1
|
Yu J, Wang X, Zhou Y, Hu J, Gu L, Zhou H, Yue C, Zhou P, Li Y, Zhao Q, Zhang C, Hu Y, Zeng F, Zhao F, Li G, Feng Y, He M, Huang S, Wu W, Huang N, Cui K, Li J. EDIL3 alleviates Mannan-induced psoriatic arthritis by slowing the intracellular glycolysis process in mononuclear-derived dendritic cells. Inflammation 2024:10.1007/s10753-024-02134-y. [PMID: 39289212 DOI: 10.1007/s10753-024-02134-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 07/12/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024]
Abstract
Psoriatic arthritis (PsA) is an immune-mediated, chronic inflammatory joint disease that commonly occurs as a complication of psoriasis. EGF-like repeats and discoidal I-like domain 3 (EDIL3) is a secreted protein with multiple structural domains and associated with various physiological functions. In this study, we employed a mannan-induced psoriatic arthritis model to investigate the impact of EDIL3 on PsA pathogenesis. Notably, a downregulation of EDIL3 expression was observed in the PsA model, which correlated with increased disease severity. EDIL3 knockout mice exhibited a more severe phenotype of PsA, which was ameliorated upon re-infusion of recombinant EDIL3 protein. The mitigation effect of EDIL3 on PsA depends on its regulation of the activation of monocyte-derived DCs (MoDCs) and T-help 17 cells (Th17). After inhibiting the function of MoDCs and Th17 cells with neutralizing antibodies, the beneficial effects of EDIL3 on PsA were lost. By inducing adenosine monophosphate (AMP)-activated protein kinase (AMPK) phosphorylation and suppressing protein kinase B (AKT) phosphorylation, EDIL3 attenuates intracellular glycolysis in MoDCs stimulated by glucose, thereby impeding their maturation and differentiation. Moreover, it diminishes the differentiation of Th17 cells and decelerates the progression of PsA. In conclusion, our findings elucidate the role and mechanism of EDIL3 in the development of PsA, providing a new target for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Jiadong Yu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoyan Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yifan Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jing Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Linna Gu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chengcheng Yue
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Pei Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ya Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qixiang Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Chen Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yawen Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fanlian Zeng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fulei Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Guolin Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuting Feng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mingxiang He
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shishi Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenling Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Nongyu Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kaijun Cui
- Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, 610041, Sichuan, China
| | - Jiong Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Roque JA, Lukesh NR, Hendy DA, Dixon TA, Islam MJ, Ontiveros-Padilla L, Pena ES, Lifshits LM, Simpson SR, Batty CJ, Bachelder EM, Ainslie KM. Enhancement of subunit vaccine delivery with zinc-carnosine coordination polymer through the addition of mannan. Int J Pharm 2024; 656:124076. [PMID: 38569976 PMCID: PMC11062752 DOI: 10.1016/j.ijpharm.2024.124076] [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: 02/08/2024] [Revised: 03/24/2024] [Accepted: 03/31/2024] [Indexed: 04/05/2024]
Abstract
Vaccines represent a pivotal health advancement for preventing infection. However, because carrier systems with repeated administration can invoke carrier-targeted immune responses that diminish subsequent immune responses (e.g., PEG antibodies), there is a continual need to develop novel vaccine platforms. Zinc carnosine microparticles (ZnCar MPs), which are composed of a one-dimensional coordination polymer formed between carnosine and the metal ion zinc, have exhibited efficacy in inducing an immune response against influenza. However, ZnCar MPs' limited suspendability hinders clinical application. In this study, we address this issue by mixing mannan, a polysaccharide derived from yeast, with ZnCar MPs. We show that the addition of mannan increases the suspendability of this promising vaccine formulation. Additionally, since mannan is an adjuvant, we illustrate that the addition of mannan increases the antibody response and T cell response when mixed with ZnCar MPs. Mice vaccinated with mannan + OVA/ZnCar MPs had elevated serum IgG and IgG1 levels in comparison to vaccination without mannan. Moreover, in the mannan + OVA/ZnCar MPs vaccinated group, mucosal washes demonstrated increased IgG, IgG1, and IgG2c titers, and antigen recall assays showed enhanced IFN-γ production in response to MHC-I and MHC-II immunodominant peptide restimulation, compared to the vaccination without mannan. These findings suggest that the use of mannan mixed with ZnCar MPs holds potential for subunit vaccination and its improved suspendability further promotes clinical translation.
Collapse
Affiliation(s)
- John A Roque
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Nicole Rose Lukesh
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Dylan A Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Timothy A Dixon
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Md Jahirul Islam
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Luis Ontiveros-Padilla
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Erik S Pena
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27599, United States
| | - Liubov M Lifshits
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Sean R Simpson
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Cole J Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27599, United States; Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| |
Collapse
|
3
|
Nenciarini S, Renzi S, di Paola M, Meriggi N, Cavalieri D. The yeast-human coevolution: Fungal transition from passengers, colonizers, and invaders. WIREs Mech Dis 2024; 16:e1639. [PMID: 38146626 DOI: 10.1002/wsbm.1639] [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: 05/19/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/27/2023]
Abstract
Fungi are the cause of more than a billion infections in humans every year, although their interactions with the host are still neglected compared to bacteria. Major systemic fungal infections are very unusual in the healthy population, due to the long history of coevolution with the human host. Humans are routinely exposed to environmental fungi and can host a commensal mycobiota, which is increasingly considered as a key player in health and disease. Here, we review the current knowledge on host-fungi coevolution and the factors that regulate their interaction. On one hand, fungi have learned to survive and inhabit the host organisms as a natural ecosystem, on the other hand, the host immune system finely tunes the response toward fungi. In turn, recognition of fungi as commensals or pathogens regulates the host immune balance in health and disease. In the human gut ecosystem, yeasts provide a fingerprint of the transient microbiota. Their status as passengers or colonizers is related to the integrity of the gut barrier and the risk of multiple disorders. Thus, the study of this less known component of the microbiota could unravel the rules of the transition from passengers to colonizers and invaders, as well as their dependence on the innate component of the host's immune response. This article is categorized under: Infectious Diseases > Environmental Factors Immune System Diseases > Environmental Factors Infectious Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
| | - Sonia Renzi
- Department of Biology, University of Florence, Florence, Italy
| | - Monica di Paola
- Department of Biology, University of Florence, Florence, Italy
| | - Niccolò Meriggi
- Department of Biology, University of Florence, Florence, Italy
| | | |
Collapse
|
4
|
Li S, Toriumi H, Takahashi D, Kamasaki T, Fujioka Y, Nagatoishi S, Li J, Liu Y, Hosokawa T, Tsumoto K, Ohba Y, Katayama Y, Murakami D, Hase K, Mori T. Safe and efficient oral allergy immunotherapy using one-pot-prepared mannan-coated allergen nanoparticles. Biomaterials 2023; 303:122381. [PMID: 37935073 DOI: 10.1016/j.biomaterials.2023.122381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/16/2023] [Accepted: 10/27/2023] [Indexed: 11/09/2023]
Abstract
Allergen immunotherapy (AIT) is the only curative treatment for allergic diseases. However, AIT has many disadvantages related to efficiency, safety, long-term duration, and patient compliance. Dendritic cells (DCs) have an important role in antigen-specific tolerance induction; thus, DC-targeting strategies to treat allergies such as glutaraldehyde crosslinked antigen to mannoprotein (MAN) have been established. However, glutaraldehyde crosslinking may reduce the antigen presentation efficiency of DCs. To overcome this, we developed a MAN-coated ovalbumin (OVA) nanoparticle (MDO), which uses intermolecular disulfide bond to crosslink OVA and MAN. MDO effectively targeted DCs resulting in tolerogenic DCs, and promoted higher antigen presentation efficiency by DCs compared with OVA or glutaraldehyde crosslinked nanoparticles. In vitro and in vivo experiments showed that DCs exposed to MDO induced Treg cells. Moreover, MDO had low reactivity with anti-OVA antibodies and did not induce anaphylaxis in allergic mice, demonstrating its high safety profile. In a mouse model of allergic asthma, MDO had significant preventative and therapeutic effects when administered orally or subcutaneously. Therefore, MDO represents a promising new approach for the efficient and safe treatment of allergies.
Collapse
Affiliation(s)
- Shunyi Li
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| | - Hiroki Toriumi
- Division of Biochemistry, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, 105-8512, Japan
| | - Daisuke Takahashi
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, 105-8512, Japan
| | - Tomoko Kamasaki
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Yoichiro Fujioka
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Satoru Nagatoishi
- The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan; Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Jinting Li
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| | - Yiwei Liu
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka, 819-0395, Japan
| | - Takanatsu Hosokawa
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| | - Kouhei Tsumoto
- The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan; Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Yusuke Ohba
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Yoshiki Katayama
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan; Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka, 819-0395, Japan; Center for Future Chemistry, Kyushu University, Fukuoka, 819-0395, Japan; International Research Center for Molecular Systems, Kyushu University, Fukuoka, 819-0395, Japan; Centre for Advanced Medicine Innovation, Kyushu University, Fukuoka, 812-8582, Japan; Department of Biomedical Engineering, Chung Yuan Christian University, Chung Li, 32023, Taiwan.
| | - Daisuke Murakami
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, 105-8512, Japan; The Institute of Fermentation Sciences (IFeS), Faculty of Food and Agricultural Sciences, Fukushima University, Kanayagawa, Fukushima, 960-1296, Japan; International Research and Development Centre for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo, 108-8639, Japan.
| | - Takeshi Mori
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan; Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka, 819-0395, Japan; Center for Future Chemistry, Kyushu University, Fukuoka, 819-0395, Japan.
| |
Collapse
|
5
|
Li S, Murakami D, Nagatoishi S, Liu Y, Tsumoto K, Katayama Y, Mori T. One-pot preparation of mannan-coated antigen nanoparticles using human serum albumin as a matrix for tolerance induction. J Colloid Interface Sci 2023; 649:955-965. [PMID: 37392685 DOI: 10.1016/j.jcis.2023.06.170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/10/2023] [Accepted: 06/25/2023] [Indexed: 07/03/2023]
Abstract
Nanoparticles (NPs) for allergen immunotherapy have garnered attention for their high efficiency and safety compared with naked antigen proteins. In this work, we present mannan-coated protein NPs, incorporating antigen proteins for antigen-specific tolerance induction. The heat-induced formation of protein NPs is a one-pot preparation method and can be applied to various proteins. Here, the NPs were formed spontaneously via heat denaturation of three component proteins: an antigen protein, human serum albumin (HSA) as a matrix protein, and mannoprotein (MAN) as a targeting ligand for dendritic cells (DCs). HSA is non-immunogenic, therefore suitable as a matrix protein, while MAN coats the surface of the NP. We applied this method to various antigen proteins and found that the self-disperse after heat denaturation was a requirement for incorporation into the NPs. We also established that the NPs could target DCs, and the incorporation of rapamycin into the NPs enhanced the induction of a tolerogenic phenotype of DC. The MAN coating provided steric hindrance and heat denaturation destroyed recognition structures, successfully preventing anti-antigen antibody binding, indicating the NPs may avoid anaphylaxis induction. The MAN-coated NPs proposed here, prepared by a simple method, have the potential for effective and safe allergies treatment for various antigens.
