1
|
Doan HT, Chiu YL, Cheng LC, Coad RA, Chiang HS. Candida tropicalis Alters Barrier Permeability and Claudin-1 Organization in Intestinal Epithelial Cells. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2025; 68:67-76. [PMID: 39918057 DOI: 10.4103/ejpi.ejpi-d-24-00090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/09/2024] [Indexed: 02/14/2025]
Abstract
ABSTRACT Inflammatory bowel disease (IBD) is an autoimmune disorder characterized by chronic inflammation of the gut and compromised intestinal barrier function, resulting from aberrant immune responses targeting the intestinal microbiota. While the involvement of Candida albicans in IBD pathogenesis is well-documented, the role of non- albicans Candida species in IBD remains less understood. Recent studies have identified a correlation between elevated levels of Candida tropicalis , a notable non- albicans opportunistic fungus, and the development of IBD. However, the precise impact of C. tropicalis on intestinal barrier function is not well elucidated. To address this knowledge gap, we utilized a cell model comprising polarized Caco-2 monolayers, which mimic the intestinal epithelium, to investigate the interaction between C. tropicalis and intestinal barrier function. Our results showed that incubation with C. tropicalis influenced transepithelial electrical resistance and increased permeability to the small molecule lucifer yellow, but did not affect permeability to the larger molecule fluorescein isothiocyanate-dextran. In addition, we observed internalization of the tight junction protein claudin-1 in the Caco-2 monolayers. Further experiments using Caco-2 monolayers exposed to the dectin-1 ligand zymosan induced similar changes in the distribution of claudin-1 but did not alter monolayer permeability. These findings suggest that C. tropicalis specifically affects intestinal barrier integrity and permeability to smaller solutes in intestinal epithelial cells.
Collapse
Affiliation(s)
- Ha The Doan
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yi-Ling Chiu
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Li-Chieh Cheng
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Rae Apaivongse Coad
- Department of Life Science, National Taiwan University, Taipei, Taiwan
- Program in Biological Sciences, Mahidol University International College, Nakorn Pathom, Thailand
| | - Hao-Sen Chiang
- Department of Life Science, National Taiwan University, Taipei, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
2
|
Huang Y, Wang Y, Huang X, Yu X. Unveiling the overlooked fungi: the vital of gut fungi in inflammatory bowel disease and colorectal cancer. Gut Pathog 2024; 16:59. [PMID: 39407244 PMCID: PMC11481806 DOI: 10.1186/s13099-024-00651-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
The fungi of the human microbiota play important roles in the nutritional metabolism and immunological balance of the host. Recently, research has increasingly emphasised the role of fungi in modulating inflammation in intestinal diseases and maintaining health in this environment. It is therefore necessary to understand more clearly the interactions and mechanisms of the microbiota/pathogen/host relationship and the resulting inflammatory processes, as well as to offer new insights into the prevention, diagnosis and treatment of inflammatory bowel disease (IBD), colorectal cancer (CRC) and other intestinal pathologies. In this review, we comprehensively elucidate the fungal-associated pathogenic mechanisms of intestinal inflammation in IBD and related CRC, with an emphasis on three main aspects: the direct effects of fungi and their metabolites on the host, the indirect effects mediated by interactions with other intestinal microorganisms and the immune regulation of the host. Understanding these mechanisms will enable the development of innovative approaches based on the use of fungi from the resident human microbiota such as dietary interventions, fungal probiotics and faecal microbiota transplantation in the prevention, diagnosis and treatment of intestinal diseases.
Collapse
Affiliation(s)
- Yilin Huang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Huankui Academy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Yang Wang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiaotian Huang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Xiaomin Yu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
3
|
Xu T, Liu Y, Zhang W, Li M, Zhang L, Li X, Zhang Y, Yue L, Li S, Lin Y, Zou X, Chen F. Specific cell subclusters of dental pulp stem cells respond to distinct pathogens through the ROS pathway. Front Cell Infect Microbiol 2024; 14:1452124. [PMID: 39328360 PMCID: PMC11424553 DOI: 10.3389/fcimb.2024.1452124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/12/2024] [Indexed: 09/28/2024] Open
Abstract
Introduction Microbial pathogens invade various human organs, including the oral cavity. Candida albicans (C.a) and Streptococcus mutans (S.m) served respectively as representative oral pathogenic fungi and bacteria to stimulate dental pulp stem cells (DPSCs) and to screen the DPSC subcluster that specifically responded to fungal infection. Methods DPSCs were obtained from the impacted third molars of six healthy subjects. Then, cells were mixed and divided into three samples, two of which were stimulated with C.a and S.m, respectively; the third sample was exposed to cell medium only (Ctrl). Single-cell mRNA sequencing analysis of treated DPSCs was performed. Results DPSCs were composed of four major clusters of which one, DPSC.7, exhibited unique changes compared to those of other subclusters. The DPSC.7 cell percentage of the C.a sample was twice those of the Ctrl and S.m samples. DPSC.7 cells expressed genes associated with the response to reactive oxygen species (ROS) response. DPSC.7 subgroup cells established characteristic aggregation under the stimulation of different pathogens in UMAP. The MAPK/ERK1/2 and NF-κB pathways were up-regulated, DUSP1/5/6 expressions were suppressed, FOS synthesis was activated, the immune-related pathway was induced, and the levels of cytokines, including IL-6 and CCL2, were up-regulated in DPSC.7 cells when stimulated with C.a. Conclusions Our study analyzed the cellular and molecular properties of DPSCs infected by oral fungi and bacteria with single-cell RNA sequencing. A subcluster of DPSCs responded specifically to infections with different pathogens, activating the MAPK and NF-κB pathways to induce immune responses via the ROS pathway. This suggests novel treatment strategies for fungal infections.
Collapse
Affiliation(s)
- Tiansong Xu
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Fifth Clinical Division, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Yangjia Liu
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Wen Zhang
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Department of Stomatology, Peking University International Hospital, Beijing, China
| | - Murong Li
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Liqi Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Xueying Li
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Yifei Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Lin Yue
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Sha Li
- Department of Implantology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Ye Lin
- Department of Implantology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Xiaoying Zou
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Center of Stomatology, Peking University Hospital, Beijing, China
| | - Feng Chen
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| |
Collapse
|
4
|
Sprague JL, Schille TB, Allert S, Trümper V, Lier A, Großmann P, Priest EL, Tsavou A, Panagiotou G, Naglik JR, Wilson D, Schäuble S, Kasper L, Hube B. Candida albicans translocation through the intestinal epithelial barrier is promoted by fungal zinc acquisition and limited by NFκB-mediated barrier protection. PLoS Pathog 2024; 20:e1012031. [PMID: 38427950 PMCID: PMC10907035 DOI: 10.1371/journal.ppat.1012031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 02/06/2024] [Indexed: 03/03/2024] Open
Abstract
The opportunistic fungal pathogen Candida albicans thrives on human mucosal surfaces as a harmless commensal, but frequently causes infections under certain predisposing conditions. Translocation across the intestinal barrier into the bloodstream by intestine-colonizing C. albicans cells serves as the main source of disseminated candidiasis. However, the host and microbial mechanisms behind this process remain unclear. In this study we identified fungal and host factors specifically involved in infection of intestinal epithelial cells (IECs) using dual-RNA sequencing. Our data suggest that host-cell damage mediated by the peptide toxin candidalysin-encoding gene ECE1 facilitates fungal zinc acquisition. This in turn is crucial for the full virulence potential of C. albicans during infection. IECs in turn exhibit a filamentation- and damage-specific response to C. albicans infection, including NFκB, MAPK, and TNF signaling. NFκB activation by IECs limits candidalysin-mediated host-cell damage and mediates maintenance of the intestinal barrier and cell-cell junctions to further restrict fungal translocation. This is the first study to show that candidalysin-mediated damage is necessary for C. albicans nutrient acquisition during infection and to explain how IECs counteract damage and limit fungal translocation via NFκB-mediated maintenance of the intestinal barrier.
Collapse
Affiliation(s)
- Jakob L. Sprague
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
| | - Tim B. Schille
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
| | - Stefanie Allert
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
| | - Verena Trümper
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
| | - Adrian Lier
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
| | - Peter Großmann
- Department of Microbiome Dynamics, Hans-Knöll-Institute, Jena, Germany
| | - Emily L. Priest
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Antzela Tsavou
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Gianni Panagiotou
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Department of Microbiome Dynamics, Hans-Knöll-Institute, Jena, Germany
- Institute of Microbiology, Friedrich-Schiller-University Jena, Jena, Germany
| | - Julian R. Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Duncan Wilson
- Medical Research Council, Centre for Medical Mycology at the University of Exeter, Exeter, United Kingdom
| | - Sascha Schäuble
- Department of Microbiome Dynamics, Hans-Knöll-Institute, Jena, Germany
| | - Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Institute of Microbiology, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
5
|
Song YY, Zhang XZ, Wang BN, Cheng YK, Guo X, Zhang X, Long SR, Liu RD, Wang ZQ, Cui J. A novel Trichinella spiralis serine proteinase disrupted gut epithelial barrier and mediated larval invasion through binding to RACK1 and activating MAPK/ERK1/2 pathway. PLoS Negl Trop Dis 2024; 18:e0011872. [PMID: 38190388 PMCID: PMC10798628 DOI: 10.1371/journal.pntd.0011872] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 01/19/2024] [Accepted: 12/19/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Gut epithelium is the first natural barrier against Trichinella spiralis larval invasion, but the mechanism by which larval penetration of gut epithelium is not completely elucidated. Previous studies showed that proteases secreted by T. spiralis intestinal infective larvae (IIL) degraded tight junctions (TJs) proteins of gut epithelium and mediated larval invasion. A new T. spiralis serine proteinase (TsSPc) was identified in the IIL surface proteins and ES proteins, rTsSPc bound to the intestinal epithelial cell (IECs) and promoted larval invasion of IECs. The aim of this study was to characterize the interacted proteins of TsSPc and IECs, and to investigate the molecular mechanisms of TsSPc mediating larval invasion of gut mucosa. METHODOLOGY/PRINCIPAL FINDING IIFT results showed natural TsSPc was detected in infected murine intestine at 6, 12 hours post infection (hpi) and 3 dpi. The results of GST pull-down, mass spectrometry (MS) and Co-IP indicated that rTsSPc bound and interacted specifically with receptor for activated protein C kinase 1 (RACK1) in Caco-2 cells. rTsSPc did not directly hydrolyze the TJs proteins. qPCR and Western blot showed that rTsSPc up-regulated RACK1 expression, activated MAPK/ERK1/2 pathway, reduced the expression levels of gut TJs (occludin and claudin-1) and adherent protein E-cad, increased the paracellular permeability and damaged the integrity of intestinal epithelial barrier. Moreover, the RACK1 inhibitor HO and ERK1/2 pathway inhibitor PD98059 abolished the rTsSPc activating ERK1/2 pathway, they also inhibited and abrogated the rTsSPc down-regulating expression of occludin, claudin-1 and E-cad in Caco-2 monolayer and infected murine intestine, impeded larval invasion and improved intestinal epithelial integrity and barrier function, reduced intestinal worm burdens and alleviated intestinal inflammation. CONCLUSIONS rTsSPc bound to RACK1 receptor in gut epithelium, activated MAPK/ERK1/2 pathway, decreased the expression of gut epithelial TJs proteins and disrupted the epithelial integrity, consequently mediated T. spiralis larval invasion of gut epithelium. The results are valuable to understand T. spiralis invasion mechanism, and TsSPc might be regarded as a vaccine target against T. spiralis invasion and infection.
Collapse
Affiliation(s)
- Yan Yan Song
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Xin Zhuo Zhang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Bo Ning Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Yong Kang Cheng
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Xi Zhang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Shao Rong Long
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Ruo Dan Liu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Zhong Quan Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Jing Cui
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
Zhu H, Zhang Y, Du S, Wang H, Zheng Y. Colonic mucosal biopsy location can not affect the results of mucosal metabolomics and mucosal microbiota analysis in IBS. Front Med (Lausanne) 2023; 10:1183484. [PMID: 37351069 PMCID: PMC10282601 DOI: 10.3389/fmed.2023.1183484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/22/2023] [Indexed: 06/24/2023] Open
Abstract
Objective To compare and analyze the mucosal metabolites and mucosal microbiota of different parts of colon in patients with IBS. Methods A total of 10 patients with IBS-D and six healthy controls (HC) were enrolled. All enrolled participants underwent two biopsies of the ileocecal and sigmoid colon during colonoscopy. Metabolomic profiling of one piece of tissue was conducted using desorption electrospray ionization-mass spectrometry (DESI-MS), and the gut flora of the other piece was examined using 16S rRNA sequencing. The metabolic profiles and flora of the ileocecal and sigmoid colonic mucosa in each group were further analyzed in this study. Results (1) Principal components analysis (PCA) indicated that mucosal metabolites did not differ in different parts of the colon in either the IBS-D or HC groups. (2) In the mucosal microbiome analyses, no differences between the microbiota of the two parts of the colon were found by using Principal Co-ordinates Analysis (PCoA). In IBS group, comparing with sigmoid mucosa, the chao1 richness indice was higher and the Shannon index was lower in the ileocecal mucosa (p = 0.40, p = 0.22). However, in the HC group, microbiome analysis of the ileocecal mucosa showed lower values for Chao 1 and Shannon indices than those of the sigmoid colon mucosa (p = 0.06, p = 0.86). (3) Compared with the HC group, 1,113 metabolic signal peaks were upregulated, whereas 594 metabolites were downregulated in the IBS-D samples. Moreover, the PCA of the metabolites showed significant separation between the IBS-D and HC groups. (4) Chao1 expression was significantly higher in the mucosal microbiota with IBS-D than in the HC (p = 0.03). The Shannon index was lower in IBS-D, but the difference was not statistically significant (p = 0.53). PCoA revealed a significant difference in the microflora structure between the IBS-D and HC groups. Conclusion The mucosal metabolic profile and mucosal flora structure of the colon were similar, despite different locations in IBS and healthy subjects. IBS had abnormal colonic mucosal metabolism and flora disturbances.