Collapse
Affiliation(s)
- Shunyi Li
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Daisuke Murakami
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - Satoru Nagatoishi
- The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Yiwei Liu
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka 819-0395, Japan
| | - Kouhei Tsumoto
- The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Yoshiki Katayama
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka 819-0395, Japan; Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka 819-0395, Japan; Center for Future Chemistry, Kyushu University, 819-0395, Japan; International Research Center for Molecular Systems, Kyushu University, Fukuoka 819-0395, Japan; Centre for Advanced Medicine Innovation, Kyushu University, Fukuoka 812-8582, Japan; Department of Biomedical Engineering, Chung Yuan Christian University, Chung Li, 32023, Taiwan, ROC.
| | - Takeshi Mori
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka 819-0395, Japan; Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka 819-0395, Japan; Center for Future Chemistry, Kyushu University, 819-0395, Japan.
| |
Collapse
|
6
|
Sabatini A, Guerrera G, Corsetti M, Ruocco G, De Bardi M, Renzi S, Cavalieri D, Battistini L, Angelini DF, Volpe E. Human Conventional and Plasmacytoid Dendritic Cells Differ in Their Ability to Respond to Saccharomyces cerevisiae. Front Immunol 2022; 13:850404. [PMID: 35634316 PMCID: PMC9131191 DOI: 10.3389/fimmu.2022.850404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/06/2022] [Indexed: 11/22/2022] Open
Abstract
Saccharomyces cerevisiae is a commensal yeast colonizer of mucosal surfaces and an emerging opportunistic pathogen in the mucosa and bloodstream. The role of S. cerevisiae has been largely characterized in peripheral blood mononuclear cells and monocyte-derived dendritic cells, where yeast cells induce the production of inflammatory cytokines through the interaction with mannose receptors, chitin receptors, DC SIGN, and dectin1. However, the response of blood-circulating dendritic cells (DCs) to S. cerevisiae has never been investigated. Among blood DCs, conventional DCs (cDCs) are producers of inflammatory cytokines, while plasmacytoid DCs (pDCs) are a specialized population producing a large amount of interferon (IFN)-α, which is involved in the antiviral immune response. Here we report that both human DC subsets are able to sense S. cerevisiae. In particular, cDCs produce interleukin (IL)-6, express activation markers, and promotes T helper 17 cell polarization in response to yeasts, behaving similarly to monocyte-derived DCs as previously described. Interestingly, pDCs, not cDCs, sense fungal nucleic acids, leading to the generation of P1-pDCs (PD-L1+CD80–), a pDC subset characterized by the production of IFN-α and the induction of a Th profile producing IL-10. These results highlight a novel role of pDCs in response to S. cerevisiae that could be important for the regulation of the host microbiota–immune system balance and of anti-fungal immune response.
Collapse
Affiliation(s)
- Andrea Sabatini
- Molecular Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy.,Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Gisella Guerrera
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Marta Corsetti
- Molecular Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| | - Gabriella Ruocco
- Molecular Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| | - Marco De Bardi
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Sonia Renzi
- Department of Biology, University of Florence, Florence, Italy
| | | | - Luca Battistini
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Daniela Francesca Angelini
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Elisabetta Volpe
- Molecular Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
7
|
The network interplay of interferon and Toll-like receptor signaling pathways in the anti-Candida immune response. Sci Rep 2021; 11:20281. [PMID: 34645905 PMCID: PMC8514550 DOI: 10.1038/s41598-021-99838-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/30/2021] [Indexed: 01/22/2023] Open
Abstract
Fungal infections represent a major global health problem affecting over a billion people that kills more than 1.5 million annually. In this study, we employed an integrative approach to reveal the landscape of the human immune responses to Candida spp. through meta-analysis of microarray, bulk, and single-cell RNA sequencing (scRNA-seq) data for the blood transcriptome. We identified across these different studies a consistent interconnected network interplay of signaling molecules involved in both Toll-like receptor (TLR) and interferon (IFN) signaling cascades that is activated in response to different Candida species (C. albicans, C. auris, C. glabrata, C. parapsilosis, and C. tropicalis). Among these molecules are several types I IFN, indicating an overlap with antiviral immune responses. scRNA-seq data confirmed that genes commonly identified by the three transcriptomic methods show cell type-specific expression patterns in various innate and adaptive immune cells. These findings shed new light on the anti-Candida immune response, providing putative molecular pathways for therapeutic intervention.
Collapse
|
8
|
Virulence Factors and in-Host Selection on Phenotypes in Infectious Probiotic Yeast Isolates ( Saccharomyces 'boulardii'). J Fungi (Basel) 2021; 7:jof7090746. [PMID: 34575784 PMCID: PMC8472476 DOI: 10.3390/jof7090746] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 12/27/2022] Open
Abstract
Saccharomyces yeast probiotics (S. ‘boulardii’) have long been applied in the treatment of several gastrointestinal conditions. Despite their widespread use, they are rare opportunistic pathogens responsible for a high proportion of Saccharomyces mycosis cases. The potential virulence attributes of S. ‘boulardii’ as well as its interactions with the human immune system have been studied, however, no information is available on how these yeasts may change due to in-host evolution. To fill this gap, we compared the general phenotypic characteristics, cell morphology, virulence factors, epithelial and immunological interactions, and pathogenicity of four probiotic product samples, two mycosis, and eight non-mycosis samples of S. ‘boulardii’. We assessed the characteristics related to major steps of yeast infections. Mycosis and non-mycosis isolates both displayed novel characters when compared to the product isolates, but in the case of most virulence factors and in pathogenicity, differences were negligible or, surprisingly, the yeasts from products showed elevated levels. No isolates inflicted considerable damage to the epithelial model or bore the hallmarks of immune evasion. Our results show that strains in probiotic products possess characteristics that enable them to act as pathogens upon permissive conditions, and their entry into the bloodstream is not due to active mechanisms but depends on the host. Survival in the host is dependent on yeast phenotypic characteristics which may change in many ways once they start evolving in the host. These facts call attention to the shortcomings of virulence phenotyping in yeast research, and the need for a more thorough assessment of probiotic use.
Collapse
|
9
|
Wu X, Xia Y, He F, Zhu C, Ren W. Intestinal mycobiota in health and diseases: from a disrupted equilibrium to clinical opportunities. MICROBIOME 2021; 9:60. [PMID: 33715629 PMCID: PMC7958491 DOI: 10.1186/s40168-021-01024-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/04/2021] [Indexed: 05/08/2023]
Abstract
Bacteria, viruses, protozoa, and fungi establish a complex ecosystem in the gut. Like other microbiota, gut mycobiota plays an indispensable role in modulating intestinal physiology. Notably, the most striking characteristics of intestinal fungi are their extraintestinal functions. Here, we provide a comprehensive review of the importance of gut fungi in the regulation of intestinal, pulmonary, hepatic, renal, pancreatic, and brain functions, and we present possible opportunities for the application of gut mycobiota to alleviate/treat human diseases. Video Abstract.
Collapse
Affiliation(s)
- Xiaoyan Wu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642 China
| | - Yaoyao Xia
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642 China
| | - Fang He
- College of Animal Science and Technology, Southwest University, Chongqing, 400716 China
| | - Congrui Zhu
- College of Veterinary Medicine, Kansas State University, Manhattan, KS USA
| | - Wenkai Ren
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642 China
| |
Collapse
|
10
|
Daliri EBM, Ofosu FK, Xiuqin C, Chelliah R, Oh DH. Probiotic Effector Compounds: Current Knowledge and Future Perspectives. Front Microbiol 2021; 12:655705. [PMID: 33746935 PMCID: PMC7965967 DOI: 10.3389/fmicb.2021.655705] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 02/12/2021] [Indexed: 12/22/2022] Open
Abstract
Understanding the mechanism behind probiotic action will enable a rational selection of probiotics, increase the chances of success in clinical studies and make it easy to substantiate health claims. However, most probiotic studies over the years have rather focused on the effects of probiotics in health and disease, whereas little is known about the specific molecules that trigger effects in hosts. This makes it difficult to describe the detailed mechanism by which a given probiotic functions. Probiotics communicate with their hosts through molecular signaling. Meanwhile, since the molecules produced by probiotics under in vitro conditions may differ from those produced in vivo, in vitro mechanistic studies would have to be conducted under conditions that mimic gastrointestinal conditions as much as possible. The ideal situation would, however, be to carry out well-designed clinical trials in humans (or the target animal) using adequate quantities of the suspected probiotic molecule(s) or adequate quantities of isogenic knock-out or knock-in probiotic mutants. In this review, we discuss our current knowledge about probiotic bacteria and yeast molecules that are involved in molecular signaling with the host. We also discuss the challenges and future perspectives in the search for probiotic effector molecules.
Collapse
Affiliation(s)
- Eric Banan-Mwine Daliri
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon, South Korea
| | - Fred Kwame Ofosu
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon, South Korea
| | - Chen Xiuqin
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon, South Korea
| | - Ramachandran Chelliah
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon, South Korea
| | - Deog-Hwan Oh
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
11
|
Jiang L, Stärkel P, Fan JG, Fouts DE, Bacher P, Schnabl B. The gut mycobiome: a novel player in chronic liver diseases. J Gastroenterol 2021; 56:1-11. [PMID: 33151407 PMCID: PMC7819863 DOI: 10.1007/s00535-020-01740-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023]
Abstract
The human gut microbiome (bacteria, fungi, viruses, and archaea) is a complex and diverse ecosystem. It plays an important role in human health, but is involved in several intestinal and extraintestinal diseases. Most research to date has focused on the role of bacteria, while studies focusing on fungi (also referred to as "mycobiome" or "fungome") are still in its infancy. In this review, we focus on the existing literature available about the gut mycobiome with an emphasis on compositional mycobiome changes associated with liver diseases, the impact on pathogenesis of disease, and its potential use as therapeutic targets. We also provide insights into current methodologies of studying mycobiome, and we highlight the interkingdom interactions in the context of disease and how they affect health of the host. Herein, by focusing on the gut mycobiome, this review provides novel insights and directions for liver research.
Collapse
Affiliation(s)
- Lu Jiang
- Department of Medicine, University of California San Diego, MC0063, 9500 Gilman Drive, La Jolla, CA 92093 USA ,Department of Medicine, VA San Diego Healthcare System, San Diego, CA USA
| | - Peter Stärkel
- Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Petra Bacher
- Institute of Immunology, Christian-Albrechts-University of Kiel and UKSH Schleswig-Holstein, Kiel, Germany ,Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, MC0063, 9500 Gilman Drive, La Jolla, CA 92093 USA ,Department of Medicine, VA San Diego Healthcare System, San Diego, CA USA
| |
Collapse
|
12
|
Černáková L, Rodrigues CF. Microbial interactions and immunity response in oral Candida species. Future Microbiol 2020; 15:1653-1677. [PMID: 33251818 DOI: 10.2217/fmb-2020-0113] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Oral candidiasis are among the most common noncommunicable diseases, related with serious local and systemic illnesses. Although these infections can occur in all kinds of patients, they are more recurrent in immunosuppressed ones such as patients with HIV, hepatitis, cancer or under long antimicrobial treatments. Candida albicans continues to be the most frequently identified Candida spp. in these disorders, but other non-C. albicans Candida are rising. Understanding the immune responses involved in oral Candida spp. infections is a key feature to a successful treatment and to the design of novel therapies. In this review, we performed a literature search in PubMed and WoS, in order to examine and analyze common oral Candida spp.-bacteria/Candida-Candida interactions and the host immunity response in oral candidiasis.
Collapse
Affiliation(s)
- Lucia Černáková
- Department of Microbiology & Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovičova 6, 842 15 Bratislava, Slovakia
| | - Célia F Rodrigues
- Department of Chemical Engineering, LEPABE - Laboratory for Process Engineering, Environment, Biotechnology & Energy, Faculty of Engineering, University of Porto, Portugal
| |
Collapse
|
13
|
Galloway-Peña JR, Kontoyiannis DP. The gut mycobiome: The overlooked constituent of clinical outcomes and treatment complications in patients with cancer and other immunosuppressive conditions. PLoS Pathog 2020; 16:e1008353. [PMID: 32240277 PMCID: PMC7117661 DOI: 10.1371/journal.ppat.1008353] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Jessica R. Galloway-Peña
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail: (JRG-P); (DPK)
| | - Dimitrios P. Kontoyiannis
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail: (JRG-P); (DPK)
| |
Collapse
|
14
|
Tang Y, Zhang H, Xu H, Zeng W, Qiu Y, Tan C, Tang S, Zhang J. Dendritic Cells Promote Treg Expansion but Not Th17 Generation in Response to Talaromyces marneffei Yeast Cells. Infect Drug Resist 2020; 13:805-813. [PMID: 32210595 PMCID: PMC7075240 DOI: 10.2147/idr.s239906] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/25/2020] [Indexed: 12/29/2022] Open
Abstract
Background Dendritic cells (DCs) with both proinflammatory and tolerogenic properties have been implicated in modulation of CD4+ T cell responses in many fungal diseases. However, the role of DC in the context of Talaromyces marneffei (T. marneffei) infection has not been determined. In this study, we aimed to study the effect of the yeast form of T. marneffei yeasts on DCs, as well as the role of DCs in modulating T helper 17 (Th17) and regulatory T (Treg) cell responses to the pathogen. Methods Mouse bone marrow-derived DCs were stimulated with T. marneffei yeasts for 24 h. Frequencies of CD80 and CD86 expression on DCs and the levels of IL-6, IL-10 and TGF-β in the culture supernatant of yeast-stimulated DCs were detected by flow cytometry and ELISA, respectively. In co-culture experiments, CD4+ T lymphocytes of mice were isolated from the spleen using magnetic beads and co-cultured with T. marneffei yeasts, with or without DCs for 24 h. The proportions of Th17 and Treg cells in co-culture were detected by flow cytometry. The mRNA levels of RORγt and Foxp3 were detected by RT-PCR. Levels of IL-10 and TGF-β in the co-culture supernatant were detected by ELISA. Results The expressions of CD80 and CD86 on DCs were increased, as well as IL-6, IL-10 and TGF-β levels in the culture supernatant of T. marneffei-stimulated DCs were higher than those in DCs cultured without T. marneffei. In co-culture experiments, in the presence of DCs, T. marneffei promoted Treg expansion and Foxp3 up-regulation but limited Th17 and downregulated RORγt. Levels of IL-10 and TGF-β were higher in the co-culture containing DCs than without DCs. Conclusion Our findings demonstrated that the interaction between DCs and T. marneffei could promote Treg expansion but not Th17 generation. These findings provide a mechanism by which DCs may promote immune tolerance in T. marneffei infection.
Collapse
Affiliation(s)
- Yanping Tang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Hui Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Haiguang Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Wen Zeng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Ye Qiu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Caimei Tan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Shudan Tang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Jianquan Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| |
Collapse
|
15
|
Paulovičová E, Paulovičová L, Farkaš P, Karelin AA, Tsvetkov YE, Krylov VB, Nifantiev NE. Importance of Candida Antigenic Factors: Structure-Driven Immunomodulation Properties of Synthetically Prepared Mannooligosaccharides in RAW264.7 Macrophages. Front Cell Infect Microbiol 2019; 9:378. [PMID: 31788453 PMCID: PMC6856089 DOI: 10.3389/fcimb.2019.00378] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 10/21/2019] [Indexed: 12/15/2022] Open
Abstract
The incidence and prevalence of serious fungal infections is rising, especially in immunosuppressed individuals. Moreover, co-administration of antibiotics and immunosuppressants has driven the emergence of new multidrug-resistant pathogens. The significant increase of multidrug-resistant pathogens, together with their ability to form biofilms, is associated with morbidity and mortality. Research on novel synthetically prepared immunomodulators as potential antifungal immunotherapeutics is of serious interest. Our study demonstrated the immunobiological activity of synthetically prepared biotinylated mannooligosaccharides mimicking Candida antigenic factors using RAW264.7 macrophages. Macrophage exposure to the set of eight structurally different mannooligosaccharides induced a release of Th1, Th2, Th17, and Treg cytokine signature patterns. The observed immune responses were tightly associated with structure, dose, exposure time, and selected signature cytokines. The viability/cytotoxicity of the mannooligosaccharide formulas was assessed based on cell proliferation. The structure-based immunomodulatory activity of the formulas was evaluated with respect to the length, branching and conformation of the various formulas. Glycoconjugate formulas with terminal β-mannosyl-units tended to be more potent in terms of Candida relevant cytokines IL-12 p70, IL-17, GM-CSF, IL-6, and TNFα induction and cell proliferation, and this tendency was associated with structural differences between the studied glycoconjugate formulas. The eight tested mannooligosaccharide conjugates can be considered potential in vitro immunomodulative agents suitable for in vitro Candida diagnostics or prospectively for subcellular anti-Candida vaccine design.
Collapse
Affiliation(s)
- Ema Paulovičová
- Cell Culture & Immunology Laboratory, Department of Immunochemistry of Glycoconjugates, Center for Glycomics, Institute of Chemistry, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lucia Paulovičová
- Cell Culture & Immunology Laboratory, Department of Immunochemistry of Glycoconjugates, Center for Glycomics, Institute of Chemistry, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Pavol Farkaš
- Cell Culture & Immunology Laboratory, Department of Immunochemistry of Glycoconjugates, Center for Glycomics, Institute of Chemistry, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Alexander A Karelin
- Laboratory of Glycoconjugate Chemistry, N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Yury E Tsvetkov
- Laboratory of Glycoconjugate Chemistry, N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Vadim B Krylov
- Laboratory of Glycoconjugate Chemistry, N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Nikolay E Nifantiev
- Laboratory of Glycoconjugate Chemistry, N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
16
|
Novel vaccines targeting dendritic cells by coupling allergoids to mannan. ALLERGO JOURNAL 2018. [DOI: 10.1007/s15007-018-1764-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
17
|
Ramazzotti M, Stefanini I, Di Paola M, De Filippo C, Rizzetto L, Berná L, Dapporto L, Rivero D, Tocci N, Weil T, Lenucci MS, Lionetti P, Cavalieri D. Population genomics reveals evolution and variation of Saccharomyces cerevisiae in the human and insects gut. Environ Microbiol 2018; 21:50-71. [PMID: 30246283 DOI: 10.1111/1462-2920.14422] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 09/07/2018] [Accepted: 09/17/2018] [Indexed: 12/16/2022]
Abstract
The quest to discover the variety of ecological niches inhabited by Saccharomyces cerevisiae has led to research in areas as diverse as wineries, oak trees and insect guts. The discovery of fungal communities in the human gastrointestinal tract suggested the host's gut as a potential reservoir for yeast adaptation. Here, we report the existence of yeast populations associated with the human gut (HG) that differ from those isolated from other human body sites. Phylogenetic analysis on 12 microsatellite loci and 1715 combined CDSs from whole-genome sequencing revealed three subclusters of HG strains with further evidence of clonal colonization within the host's gut. The presence of such subclusters was supported by other genomic features, such as copy number variation, absence/introgressions of CDSs and relative polymorphism frequency. Functional analysis of CDSs specific of the different subclusters suggested possible alterations in cell wall composition and sporulation features. The phenotypic analysis combined with immunological profiling of these strains further showed that sporulation was related with strain-specific genomic characteristics in the immune recognition pattern. We conclude that both genetic and environmental factors involved in cell wall remodelling and sporulation are the main drivers of adaptation in S. cerevisiae populations in the human gut.