Collapse
Affiliation(s)
- Huiting Zhu
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
- Department of Gastroenterology, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Yanli Zhang
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| | - Shiyu Du
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| | - Huifen Wang
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| | - Yue Zheng
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
- Department of Gastroenterology, First Hospital of Qinhuangdao, Qinhuangdao, China
| |
Collapse
|
7
|
Chen X, Luo D, Jia G, Zhao H, Liu G, Huang Z. L-theanine attenuates porcine intestinal tight junction damage induced by LPS via p38 MAPK/NLRP3 signaling in IPEC-J2 cells. Food Chem Toxicol 2023:113870. [PMID: 37271275 DOI: 10.1016/j.fct.2023.113870] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/27/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023]
Abstract
L-theanine is a natural bioactive component in tea leaves and has anti-inflammatory effects. The study aimed to investigated the effects and underlying mechanisms of L-theanine on lipopolysaccharide (LPS)-induced intestinal tight junction damage in IPEC-J2 cells. Results showed that LPS induced tight junction damage by increasing reactive oxygen species production and lactate dehydrogenase (LDH) release and decreasing the mRNA expression of tight junction proteins related genes zonula occludens-1 (ZO-1, also known as Tjp1), Occludin and Claudin-1, while L-theanine reversed such an effect and attenuated the increase of p38 mitogen-activated protein kinase (p38 MAPK) mRNA expression. The p38 MAPK inhibitor (SB203580) attenuated the mRNA expression of nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (Nlrp3) inflammasome and interleukin-1β (Il-1β), and increased the mRNA expression of Tjp1, Occludin and Claudin-1, which showed a similar effect with L-theanine. In addition, NLRP3 inhibitor MCC950 attenuated the Il-1β expression and LDH release, while increased the expression of tight-junction protein-related genes. In conclusion, L-theanine could protect LPS-induced intestinal tight junction damage by inhibiting the activation of p38 MAPK-mediated NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Xiaoling Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Diaoyun Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Gang Jia
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Hua Zhao
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Guangmang Liu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China.
| |
Collapse
|
8
|
Sprague JL, Kasper L, Hube B. From intestinal colonization to systemic infections: Candida albicans translocation and dissemination. Gut Microbes 2022; 14:2154548. [PMID: 36503341 PMCID: PMC9746630 DOI: 10.1080/19490976.2022.2154548] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Candida species are the most prevalent cause of invasive fungal infections, of which Candida albicans is the most common. Translocation across the epithelial barrier into the bloodstream by intestinal-colonizing C. albicans cells serves as the main source for systemic infections. Understanding the fungal mechanisms behind this process will give valuable insights on how to prevent such infections and keep C. albicans in the commensal state in patients with predisposing conditions. This review will focus on recent developments in characterizing fungal translocation mechanisms, compare what we know about enteric bacterial pathogens with C. albicans, and discuss the different proposed hypotheses for how C. albicans enters and disseminates through the bloodstream immediately following translocation.
Collapse
Affiliation(s)
- Jakob L. Sprague
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany,Contact: Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Beutenbergstrasse 11a, 07745Jena, Germany
| | - Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany,Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
9
|
Singh S, Singh S, Kumar A. Systemic Candida albicans Infection in Mice Causes Endogenous Endophthalmitis via Breaching the Outer Blood-Retinal Barrier. Microbiol Spectr 2022; 10:e0165822. [PMID: 35913202 PMCID: PMC9431129 DOI: 10.1128/spectrum.01658-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 07/15/2022] [Indexed: 11/20/2022] Open
Abstract
Candida albicans is the leading cause of endogenous fungal endophthalmitis; however, its pathobiology studies are limited. Moreover, the contribution of host factors in the pathogenesis of Candida endophthalmitis remains unclear. In the present study, we developed a murine model of C. albicans endogenous endophthalmitis and investigated the molecular pathobiology of ocular candidiasis and blood-retinal barrier permeability. Our data show that intravenous injection of C. albicans in immunocompetent C57BL/6 mice led to endogenous endophthalmitis without causing mortality, and C. albicans was detected in the eyes at 3 days postinfection and persisted for up to 10 days. The intraocular presence of C. albicans coincided with a decrease in retinal function and increased expression of inflammatory mediators (tumor necrosis factor alpha [TNF-α], interleukin 1β [IL-1β], MIP2, and KC) and antimicrobial peptides (human β-defensins [hBDs] and LL37) in mouse retinal tissue. C. albicans infection disrupted the blood-retinal barrier (BRB) by decreasing the expression of tight junction (ZO-1) and adherens junction (E-cadherin, N/R-cadherin) proteins. In vitro studies using human retinal pigment epithelial (ARPE-19) cells showed time-dependent activation of eIF2α, extracellular signal-related kinase (ERK), and NF-κB signaling and decreased activity of AMP-activated protein kinase (AMPK) leading to the induction of an inflammatory response upon C. albicans infection. Moreover, C. albicans-infected cells exhibited increased cellular permeability coinciding with a reduction in cellular junction proteins. Overall, our study provides new insight into the molecular pathogenesis of C. albicans endogenous endophthalmitis. Furthermore, the experimental models developed in the study can be used to identify newer therapeutic targets or test the efficacy of drugs to treat and prevent fungal endophthalmitis. IMPORTANCE Patients with candidemia often experience endophthalmitis, a blinding infectious eye disease. However, the pathogenesis of Candida endophthalmitis is not well understood. Here, using in vivo and in vitro experimental models, we describe events leading to the invasion of Candida into the eye. We show that Candida from the systemic circulation disrupts the protective blood-retinal barrier and causes endogenous endophthalmitis. Our study highlights an important role of retinal pigment epithelial cells in evoking innate inflammatory and antimicrobial responses toward C. albicans infection. This study allows a better understanding of the pathobiology of fungal endophthalmitis, which can lead to the discovery of novel therapeutic targets to treat ocular fungal infections.