Collapse
Affiliation(s)
- Matteo Ramazzotti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Irene Stefanini
- Division of Biomedical Sciences, University of Warwick, Coventry, UK
| | - Monica Di Paola
- Department of Biology, University of Florence, Florence, Italy
| | - Carlotta De Filippo
- Institute of Agricultural Biology and Biotechnology, National Research Council (CNR), Pisa, Italy
| | - Lisa Rizzetto
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, San Michele All'Adige (Trento), Italy
| | - Luisa Berná
- Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | | | - Damariz Rivero
- Department of Biology, University of Florence, Florence, Italy
| | - Noemi Tocci
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, San Michele All'Adige (Trento), Italy
| | - Tobias Weil
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, San Michele All'Adige (Trento), Italy
| | - Marcello S Lenucci
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali (Di.S.Te.B.A.), Università del Salento, Lecce, Italy
| | - Paolo Lionetti
- Department of Neuroscience, Psychology, Drug Research and Child Health, Meyer Children Hospital, University of Florence, Florence, Italy
| | | |
Collapse
|
18
|
Lai GC, Tan TG, Pavelka N. The mammalian mycobiome: A complex system in a dynamic relationship with the host. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2018; 11:e1438. [PMID: 30255552 PMCID: PMC6586165 DOI: 10.1002/wsbm.1438] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 08/26/2018] [Accepted: 08/28/2018] [Indexed: 12/21/2022]
Abstract
Mammalian barrier surfaces are densely populated by symbiont fungi in much the same way the former are colonized by symbiont bacteria. The fungal microbiota, otherwise known as the mycobiota, is increasingly recognized as a critical player in the maintenance of health and homeostasis of the host. Here we discuss the impact of the mycobiota on host physiology and disease, the factors influencing mycobiota composition, and the current technologies used for identifying symbiont fungal species. Understanding the tripartite interactions among the host, mycobiota, and other members of the microbiota, will help to guide the development of novel prevention and therapeutic strategies for a variety of human diseases. This article is categorized under:
Physiology > Mammalian Physiology in Health and Disease Laboratory Methods and Technologies > Genetic/Genomic Methods Models of Systems Properties and Processes > Organismal Models
Collapse
|
19
|
Benito-Villalvilla C, Soria I, Subiza JL, Palomares O. Novel vaccines targeting dendritic cells by coupling allergoids to mannan. ACTA ACUST UNITED AC 2018; 27:256-262. [PMID: 30546997 PMCID: PMC6267119 DOI: 10.1007/s40629-018-0069-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 04/09/2018] [Indexed: 01/27/2023]
Abstract
Allergen-specific immunotherapy (AIT) is the single disease-modifying treatment for allergy. Clinical trials show AIT to be safe and effective for many patients; however, it still faces problems related to efficacy, safety, long treatment duration and low patient adherence. There has been intensive research to develop alternative strategies, including novel administration routes, adjuvants or hypoallergenic molecules. Promising results are reported for some of them, but clinical progress is still moderate. Allergoids conjugated to nonoxidized mannan from Saccharomyces cerevisiae have emerged as a novel concept of vaccine targeting dendritic cells (DCs). Preclinical human and animal models demonstrated that allergoids conjugated to mannan enhance allergen uptake, promote healthy responses to allergens by inducing Th1 and T regulatory (Treg) cells, and show clinical efficacy in veterinary medicine. Dose-finding phase II clinical trials in humans are currently ongoing. We review the current stage of allergoids conjugated to mannan as next generation vaccines for AIT.
Collapse
Affiliation(s)
- Cristina Benito-Villalvilla
- 1Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, 28040 Madrid, Spain
| | | | | | - Oscar Palomares
- 1Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
20
|
Deciphering the mechanism of action of 089, a compound impairing the fungal cell cycle. Sci Rep 2018; 8:5964. [PMID: 29654251 PMCID: PMC5899093 DOI: 10.1038/s41598-018-24341-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 03/29/2018] [Indexed: 01/28/2023] Open
Abstract
Fungal infections represent an increasingly relevant clinical problem, primarily because of the increased survival of severely immune-compromised patients. Despite the availability of active and selective drugs and of well-established prophylaxis, classical antifungals are often ineffective as resistance is frequently observed. The quest for anti-fungal drugs with novel mechanisms of action is thus important. Here we show that a new compound, 089, acts by arresting fungal cells in the G2 phase of the cell cycle through targeting of SWE1, a mechanism of action unexploited by current anti-fungal drugs. The cell cycle impairment also induces a modification of fungal cell morphology which makes fungal cells recognizable by immune cells. This new class of molecules holds promise to be a valuable source of novel antifungals, allowing the clearance of pathogenic fungi by both direct killing of the fungus and enhancing the recognition of the pathogen by the host immune system.
Collapse
|
21
|
Xiao W, He H, Tong Y, Cai M, Shi Y, Liu B, Wang J, Qin Y, Lai S. Transcriptome analysis of Trichophyton mentagrophytes–induced rabbit (Oryctolagus cuniculus) dermatophytosis. Microb Pathog 2018; 114:350-356. [DOI: 10.1016/j.micpath.2017.12.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/01/2017] [Accepted: 12/06/2017] [Indexed: 12/28/2022]
|
22
|
Nguyen TNY, Matangkasombut O, Ritprajak P. Differential dendritic cell responses to cell wall mannan of Candida albicans, Candida parapsilosis, and Candida dubliniensis . J Oral Sci 2018; 60:557-566. [DOI: 10.2334/josnusd.17-0426] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Thu N. Y. Nguyen
- Graduate program in Oral Biology, Faculty of Dentistry, Chulalongkorn University
- Research Unit on Oral Microbiology and Immunology and Department of Microbiology, Faculty of Dentistry, Chulalongkorn University
| | - Oranart Matangkasombut
- Research Unit on Oral Microbiology and Immunology and Department of Microbiology, Faculty of Dentistry, Chulalongkorn University
- Laboratory of Biotechnology, Chulabhorn Research Institute
| | - Patcharee Ritprajak
- Research Unit on Oral Microbiology and Immunology and Department of Microbiology, Faculty of Dentistry, Chulalongkorn University
- Oral Biology Research Center, Faculty of Dentistry, Chulalongkorn University
| |
Collapse
|
23
|
Ness T, Abdallah M, Adams J, Alvarado C, Gunn E, House B, Lamb J, Macguire J, Norris E, Robinson R, Sapp M, Sharma J, Garner R. Candida albicans-derived mannoproteins activate NF-κB in reporter cells expressing TLR4, MD2 and CD14. PLoS One 2017; 12:e0189939. [PMID: 29281684 PMCID: PMC5744952 DOI: 10.1371/journal.pone.0189939] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 12/05/2017] [Indexed: 01/10/2023] Open
Abstract
The ability of soluble C. albicans 20A (serotype A) mannoprotein (CMP) to serve as a ligand for toll-like receptor 4 (TLR4) and its co-receptors was examined using commercially available and stably-transfected HEK293 cells that express human TLR4, MD2 and CD14, but not MR. These TLR4 reporter cells also express an NF-κB-dependent, secreted embryonic alkaline phosphatase (SEAP) reporter gene. TLR4-reporter cells exhibited a dose-dependent SEAP response to both LPS and CMP, wherein peak activation was achieved after stimulation with 40–50 μg/mL of CMP. Incubation on polymyxin B resin had no effect on CMP’s ligand activity, but neutralized LPS-spiked controls. HEK293 Null cells lacking TLR4 and possessing the same SEAP reporter failed to respond to LPS or CMP, but produced SEAP when activated with TNFα. Reporter cell NF-κB responses were accompanied by transcription of IL-8, TNFα, and COX-2 genes. Celecoxib inhibited LPS-, CMP-, and TNFα-dependent NF-κB responses; whereas, indomethacin had limited effect on LPS and CMP responses. SEAP production in response to C. albicans A9 mnn4Δ mutant CMP, lacking phosphomannosylations on N-linked glycans, was significantly greater (p ≤ 0.005) than SEAP responses to CMP derived from parental A9 (both serotype B). These data confirm that engineered human cells expressing TLR4, MD2 and CD14 can respond to CMP with NF-κB activation and the response can be influenced by variations in CMP-mannosylation. Future characterizations of CMPs from other sources and their application in this model may provide further insight into variations observed with TLR4 dependent innate immune responses targeting different C. albicans strains.
Collapse
Affiliation(s)
- Traci Ness
- Department of Biology, Armstrong State University, Savannah, Georgia, United States of America
| | - Mahmud Abdallah
- Department of Biology, Armstrong State University, Savannah, Georgia, United States of America
| | - Jaime Adams
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia, United States of America
| | - Claudia Alvarado
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia, United States of America
| | - Edwin Gunn
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia, United States of America
| | - Brittany House
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia, United States of America
| | - John Lamb
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia, United States of America
| | - Jack Macguire
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia, United States of America
| | - Emily Norris
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia, United States of America
| | - Rebekah Robinson
- Department of Biology, Armstrong State University, Savannah, Georgia, United States of America
| | - Morgan Sapp
- Department of Biology, Armstrong State University, Savannah, Georgia, United States of America
| | - Jill Sharma
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia, United States of America
| | - Ronald Garner
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia, United States of America
- * E-mail:
| |
Collapse
|
24
|
Maturation and cytokine pattern of human dendritic cells in response to different yeasts. Med Microbiol Immunol 2017; 207:75-81. [DOI: 10.1007/s00430-017-0528-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 11/14/2017] [Indexed: 12/20/2022]
|
25
|
Beneficial Effects of Prebiotic Saccharomyces cerevisiae Mannan on Allergic Asthma Mouse Models. J Immunol Res 2017; 2017:3432701. [PMID: 28835901 PMCID: PMC5556605 DOI: 10.1155/2017/3432701] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/31/2017] [Accepted: 06/04/2017] [Indexed: 12/21/2022] Open
Abstract
One of the unmet needs for asthma management is a new therapeutic agent with both anti-inflammatory and anti-smooth muscle (ASM) remodeling effects. The mannose receptor (MR) family plays an important role in allergen uptake and processing of major allergens Der p 1 and Fel d 1. We have previously reported that ASM cells express a mannose receptor (ASM-MR) and that mannan derived from Saccharomyces cerevisiae (SC-MN) inhibits mannosyl-rich lysosomal hydrolase-induced bovine ASM cell proliferation. Using a humanized transgenic mouse strain (huASM-MRC2) expressing the human MRC2 receptor in a SM tissue-specific manner, we have demonstrated that ASM hyperplasia/hypertrophy can occur as early as 15 days after allergen challenge in this mouse model and this phenomenon is preventable with SC-MN treatment. This proof-of-concept study would facilitate future development of a potential asthma therapeutic agent with dual function of anti-inflammatory and anti-smooth muscle remodeling effects.
Collapse
|
26
|
Airway Epithelial Repair by a Prebiotic Mannan Derived from Saccharomyces cerevisiae. J Immunol Res 2017; 2017:8903982. [PMID: 28770233 PMCID: PMC5523272 DOI: 10.1155/2017/8903982] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/05/2017] [Accepted: 05/30/2017] [Indexed: 01/01/2023] Open
Abstract
In asthmatic airways, repeated epithelial damage and repair occur. No current therapy directly targets this process. We aimed to determine the effects of mannan derived from S. cerevisiae (SC-MN) on airway epithelial wound repair, in vitro. The presence of functional mannose receptors in bronchial epithelial cells was shown by endocytosis of colloidal gold-Man BSA via clathrin-coated pits in 16HBE cells. In primary normal human bronchial epithelial cells (NHBEC), SC-MN significantly facilitated wound closure. Treatment with SC-MN stimulated cell spreading as indicated by a significant increase in the average lamellipodial width of wound edge 16HBE cells. In addition, NHBEC treated with SC-MN showed increased expression and activation of Krüppel-like factors (KLFs) 4 and 5, transcription factors important in epithelial cell survival and regulation of epithelial-mesenchymal transition. We conclude that SC-MN facilitates wound repair in human bronchial epithelium, involving mannose receptors.
Collapse
|
27
|
Niu XX, Li T, Zhang X, Wang SX, Liu ZH. Lactobacillus crispatus Modulates Vaginal Epithelial Cell Innate Response to Candida albicans. Chin Med J (Engl) 2017; 130:273-279. [PMID: 28139509 PMCID: PMC5308008 DOI: 10.4103/0366-6999.198927] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Vulvovaginal candidiasis is caused by Candida albicans. The vaginal epithelium, as the first site of the initial stage of infection by pathogens, plays an important role in resisting genital tract infections. Moreover, lactobacilli are predominant members of the vaginal microbiota that help to maintain a normal vaginal microenvironment. Therefore, Lactobacillus crispatus was explored for its capacity to intervene in the immune response of vaginal epithelial cells VK2/E6E7 to C. albicans. METHODS We examined the interleukin-2 (IL-2), 4, 6, 8, and 17 produced by VK2/E6E7 cells infected with C. albicans and treated with L. crispatus in vitro. The capacity of L. crispatus to adhere to VK2/E6E7 and inhibit C. albicans growth was also tested by scanning electron microscopy (SEM) and adhesion experiments. RESULTS Compared with group VK2/E6E7 with C. albicans, when treated with L. crispatus, the adhesion of C. albicans to VK2/E6E7 cells decreased significantly by 52.87 ± 1.22%, 47.03 ± 1.35%, and 42.20 ± 1.55% under competition, exclusion, and displacement conditions, respectively. SEM revealed that the invasion of C. albicans into VK2/E6E7 cells was caused by induced endocytosis and active penetration. L. crispatus could effectively protect the cells from the virulence of hyphae and spores of C. albicans and enhance the local immune function of the VK2/E6E7 cells. The concentrations of IL-2, 6, and 17 were upregulated significantly (P < 0.01) and that of IL-8 were downregulated significantly (P < 0.01) in infected VK2/E6E7 cells treated with L. crispatus. The concentration of IL-4 was similar to that of the group VK2/E6E7 with C. albicans (24.10 ± 0.97 vs. 23.12 ± 0.76 pg/ml, P = 0.221). CONCLUSIONS L. crispatus can attenuate the virulence of C. albicans, modulate the secretion of cytokines and chemokines, and enhance the immune response of VK2/E6E7 cells in vitro. The vaginal mucosa has a potential function in the local immune responses against pathogens that can be promoted by L. crispatus.
Collapse
Affiliation(s)
- Xiao-Xi Niu
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing 100034, China
| | - Ting Li
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing 100034, China
| | - Xu Zhang
- Laboratory of Electron Microscopy, Ultrastructural Pathology Center, Peking University First Hospital, Beijing 100034, China
| | - Su-Xia Wang
- Laboratory of Electron Microscopy, Ultrastructural Pathology Center, Peking University First Hospital, Beijing 100034, China
| | - Zhao-Hui Liu
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
28
|
Li KB, Li N, Zang Y, Chen GR, Li J, James TD, He XP, Tian H. Foldable glycoprobes capable of fluorogenic crosslinking of biomacromolecules. Chem Sci 2016; 7:6325-6329. [PMID: 28567244 PMCID: PMC5450440 DOI: 10.1039/c6sc02366e] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 07/11/2016] [Indexed: 01/10/2023] Open
Abstract
We demonstrate a foldable, fluorogenic glycoprobe that can recognize and simultaneously crosslink a receptor protein to form unique supramolecular bio-ensembles.
Small-molecular probes capable of monitoring and interfering with the activity of biomacromolecules – such as polysaccharides, nucleotides and proteins – are of paramount importance to the advancement of life science. However, such probes that can detect and simultaneously modulate the construction of biomacromolecules are elusive. Here we report a fluorogenic, foldable glycoprobe that can recognize and assemble a protein receptor in a synchronous fashion. The glycoprobe synthesized by introducing a glycoligand (for protein recognition) to a bola-type bis-fluorophore conjugate shows a “self-shielded” fluorescence in the folded state. Association with a receptor protein rapidly unfolds the probe, releasing a fluorophore capable of crosslinking the proteins – as determined using small-angle X-ray scattering – thereby producing a unique fluorescent supramolecular construct. We have demonstrated the use of the foldable glycoprobe in order to track the endocytic cycle of a transmembrane receptor.
Collapse
Affiliation(s)
- Kai-Bin Li
- Key Laboratory for Advanced Materials & Institute of Fine Chemicals , School of Chemistry and Molecular Engineering , East China University of Science and Technology , 130 Meilong Rd. , Shanghai 200237 , PR China .
| | - Na Li
- National Center for Protein Science Shanghai , Shanghai Institutes of Biological Sciences , Chinese Academy of Sciences , Shanghai 200031 , PR China
| | - Yi Zang
- National Center for Drug Screening , State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica , Chinese Academy of Sciences , 189 Guo Shoujing Rd. , Shanghai 201203 , PR China .
| | - Guo-Rong Chen
- Key Laboratory for Advanced Materials & Institute of Fine Chemicals , School of Chemistry and Molecular Engineering , East China University of Science and Technology , 130 Meilong Rd. , Shanghai 200237 , PR China .
| | - Jia Li
- National Center for Drug Screening , State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica , Chinese Academy of Sciences , 189 Guo Shoujing Rd. , Shanghai 201203 , PR China .
| | - Tony D James
- Department of Chemistry , University of Bath , Bath , BA2 7AY , UK
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials & Institute of Fine Chemicals , School of Chemistry and Molecular Engineering , East China University of Science and Technology , 130 Meilong Rd. , Shanghai 200237 , PR China .
| | - He Tian
- Key Laboratory for Advanced Materials & Institute of Fine Chemicals , School of Chemistry and Molecular Engineering , East China University of Science and Technology , 130 Meilong Rd. , Shanghai 200237 , PR China .
| |
Collapse
|
29
|
Abstract
An increasing body of literature is addressing the immuno-modulating functions of miRNAs which include paracrine signaling via exosome-mediated intercellular miRNA. In view of the recent evidence of intake and bioavailability of dietary miRNAs in humans and animals we explored the immuno-modulating capacity of plant derived miRNAs. Here we show that transfection of synthetic miRNAs or native miRNA-enriched fractions obtained from a wide range of plant species and organs modifies dendritic cells ability to respond to inflammatory agents by limiting T cell proliferation and consequently dampening inflammation. This immuno-modulatory effect appears associated with binding of plant miRNA on TLR3 with ensuing impairment of TRIF signaling. Similarly, in vivo, plant small RNAs reduce the onset of severity of Experimental Autoimmune Encephalomyelities by limiting dendritic cell migration and dampening Th1 and Th17 responses in a Treg-independent manner. Our results indicate a potential for therapeutic use of plant miRNAs in the prevention of chronic-inflammation related diseases.
Collapse
|
30
|
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
|
31
|
Pai J, Hyun JY, Jeong J, Loh S, Cho EH, Kang YS, Shin I. Carbohydrate microarrays for screening functional glycans. Chem Sci 2016; 7:2084-2093. [PMID: 29899934 PMCID: PMC5968531 DOI: 10.1039/c5sc03789a] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/10/2015] [Indexed: 12/27/2022] Open
Abstract
Carbohydrate microarrays were used for the simultaneous screening of various glycans whose binding to the cell-surface lectin elicits cellular response.
Carbohydrate microarrays have become robust and powerful tools for the rapid analysis of glycan-associated binding events. However, this microarray technology has rarely been applied in studies of glycan-mediated cellular responses. Herein we describe a carbohydrate microarray-based approach for the rapid screening of biologically active glycans that stimulate the production of reactive oxygen species (ROS) through binding to the cell-surface lectin. We employed a microarray assay and a fluorescent ROS probe to identify the functional glycans which enhance ROS production. Cells binding to glycans on the microarrays produced ROS, whose levels were decreased in the presence of a ROS scavenger or a NADPH oxidase inhibitor. The present study leads us to suggest that glycan microarrays are applicable to the simultaneous screening of various glycans whose binding to the cell-surface lectin elicits cellular response.