Collapse
Affiliation(s)
- Sneha Singh
- Department of Ophthalmology, Visual and Anatomical Sciences/Kresge Eye Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Sukhvinder Singh
- Department of Ophthalmology, Visual and Anatomical Sciences/Kresge Eye Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Ashok Kumar
- Department of Ophthalmology, Visual and Anatomical Sciences/Kresge Eye Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
10
|
Lachat J, Pascault A, Thibaut D, Le Borgne R, Verbavatz JM, Weiner A. Trans-cellular tunnels induced by the fungal pathogen Candida albicans facilitate invasion through successive epithelial cells without host damage. Nat Commun 2022; 13:3781. [PMID: 35773250 PMCID: PMC9246882 DOI: 10.1038/s41467-022-31237-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 06/09/2022] [Indexed: 11/09/2022] Open
Abstract
The opportunistic fungal pathogen Candida albicans is normally commensal, residing in the mucosa of most healthy individuals. In susceptible hosts, its filamentous hyphal form can invade epithelial layers leading to superficial or severe systemic infection. Although invasion is mainly intracellular, it causes no apparent damage to host cells at early stages of infection. Here, we investigate C. albicans invasion in vitro using live-cell imaging and the damage-sensitive reporter galectin-3. Quantitative single cell analysis shows that invasion can result in host membrane breaching at different stages and host cell death, or in traversal of host cells without membrane breaching. Membrane labelling and three-dimensional 'volume' electron microscopy reveal that hyphae can traverse several host cells within trans-cellular tunnels that are progressively remodelled and may undergo 'inflations' linked to host glycogen stores. Thus, C. albicans early invasion of epithelial tissues can lead to either host membrane breaching or trans-cellular tunnelling.
Collapse
Affiliation(s)
- Joy Lachat
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, 75013, Paris, France
| | - Alice Pascault
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, 75013, Paris, France
| | - Delphine Thibaut
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, 75013, Paris, France
| | - Rémi Le Borgne
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013, Paris, France
| | | | - Allon Weiner
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, 75013, Paris, France.
| |
Collapse
|
11
|
Huo J, Wu Z, Sun W, Wang Z, Wu J, Huang M, Wang B, Sun B. Protective Effects of Natural Polysaccharides on Intestinal Barrier Injury: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:711-735. [PMID: 35078319 DOI: 10.1021/acs.jafc.1c05966] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Owing to their minimal side effects and effective protection from oxidative stress, inflammation, and malignant growth, natural polysaccharides (NPs) are a potential adjuvant therapy for several diseases caused by intestinal barrier injury (IBI). More studies are accumulating on the protective effects of NPs with respect to IBI, but the underlying mechanisms remain unclear. Thus, this review aims to represent current studies that investigate the protective effects of NPs on IBI by directly maintaining intestinal epithelial barrier integrity (inhibiting oxidative stress, regulating inflammatory cytokine expression, and increasing tight junction protein expression) and indirectly regulating intestinal immunity and microbiota. Furthermore, the mechanisms underlying IBI development are briefly introduced, and the structure-activity relationships of polysaccharides with intestinal barrier protection effects are discussed. Potential developments and challenges associated with NPs exhibiting protective effects against IBI have also been highlighted to guide the application of NPs in the treatment of intestinal diseases caused by IBI.