Collapse
Affiliation(s)
- Jaeyoung Pai
- Center for Biofunctional Molecules , Department of Chemistry , Yonsei University , Seoul 03722 , Korea .
| | - Ji Young Hyun
- Center for Biofunctional Molecules , Department of Chemistry , Yonsei University , Seoul 03722 , Korea .
| | - Jieun Jeong
- Center for Biofunctional Molecules , Department of Chemistry , Yonsei University , Seoul 03722 , Korea .
| | - Sohee Loh
- Department of Biomedical Science and Technology , Department of Veterinary Pharmacology and Toxicology , College of Veterinary Medicine , Konkuk University , Seoul 05029 , Korea
| | - Eun-Hee Cho
- Department of Biomedical Science and Technology , Department of Veterinary Pharmacology and Toxicology , College of Veterinary Medicine , Konkuk University , Seoul 05029 , Korea
| | - Young-Sun Kang
- Department of Biomedical Science and Technology , Department of Veterinary Pharmacology and Toxicology , College of Veterinary Medicine , Konkuk University , Seoul 05029 , Korea
| | - Injae Shin
- Center for Biofunctional Molecules , Department of Chemistry , Yonsei University , Seoul 03722 , Korea .
| |
Collapse
|
32
|
Field KA, Johnson JS, Lilley TM, Reeder SM, Rogers EJ, Behr MJ, Reeder DM. The White-Nose Syndrome Transcriptome: Activation of Anti-fungal Host Responses in Wing Tissue of Hibernating Little Brown Myotis. PLoS Pathog 2015; 11:e1005168. [PMID: 26426272 PMCID: PMC4591128 DOI: 10.1371/journal.ppat.1005168] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 08/25/2015] [Indexed: 01/08/2023] Open
Abstract
White-nose syndrome (WNS) in North American bats is caused by an invasive cutaneous infection by the psychrophilic fungus Pseudogymnoascus destructans (Pd). We compared transcriptome-wide changes in gene expression using RNA-Seq on wing skin tissue from hibernating little brown myotis (Myotis lucifugus) with WNS to bats without Pd exposure. We found that WNS caused significant changes in gene expression in hibernating bats including pathways involved in inflammation, wound healing, and metabolism. Local acute inflammatory responses were initiated by fungal invasion. Gene expression was increased for inflammatory cytokines, including interleukins (IL) IL-1β, IL-6, IL-17C, IL-20, IL-23A, IL-24, and G-CSF and chemokines, such as Ccl2 and Ccl20. This pattern of gene expression changes demonstrates that WNS is accompanied by an innate anti-fungal host response similar to that caused by cutaneous Candida albicans infections. However, despite the apparent production of appropriate chemokines, immune cells such as neutrophils and T cells do not appear to be recruited. We observed upregulation of acute inflammatory genes, including prostaglandin G/H synthase 2 (cyclooxygenase-2), that generate eicosanoids and other nociception mediators. We also observed differences in Pd gene expression that suggest host-pathogen interactions that might determine WNS progression. We identified several classes of potential virulence factors that are expressed in Pd during WNS, including secreted proteases that may mediate tissue invasion. These results demonstrate that hibernation does not prevent a local inflammatory response to Pd infection but that recruitment of leukocytes to the site of infection does not occur. The putative virulence factors may provide novel targets for treatment or prevention of WNS. These observations support a dual role for inflammation during WNS; inflammatory responses provide protection but excessive inflammation may contribute to mortality, either by affecting torpor behavior or causing damage upon emergence in the spring. White-nose syndrome is the most devastating epizootic wildlife disease of mammals in history, having killed millions of hibernating bats in North America since 2007. We have used next-generation RNA sequencing to provide a survey of the gene expression changes that accompany this disease in the skin of bats infected with the causative fungus. We identified possible new mechanisms that may either provide protection or contribute to mortality, including inflammatory immune responses. Contrary to expectations that hibernation represents a period of dormancy, we found that gene expression pathways were responsive to the environment. We also examined which genes were expressed in the pathogen and identified several classes of genes that could contribute to the virulence of this disease. Gene expression changes in the host were associated with local inflammation despite the fact that the bats were hibernating. However, we found that hibernating bats with white-nose syndrome lack some of the responses known to defend other mammals from fungal infection. We propose that bats could be protected from white-nose syndrome if these responses could be established prior to hibernation or if treatments could block the virulence factors expressed by the pathogen.
Collapse
Affiliation(s)
- Kenneth A. Field
- Department of Biology, Bucknell University, Lewisburg, Pennsylvania, United States of America
- * E-mail:
| | - Joseph S. Johnson
- Department of Biology, Bucknell University, Lewisburg, Pennsylvania, United States of America
| | - Thomas M. Lilley
- Department of Biology, Bucknell University, Lewisburg, Pennsylvania, United States of America
| | - Sophia M. Reeder
- Department of Biology, Bucknell University, Lewisburg, Pennsylvania, United States of America
| | - Elizabeth J. Rogers
- Department of Biology, Bucknell University, Lewisburg, Pennsylvania, United States of America
| | - Melissa J. Behr
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - DeeAnn M. Reeder
- Department of Biology, Bucknell University, Lewisburg, Pennsylvania, United States of America
| |
Collapse
|
33
|
Clemente AM, Rizzetto L, Castronovo G, Perissi E, Tanturli M, Cozzolino F, Cavalieri D, Fusi F, Cialdai F, Vignali L, Torcia MG, Monici M. Effects of near-infrared laser radiation on the survival and inflammatory potential of Candida spp. involved in the pathogenesis of chemotherapy-induced oral mucositis. Eur J Clin Microbiol Infect Dis 2015; 34:1999-2007. [PMID: 26173694 DOI: 10.1007/s10096-015-2443-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/26/2015] [Indexed: 02/06/2023]
Abstract
Candida spp. usually colonize ulcerative lesions of atrophic mucosa in patients with chemotherapy-induced oral mucositis inducing severe inflammation. The spread of antifungal-resistant strains strongly encouraged the search of complementary or alternative therapeutic strategies to cure inflamed mucosa. In this paper, we studied the effects of a near-infrared (NIR) laser system with dual-wavelength emission (808 nm + 904 nm) on the survival and inflammatory potential of C. albicans, C. glabrata, and C. parapsilosis. Laser treatment was performed with a Multiwave Locked System laser. Survival and apoptosis of fungal strains were evaluated by colony-forming units (CFU) counting and annexin V staining. Cytokine production was evaluated by ImmunoPlex array. Laser treatment significantly affected the survival of Candida spp. by inducing apoptosis and induced a lower production of inflammatory cytokines by dendritic cells compared to untreated fungi. No differences in the survival and inflammatory potential were recorded in treated or untreated Saccharomyces cerevisiae cells, used as the control non-pathogenic microorganism. Laser treatment altered the survival and inflammatory potential of pathogenic Candida spp. These data provide experimental support to the use of NIR laser radiation as a co-adjuvant of antifungal therapy in patients with oral mucositis (OM) complicated by Candida infections.
Collapse
Affiliation(s)
- A M Clemente
- Department of Clinical and Experimental Medicine, University of Firenze, Firenze, FI, Italy
| | - L Rizzetto
- Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, TN, Italy
| | - G Castronovo
- Department of Experimental and Clinical Biomedical Sciences, University of Firenze, Firenze, FI, Italy
| | - E Perissi
- Department of Experimental and Clinical Biomedical Sciences, University of Firenze, Firenze, FI, Italy
| | - M Tanturli
- Department of Experimental and Clinical Biomedical Sciences, University of Firenze, Firenze, FI, Italy
| | - F Cozzolino
- Department of Experimental and Clinical Biomedical Sciences, University of Firenze, Firenze, FI, Italy
| | - D Cavalieri
- Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, TN, Italy
| | - F Fusi
- Medical Physics Unit, Department of Experimental and Clinical Biomedical Sciences, University of Firenze, Firenze, FI, Italy
| | - F Cialdai
- ASAcampus Joint Laboratory, ASA Research Division, Department of Experimental and Clinical Biomedical Sciences, University of Firenze, Firenze, FI, Italy
| | - L Vignali
- ASAcampus Joint Laboratory, ASA Research Division, Department of Experimental and Clinical Biomedical Sciences, University of Firenze, Firenze, FI, Italy
| | - M G Torcia
- Department of Clinical and Experimental Medicine, University of Firenze, Firenze, FI, Italy.
| | - M Monici
- ASAcampus Joint Laboratory, ASA Research Division, Department of Experimental and Clinical Biomedical Sciences, University of Firenze, Firenze, FI, Italy
| |
Collapse
|
34
|
Park S, Kim GH, Park SH, Pai J, Rathwell D, Park JY, Kang YS, Shin I. Probing cell-surface carbohydrate binding proteins with dual-modal glycan-conjugated nanoparticles. J Am Chem Soc 2015; 137:5961-8. [PMID: 25939670 DOI: 10.1021/jacs.5b00592] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Dual-modal fluorescent magnetic glyconanoparticles have been prepared and shown to be powerful in probing lectins displayed on pathogenic and mammalian cell surfaces. Blood group H1- and Le(b)-conjugated nanoparticles were found to bind to BabA displaying Helicobacter pylori, and Le(a)- and Le(b)-modified nanoparticles are both recognized by and internalized into DC-SIGN and SIGN-R1 expressing mammalian cells via lectin-mediated endocytosis. In addition, glyconanoparticles block adhesion of H. pylori to mammalian cells, suggesting that they can serve as inhibitors of infection of host cells by this pathogen. It has been also shown that owing to their magnetic properties, glyconanoparticles are useful tools to enrich lectin expressing cells. The combined results indicate that dual-modal glyconanoparticles are biocompatible and that they can be employed in lectin-associated biological studies and biomedical applications.