Collapse
Affiliation(s)
- Jiaying Huo
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, People's Republic of China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, People's Republic of China
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510640, People's Republic of China
| | - Ziyan Wu
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, People's Republic of China
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, People's Republic of China
| | - Weizheng Sun
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510640, People's Republic of China
| | - Zhenhua Wang
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai, Shandong 264005, People's Republic of China
| | - Jihong Wu
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, People's Republic of China
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, People's Republic of China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, People's Republic of China
| | - Mingquan Huang
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, People's Republic of China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, People's Republic of China
| | - Bowen Wang
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, People's Republic of China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, People's Republic of China
| | - Baoguo Sun
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, People's Republic of China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, People's Republic of China
| |
Collapse
|
12
|
Impacts of Fructose on Intestinal Barrier Function, Inflammation and Microbiota in a Piglet Model. Nutrients 2021; 13:nu13103515. [PMID: 34684516 PMCID: PMC8541567 DOI: 10.3390/nu13103515] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 02/07/2023] Open
Abstract
The metabolic disorder caused by excessive fructose intake was reported extensively and often accompanied by intestinal barrier dysfunction. And the rising dietary fructose was consumed at an early age of human. However, related researches were almost conducted in rodent models, while in the anatomy and physiology of gastrointestinal tract, pig is more similar to human beings than rodents. Hence, weaned piglets were chosen as the model animals in our study to investigate the fructose’s impacts on intestinal tight junction, inflammation response and microbiota structure of piglets. Herein, growth performance, inflammatory response, oxidation resistance and ileal and colonic microbiota of piglet were detected after 35-day fructose supplementation. Our results showed decreased tight junction gene expressions in piglets after fructose addition, with no obvious changes in the growth performance, antioxidant resistance and inflammatory response. Moreover, fructose supplementation differently modified the microbiota structures in ileum and colon. In ileum, the proportions of Streptococcus and Faecalibacterium were higher in Fru group (fructose supplementation). In colon, the proportions of Blautia and Clostridium sensu stricto 1 were higher in Fru group. All the results suggested that tight junction dysfunction might be an earlier fructose-induced event than inflammatory response and oxidant stress and that altered microbes in ileum and colon might be the potential candidates to alleviate fructose-induced intestinal permeability alteration.
Collapse
|
13
|
Last A, Maurer M, S. Mosig A, S. Gresnigt M, Hube B. In vitro infection models to study fungal-host interactions. FEMS Microbiol Rev 2021; 45:fuab005. [PMID: 33524102 PMCID: PMC8498566 DOI: 10.1093/femsre/fuab005] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 01/14/2021] [Indexed: 12/14/2022] Open
Abstract
Fungal infections (mycoses) affect over a billion people per year. Approximately, two million of these infections are life-threatening, especially for patients with a compromised immune system. Fungi of the genera Aspergillus, Candida, Histoplasma and Cryptococcus are opportunistic pathogens that contribute to a substantial number of mycoses. To optimize the diagnosis and treatment of mycoses, we need to understand the complex fungal-host interplay during pathogenesis, the fungal attributes causing virulence and how the host resists infection via immunological defenses. In vitro models can be used to mimic fungal infections of various tissues and organs and the corresponding immune responses at near-physiological conditions. Furthermore, models can include fungal interactions with the host-microbiota to mimic the in vivo situation on skin and mucosal surfaces. This article reviews currently used in vitro models of fungal infections ranging from cell monolayers to microfluidic 3D organ-on-chip (OOC) platforms. We also discuss how OOC models can expand the toolbox for investigating interactions of fungi and their human hosts in the future.
Collapse
Affiliation(s)
- Antonia Last
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knoell Institute, Beutenbergstrasse 11a, 07745, Jena, Germany
| | - Michelle Maurer
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
- Institute of Biochemistry II, Jena University Hospital, Nonnenplan 2,07743, Jena, Germany
| | - Alexander S. Mosig
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
- Institute of Biochemistry II, Jena University Hospital, Nonnenplan 2,07743, Jena, Germany
| | - Mark S. Gresnigt
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knoell Institute, Beutenbergstrasse 11a, 07745, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knoell Institute, Beutenbergstrasse 11a, 07745, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Neugasse 24, 07743, Jena, Germany
| |
Collapse
|
14
|
Deng W, Su Z, Liang P, Ma Y, Liu Y, Zhang K, Zhang Y, Liang T, Shao J, Liu X, Han W, Li R. Single-cell immune checkpoint landscape of PBMCs stimulated with Candida albicans. Emerg Microbes Infect 2021; 10:1272-1283. [PMID: 34120578 PMCID: PMC8238073 DOI: 10.1080/22221751.2021.1942228] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Immune checkpoints play various important roles in tumour immunity, which usually contribute to T cells’ exhaustion, leading to immunosuppression in the tumour microenvironment. However, the roles of immune checkpoints in infectious diseases, especially fungal infection, remain elusive. Here, we reanalyzed a recent published single-cell RNA-sequencing (scRNA-seq) data of peripheral blood mononuclear cells (PBMCs) stimulated with Candida albicans (C. albicans), to explore the expression patterns of immune checkpoints after C. albicans bloodstream infection. We characterized the heterogeneous pathway activities among different immune cell subpopulations after C. albicans infection. The CTLA-4 pathway was up-regulated in stimulated CD4+ and CD8+ T cells, while the PD-1 pathway showed high activity in stimulated plasmacytoid dendritic cell (pDC) and monocytes. Importantly, we found that immunosuppressive checkpoints HAVCR2 and LAG3 were only expressed in stimulated NK and CD8+ T cells, respectively. Their viabilities were validated by flow cytometry. We also identified three overexpressed genes (ISG20, LY6E, ISG15) across all stimulated cells. Also, two monocyte-specific overexpressed genes (SNX10, IDO1) were screened out in this study. Together, these results supplemented the landscape of immune checkpoints in fungal infection, which may serve as potential therapeutic targets for C. albicans infection. Moreover, the genes with the most relevant for C. albicans infection were identified in this study.