Collapse
Affiliation(s)
- Sungjin Park
- †Department of Chemistry, Yonsei University, Seoul 120-749 Korea
| | - Gun-Hee Kim
- †Department of Chemistry, Yonsei University, Seoul 120-749 Korea
| | - Seong-Hyun Park
- †Department of Chemistry, Yonsei University, Seoul 120-749 Korea
| | - Jaeyoung Pai
- †Department of Chemistry, Yonsei University, Seoul 120-749 Korea
| | - Dominea Rathwell
- †Department of Chemistry, Yonsei University, Seoul 120-749 Korea
| | - Jin-Yeon Park
- ‡Department of Veterinary Medicine, Department of Biomedical Science and Technology, Konkuk University, Seoul 143-701 Korea
| | - Young-Sun Kang
- ‡Department of Veterinary Medicine, Department of Biomedical Science and Technology, Konkuk University, Seoul 143-701 Korea
| | - Injae Shin
- †Department of Chemistry, Yonsei University, Seoul 120-749 Korea
| |
Collapse
|
35
|
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
|
36
|
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
|
37
|
Kidane YH, Lawrence C, Murali TM. Computational approaches for discovery of common immunomodulators in fungal infections: towards broad-spectrum immunotherapeutic interventions. BMC Microbiol 2013; 13:224. [PMID: 24099000 PMCID: PMC3853472 DOI: 10.1186/1471-2180-13-224] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 09/17/2013] [Indexed: 01/16/2023] Open
Abstract
Background Fungi are the second most abundant type of human pathogens. Invasive fungal pathogens are leading causes of life-threatening infections in clinical settings. Toxicity to the host and drug-resistance are two major deleterious issues associated with existing antifungal agents. Increasing a host’s tolerance and/or immunity to fungal pathogens has potential to alleviate these problems. A host’s tolerance may be improved by modulating the immune system such that it responds more rapidly and robustly in all facets, ranging from the recognition of pathogens to their clearance from the host. An understanding of biological processes and genes that are perturbed during attempted fungal exposure, colonization, and/or invasion will help guide the identification of endogenous immunomodulators and/or small molecules that activate host-immune responses such as specialized adjuvants. Results In this study, we present computational techniques and approaches using publicly available transcriptional data sets, to predict immunomodulators that may act against multiple fungal pathogens. Our study analyzed data sets derived from host cells exposed to five fungal pathogens, namely, Alternaria alternata, Aspergillus fumigatus, Candida albicans, Pneumocystis jirovecii, and Stachybotrys chartarum. We observed statistically significant associations between host responses to A. fumigatus and C. albicans. Our analysis identified biological processes that were consistently perturbed by these two pathogens. These processes contained both immune response-inducing genes such as MALT1, SERPINE1, ICAM1, and IL8, and immune response-repressing genes such as DUSP8, DUSP6, and SPRED2. We hypothesize that these genes belong to a pool of common immunomodulators that can potentially be activated or suppressed (agonized or antagonized) in order to render the host more tolerant to infections caused by A. fumigatus and C. albicans. Conclusions Our computational approaches and methodologies described here can now be applied to newly generated or expanded data sets for further elucidation of additional drug targets. Moreover, identified immunomodulators may be used to generate experimentally testable hypotheses that could help in the discovery of broad-spectrum immunotherapeutic interventions. All of our results are available at the following supplementary website: http://bioinformatics.cs.vt.edu/~murali/supplements/2013-kidane-bmc
Collapse
Affiliation(s)
- Yared H Kidane
- Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, VA 24061, USA.
| | | | | |
Collapse
|
38
|
Eriksson M, Johannssen T, von Smolinski D, Gruber AD, Seeberger PH, Lepenies B. The C-Type Lectin Receptor SIGNR3 Binds to Fungi Present in Commensal Microbiota and Influences Immune Regulation in Experimental Colitis. Front Immunol 2013; 4:196. [PMID: 23882266 PMCID: PMC3712271 DOI: 10.3389/fimmu.2013.00196] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 07/02/2013] [Indexed: 01/14/2023] Open
Abstract
Inflammatory bowel disease is a condition of acute and chronic inflammation of the gut. An important factor contributing to pathogenesis is a dysregulated mucosal immunity against commensal bacteria and fungi. Host pattern-recognition receptors (PRRs) sense commensals in the gut and are involved in maintaining the balance between controlled responses to pathogens and overwhelming innate immune activation. C-type lectin receptors (CLRs) are PRRs recognizing glycan structures on pathogens and self-antigens. Here we examined the role of the murine CLR specific intracellular adhesion molecule-3 grabbing non-integrin homolog-related 3 (SIGNR3) in the recognition of commensals and its involvement in intestinal immunity. SIGNR3 is the closest murine homolog of the human dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) receptor recognizing similar carbohydrate ligands such as terminal fucose or high-mannose glycans. We discovered that SIGNR3 recognizes fungi present in the commensal microbiota. To analyze whether this interaction impacts the intestinal immunity against microbiota, the dextran sulfate sodium-induced colitis model was employed. SIGNR3−/− mice exhibited an increased weight loss associated with more severe colitis symptoms compared to wild-type control mice. The increased inflammation in SIGNR3−/− mice was accompanied by a higher level of TNF-α in colon. Our findings demonstrate for the first time that SIGNR3 recognizes intestinal fungi and has an immune regulatory role in colitis.
Collapse
Affiliation(s)
- Magdalena Eriksson
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces , Potsdam , Germany ; Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin , Berlin , Germany
| | | | | | | | | | | |
Collapse
|
39
|
Rizzetto L, De Filippo C, Rivero D, Riccadonna S, Beltrame L, Cavalieri D. Systems biology of host-mycobiota interactions: dissecting Dectin-1 and Dectin-2 signalling in immune cells with DC-ATLAS. Immunobiology 2013; 218:1428-37. [PMID: 23932568 DOI: 10.1016/j.imbio.2013.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 07/02/2013] [Accepted: 07/06/2013] [Indexed: 01/04/2023]
Abstract
Modelling the networks sustaining the fruitful coexistence between fungi and their mammalian hosts is becoming increasingly important to control emerging fungal pathogens. The C-type lectins Dectin-1 and Dectin-2 are involved in host defense mechanisms against fungal infection driving inflammatory and adaptive immune responses and complement in containing fungal burdens. Recognizing carbohydrate structures in pathogens, their engagement induces maturation of dendritic cells (DCs) into potent immuno-stimulatory cells endowed with the capacity to efficiently prime T cells. Owing to these properties, Dectin-1 and Dectin-2 agonists are currently under investigation as promising adjuvants in vaccination procedures for the treatment of fungal infection. Thus, a detailed understanding of events' cascade specifically triggered in DCs upon engagement is of great interest in translational research. Here, we summarize the current knowledge on Dectin-1 and Dectin-2 signalling in DCs highlighting similarities and differences. Detailed maps are annotated, using the Biological Connection Markup Language (BCML) data model, and stored in DC-ATLAS, a versatile resource for the interpretation of high-throughput data generated perturbing the signalling network of DCs.
Collapse
Affiliation(s)
- Lisa Rizzetto
- Fondazione Edmund Mach, Research and Innovation Centre, San Michele all'Adige (TN), Italy
| | | | | | | | | | | |
Collapse
|
40
|
Beltrame L, Bianco L, Fontana P, Cavalieri D. Pathway Processor 2.0: a web resource for pathway-based analysis of high-throughput data. ACTA ACUST UNITED AC 2013; 29:1825-6. [PMID: 23740747 PMCID: PMC3702260 DOI: 10.1093/bioinformatics/btt292] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Summary: Pathway Processor 2.0 is a web application designed to analyze high-throughput datasets, including but not limited to microarray and next-generation sequencing, using a pathway centric logic. In addition to well-established methods such as the Fisher’s test and impact analysis, Pathway Processor 2.0 offers innovative methods that convert gene expression into pathway expression, leading to the identification of differentially regulated pathways in a dataset of choice. Availability and implementation: Pathway Processor 2.0 is available as a web service at http://compbiotoolbox.fmach.it/pathwayProcessor/. Sample datasets to test the functionality can be used directly from the application. Contact:duccio.cavalieri@fmach.it Supplementary information:Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Luca Beltrame
- Translational Genomics Unit, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via La Masa 19, 20159 Milano, Italy
| | | | | | | |
Collapse
|
41
|
Rizzetto L, Giovannini G, Bromley M, Bowyer P, Romani L, Cavalieri D. Strain dependent variation of immune responses to A. fumigatus: definition of pathogenic species. PLoS One 2013; 8:e56651. [PMID: 23441211 PMCID: PMC3575482 DOI: 10.1371/journal.pone.0056651] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 01/11/2013] [Indexed: 11/19/2022] Open
Abstract
For over a century microbiologists and immunologist have categorized microorganisms as pathogenic or non-pathogenic species or genera. This definition, clearly relevant at the strain and species level for most bacteria, where differences in virulence between strains of a particular species are well known, has never been probed at the strain level in fungal species. Here, we tested the immune reactivity and the pathogenic potential of a collection of strains from Aspergillus spp, a fungus that is generally considered pathogenic in immuno-compromised hosts. Our results show a wide strain-dependent variation of the immune response elicited indicating that different isolates possess diverse virulence and infectivity. Thus, the definition of markers of inflammation or pathogenicity cannot be generalized. The profound understanding of the molecular mechanisms subtending the different immune responses will result solely from the comparative study of strains with extremely diverse properties.
Collapse
Affiliation(s)
- Lisa Rizzetto
- Department of Neuroscience, Pharmacology and Child’s Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Gloria Giovannini
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Michael Bromley
- National Aspergillosis Centre and Mycology Reference Centre, University Hospital of South Manchester, University of Manchester, Manchester, United Kingdom
- School of Translational Medicine, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Paul Bowyer
- National Aspergillosis Centre and Mycology Reference Centre, University Hospital of South Manchester, University of Manchester, Manchester, United Kingdom
- School of Translational Medicine, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Luigina Romani
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Duccio Cavalieri
- Department of Neuroscience, Pharmacology and Child’s Health (NEUROFARBA), University of Florence, Florence, Italy
- Innovation and Research Center, Edmund Mach Fondation, San Michele all’Adige (TN), Italy
- * E-mail:
| |
Collapse
|
42
|
Chow BDW, Linden JR, Bliss JM. Candida parapsilosis and the neonate: epidemiology, virulence and host defense in a unique patient setting. Expert Rev Anti Infect Ther 2013; 10:935-46. [PMID: 23030332 DOI: 10.1586/eri.12.74] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Invasive candidiasis is a common problem in premature infants that leads to high morbidity and mortality. Although Candida albicans has historically been the most prominent species involved in these infections and has therefore been the subject of the most study, Candida parapsilosis is increasing in frequency, and neonates are disproportionately affected. This article reviews unique aspects of the epidemiology of this organism as well as strategies for prophylaxis against invasive candidiasis in general. Additionally, important differences between C. parapsilosis and C. albicans are coming to light related to virulence determinants and interactions with components of host immunity. These developments are reviewed while highlighting the significant gaps in our understanding that remain to be elucidated.
Collapse
Affiliation(s)
- Brian D W Chow
- Department of Pediatrics, Hasbro Children's Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | | | | |
Collapse
|
43
|
Linden JR, Kunkel D, Laforce-Nesbitt SS, Bliss JM. The role of galectin-3 in phagocytosis of Candida albicans and Candida parapsilosis by human neutrophils. Cell Microbiol 2013; 15:1127-42. [PMID: 23279221 DOI: 10.1111/cmi.12103] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 12/11/2012] [Accepted: 12/17/2012] [Indexed: 11/30/2022]
Abstract
Candida albicans causes the majority of invasive candidiasis in immunocompromised adults while Candida parapsilosis is a leading cause of neonatal candidiasis. While much work has focused on how the immune system recognizes and responds to C. albicans, less is known about host interaction with C. parapsilosis. This study investigates the human neutrophil phagocytic response to these species. Neutrophils underwent phagocytosis of C. parapsilosis yeast and C. albicans hyphae much more efficiently than C. albicans yeast. Treatment of neutrophils with a galectin-3 (gal3) blocking antibody inhibited phagocytosis of C. parapsilosis yeast and C. albicans hyphae, but not C. albicans yeast. The majority of neutrophil gal3 was expressed intracellularly and was secreted from neutrophils after treatment with C. parapsilosis mannan. When neutrophils were treated with exogenous gal3, phagocytosis of both C. albicans and C. parapsilosis yeast increased. Exposure of neutrophils to C. parapsilosis yeast increased phagocytosis of C. albicans yeast and was inhibited by gal3 blocking antibody. Taken together, these data indicate that gal3 secreted from neutrophils may act as a pro-inflammatory autocrine/paracrine signal in neutrophil phagocytosis and suggest that gal3 has a unique role in neutrophil response to C. parapsilosis yeast and C. albicans hyphae distinct from C. albicans yeast.
Collapse
Affiliation(s)
- Jennifer R Linden
- Graduate Program in Pathobiology, Brown University, Providence, RI, USA
| | | | | | | |
Collapse
|
44
|
Mannose Receptor Ligands Regulate the Gene Expression of Toll-like Receptors in Chicken Monocytes. J Poult Sci 2013. [DOI: 10.2141/jpsa.0120178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
45
|
Abstract
Pathologies attributable to fungal infections represent a growing concern in both developed and developing countries. Initially discovered as opportunistic pathogens of immunocompromised hosts, fungi such as Candida albicans are now being placed at the centre of a more complex and dynamic picture in which the outcome of an infection is the result of an intricate network of molecular interactions between the fungus, the host and the commensal microflora co-inhabiting various host niches, and especially the gastrointestinal (GI) tract. The complexity of the host-fungal interaction begins with the numerous pathogen-associated molecular patterns (PAMPs) present on the fungal cell wall that are recognized by multiple pathogen-recognition receptors (PRRs), expressed by several types of host cells. PAMP-PRR interactions elicit a variety of intracellular signalling pathways leading to a wide array of immune responses, some of which promote fungal clearance while others contribute to pathogenesis. The picture is further complicated by the fact that numerous commensal bacteria normally co-inhabiting the host's GI tract produce molecules that either directly modulate the survival and virulence of commensal fungi such as C. albicans or indirectly modulate the host's antifungal immune responses. On top of this complexity, this host-microbiome-fungal interaction exhibits features of a dynamic system, in which the same fungi can easily switch between different morphological forms presenting different PAMPs at different moments of time. Furthermore, fungal pathogens can rapidly accumulate genomic alterations that further modify their recognition by the immune system, their virulence and their resistance to antifungal compounds. Thus, based on available molecular data alone, it is currently difficult to construct a coherent model able to explain the balance between commensalism and virulence and to predict the outcome of a fungal infection. Here, we review current advances in our understanding of this complex and dynamic system and propose new avenues of investigation to assemble a more complete picture of the host-fungal interaction, integrating microbiological and immunological data under the lens of systems biology and evolutionary genomics.
Collapse
|
46
|
Rizzetto L, Buschow SI, Beltrame L, Figdor CG, Schierer S, Schuler G, Cavalieri D. The modular nature of dendritic cell responses to commensal and pathogenic fungi. PLoS One 2012; 7:e42430. [PMID: 22879980 PMCID: PMC3411757 DOI: 10.1371/journal.pone.0042430] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 07/09/2012] [Indexed: 11/29/2022] Open
Abstract
The type of adaptive immune response following host-fungi interaction is largely determined at the level of the antigen-presenting cells, and in particular by dendritic cells (DCs). The extent to which transcriptional regulatory events determine the decision making process in DCs is still an open question. By applying the highly structured DC-ATLAS pathways to analyze DC responses, we classified the various stimuli by revealing the modular nature of the different transcriptional programs governing the recognition of either pathogenic or commensal fungi. Through comparison of the network parts affected by DC stimulation with fungal cells and purified single agonists, we could determine the contribution of each receptor during the recognition process. We observed that initial recognition of a fungus creates a temporal window during which the simultaneous recruitment of cell surface receptors can intensify, complement and sustain the DC activation process. The breakdown of the response to whole live cells, through the purified components, showed how the response to invading fungi uses a set of specific modules. We find that at the start of fungal recognition, DCs rapidly initiate the activation process. Ligand recognition is further enhanced by over-expression of the receptor genes, with a significant correspondence between gene expression and protein levels and function. Then a marked decrease in the receptor levels follows, suggesting that at this moment the DC commits to a specific fate. Overall our pathway based studies show that the temporal window of the fungal recognition process depends on the availability of ligands and is different for pathogens and commensals. Modular analysis of receptor and signalling-adaptor expression changes, in the early phase of pathogen recognition, is a valuable tool for rapid and efficient dissection of the pathogen derived components that determine the phenotype of the DC and thereby the type of immune response initiated.
Collapse
Affiliation(s)
- Lisa Rizzetto
- Department of Preclinical and Clinical Pharmacology, University of Florence, Florence, Italy
| | | | | | | | | | | | | |
Collapse
|
47
|
Tierney L, Kuchler K, Rizzetto L, Cavalieri D. Systems biology of host-fungus interactions: turning complexity into simplicity. Curr Opin Microbiol 2012; 15:440-6. [PMID: 22717554 PMCID: PMC3501689 DOI: 10.1016/j.mib.2012.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 04/24/2012] [Accepted: 05/01/2012] [Indexed: 12/15/2022]
Abstract
Modeling interactions between fungi and their hosts at the systems level requires a molecular understanding both of how the host orchestrates immune surveillance and tolerance, and how this activation, in turn, affects fungal adaptation and survival. The transition from the commensal to pathogenic state, and the co-evolution of fungal strains within their hosts, necessitates the molecular dissection of fungal traits responsible for these interactions. There has been a dramatic increase in publically available genome-wide resources addressing fungal pathophysiology and host–fungal immunology. The integration of these existing data and emerging large-scale technologies addressing host–pathogen interactions requires novel tools to connect genome-wide data sets and theoretical approaches with experimental validation so as to identify inherent and emerging properties of host–pathogen relationships and to obtain a holistic view of infectious processes. If successful, a better understanding of the immune response in health and microbial diseases will eventually emerge and pave the way for improved therapies.
Collapse
Affiliation(s)
- Lanay Tierney
- Medical University of Vienna, Christian Doppler Laboratory Infection Biology, Max F. Perutz Laboratories, A-1030 Vienna, Austria
| | - Karl Kuchler
- Medical University of Vienna, Christian Doppler Laboratory Infection Biology, Max F. Perutz Laboratories, A-1030 Vienna, Austria
| | - Lisa Rizzetto
- Department of Preclinical and Clinical Pharmacology, University of Florence, 50139 Firenze, Italy
| | - Duccio Cavalieri
- Department of Preclinical and Clinical Pharmacology, University of Florence, 50139 Firenze, Italy
- Research and Innovation Centre, Edmund Mach Foundation, San Michele all’Adige, 38010, Trento, Italy
| |
Collapse
|
48
|
Espinosa V, Rivera A. Cytokines and the regulation of fungus-specific CD4 T cell differentiation. Cytokine 2011; 58:100-6. [PMID: 22133343 DOI: 10.1016/j.cyto.2011.11.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 11/02/2011] [Accepted: 11/03/2011] [Indexed: 12/11/2022]
Abstract
CD4 T cells play important and non-redundant roles in protection against infection with diverse fungi. Distinct CD4 T cell subsets can mediate protection against fungal disease where Th1 and Th17 CD4 T cell subsets have been found to promote fungal clearance and protective immunity against diverse fungal pathogens. The differentiation of naïve CD4 T cells into Th1 or Th17 cells is crucially controlled by their interaction with dendritic cells and instructed by cytokines. IL-12 and IFN-γ promote Th1 differentiation while TGF-β, IL-6, IL-1, IL-21 and IL-23 promote Th17 differentiation and maintenance. The production of these cytokines by DCs is in turn regulated by innate receptors triggered in response to fungal infection. In this review we will discuss the contributions of cytokines found to influence fungus-specific CD4 T cell differentiation and their role in defense against fungal disease. We will also highlight the contributions of innate receptors involved in recognition of fungi and how they shape cytokine secretion and CD4 T cell differentiation.
Collapse
Affiliation(s)
- Vanessa Espinosa
- Graduate School of Biomedical Sciences, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, 185 So Orange Avenue, MSB-F601, Newark, NJ 07101, USA.
| | | |
Collapse
|
49
|
Friend or foe: using systems biology to elucidate interactions between fungi and their hosts. Trends Microbiol 2011; 19:509-15. [DOI: 10.1016/j.tim.2011.07.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 07/26/2011] [Accepted: 07/27/2011] [Indexed: 11/20/2022]
|
50
|
The economy of inflammation: when is less more? Trends Parasitol 2011; 27:382-7. [DOI: 10.1016/j.pt.2011.05.004] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 05/12/2011] [Accepted: 05/12/2011] [Indexed: 12/23/2022]
|