Collapse
Affiliation(s)
- Weiwei Deng
- Department of Dermatology and Venerology, Peking University First Hospital, Peking University; National Clinical Research Center for Skin and Immune Diseases; Beijing Key Laboratory of Molecular Diagnosis of Dermatoses, Beijing, People's Republic of China
| | - Zhen Su
- Department of Dermatology and Venerology, The Third Affiliated Hospital of Sun Yat-Sen university, Guangzhou, People's Republic of China
| | - Panpan Liang
- Clinical laboratory, The Third Affiliated Hospital of Sun Yat-Sen university, Guangzhou, People's Republic of China
| | - Yubo Ma
- Department of Dermatology and Venerology, Peking University First Hospital, Peking University; National Clinical Research Center for Skin and Immune Diseases; Beijing Key Laboratory of Molecular Diagnosis of Dermatoses, Beijing, People's Republic of China
| | - Yufang Liu
- Department of Dermatology and Venerology, The Third Affiliated Hospital of Sun Yat-Sen university, Guangzhou, People's Republic of China
| | - Kai Zhang
- Department of Dermatology and Venerology, Peking University First Hospital, Peking University; National Clinical Research Center for Skin and Immune Diseases; Beijing Key Laboratory of Molecular Diagnosis of Dermatoses, Beijing, People's Republic of China
| | - Yi Zhang
- Department of Dermatology and Venerology, Peking University First Hospital, Peking University; National Clinical Research Center for Skin and Immune Diseases; Beijing Key Laboratory of Molecular Diagnosis of Dermatoses, Beijing, People's Republic of China
| | - Tianyu Liang
- Department of Dermatology and Venerology, Peking University First Hospital, Peking University; National Clinical Research Center for Skin and Immune Diseases; Beijing Key Laboratory of Molecular Diagnosis of Dermatoses, Beijing, People's Republic of China
| | - Jin Shao
- Department of Dermatology and Venerology, Peking University First Hospital, Peking University; National Clinical Research Center for Skin and Immune Diseases; Beijing Key Laboratory of Molecular Diagnosis of Dermatoses, Beijing, People's Republic of China
| | - Xiao Liu
- Department of Dermatology and Venerology, Peking University First Hospital, Peking University; National Clinical Research Center for Skin and Immune Diseases; Beijing Key Laboratory of Molecular Diagnosis of Dermatoses, Beijing, People's Republic of China
| | - Wenling Han
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Peking University Center for Human Disease Genomics, Key Laboratory of Medical Immunology, Ministry of Health, Beijing, People's Republic of China
| | - Ruoyu Li
- Department of Dermatology and Venerology, Peking University First Hospital, Peking University; National Clinical Research Center for Skin and Immune Diseases; Beijing Key Laboratory of Molecular Diagnosis of Dermatoses, Beijing, People's Republic of China
| |
Collapse
|
15
|
Li C, Bai X, Liu X, Zhang Y, Liu L, Zhang L, Xu F, Yang Y, Liu M. Disruption of Epithelial Barrier of Caco-2 Cell Monolayers by Excretory Secretory Products of Trichinella spiralis Might Be Related to Serine Protease. Front Microbiol 2021; 12:634185. [PMID: 33815318 PMCID: PMC8013981 DOI: 10.3389/fmicb.2021.634185] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/19/2021] [Indexed: 11/16/2022] Open
Abstract
The physical barrier is composed of epithelial cells which are joined together through intercellular connections. It serves to prevent pathogenic microorganisms from departing the intestinal lumen to invade the host. The excretory secretory (ES) products of Trichinella spiralis are critical for invasion. However, whether ES products of T. spiralis can act on the intestinal barrier is still unknown. In this study, the role of ES products of T. spiralis muscle larvae (Ts-ML-ES) in host invasion was studied by establishing an in vitro cell monolayers model. Barrier integrity analysis by a transmembrane resistance test and a paracellular permeability assay revealed that the Ts-ML-ES was able to destroy barrier function. It occurred via a reduction in the expression of tight junction (TJ) proteins, which was induced by serine protease. Furthermore, Western bolt analysis indicated that Ts-ML-ES reduced the expression of TJ proteins via the MAPK signaling pathway. Based on these data, we conclude that serine protease are likely the main factors from Ts-ML-ES that affect host intestinal barrier integrity by reducing the expression of TJs via the P38-MAPK signaling pathway. Serine protease in Ts-ML-ES might be a key invasion factor in T. spiralis.
Collapse
Affiliation(s)
- Chengyao Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Xue Bai
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Xiaolei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Yuanyuan Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Lei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Lixiao Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Fengyan Xu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Yong Yang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Mingyuan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
16
|
Tight Junctions as a Key for Pathogens Invasion in Intestinal Epithelial Cells. Int J Mol Sci 2021; 22:ijms22052506. [PMID: 33801524 PMCID: PMC7958858 DOI: 10.3390/ijms22052506] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Tight junctions play a major role in maintaining the integrity and impermeability of the intestinal barrier. As such, they act as an ideal target for pathogens to promote their translocation through the intestinal mucosa and invade their host. Different strategies are used by pathogens, aimed at directly destabilizing the junctional network or modulating the different signaling pathways involved in the modulation of these junctions. After a brief presentation of the organization and modulation of tight junctions, we provide the state of the art of the molecular mechanisms leading to permeability breakdown of the gut barrier as a consequence of tight junctions’ attack by pathogens, including bacteria, viruses, fungi, and parasites.
Collapse
|
17
|
Abstract
In the last decades, Candida albicans has served as the leading causal agent of life-threatening invasive infections with mortality rates approaching 40% despite treatment. Candida albicans (C. albicans) exists in three biological phases: yeast, pseudohyphae, and hyphae. Hyphae, which represent an important phase in the disease process, can cause tissue damage by invading mucosal epithelial cells then leading to blood infection. In this review, we summarized recent results from different fields of fungal cell biology that are instrumental in understanding hyphal growth. This includes research on the differences among C. albicans phases; the regulatory mechanism of hyphal growth, extension, and maintaining cutting-edge polarity; cross regulations of hyphal development and the virulence factors that cause serious infection. With a better understanding of the mechanism on mycelium formation, this review provides a theoretical basis for the identification of targets in candidiasis treatment. It also gives some reference to the study of antifungal drugs.
Collapse
Affiliation(s)
- Hui Chen
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Biao Ren
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Kumamoto CA, Gresnigt MS, Hube B. The gut, the bad and the harmless: Candida albicans as a commensal and opportunistic pathogen in the intestine. Curr Opin Microbiol 2020; 56:7-15. [PMID: 32604030 PMCID: PMC7744392 DOI: 10.1016/j.mib.2020.05.006] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/04/2020] [Accepted: 05/11/2020] [Indexed: 12/26/2022]
Abstract
Candida albicans is a regular member of the intestinal microbiota in the majority of the human population. This underscores C. albicans' adaptation to life in the intestine without inducing competitive interactions with other microbes, or immune responses detrimental to its survival. However, specific conditions such as a dysbalanced microbiome, a suppression of the immune system, and an impaired intestinal barrier can predispose for invasive, mostly nosocomial, C. albicans infections. Colonization of the intestine and translocation through the intestinal barrier are fundamental aspects of the processes preceding life-threatening systemic candidiasis. Insights into C. albicans' commensal lifestyle and translocation can thus help us to understand how patients develop candidiasis, and may provide leads for therapeutic strategies aimed at preventing infection. In this review, we discuss the commensal lifestyle of C. albicans in the intestine, the role of morphology for commensalism, the influence of diet, and the interactions with bacteria of the microbiota.
Collapse
Affiliation(s)
- Carol A Kumamoto
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02111, USA
| | - Mark S Gresnigt
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knoell-Institute, Beutenbergstraße 11a, 07745 Jena, Germany; Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11a 07745, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knoell-Institute, Beutenbergstraße 11a, 07745 Jena, Germany; Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University, 07743 Jena, Germany.
| |
Collapse
|
19
|
Tao Y, Yue M, Lv C, Yun X, Qiao S, Fang Y, Wei Z, Xia Y, Dai Y. Pharmacological activation of ERβ by arctigenin maintains the integrity of intestinal epithelial barrier in inflammatory bowel diseases. FASEB J 2019; 34:3069-3090. [PMID: 31908053 DOI: 10.1096/fj.201901638rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 12/09/2019] [Accepted: 12/17/2019] [Indexed: 12/29/2022]
Abstract
Intestinal epithelial barrier dysfunction is deeply involved in the pathogenesis of inflammatory bowel diseases (IBD). Arctigenin, the main active constituent in Fructus Arctii (a traditional Chinese medicine), has previously been found to attenuate colitis induced by dextran sulfate sodium (DSS) in mice. The present study investigated whether and how arctigenin protects against the disruption of the intestinal epithelial barrier in IBD. Arctigenin maintained the intestinal epithelial barrier function of mice with DSS- and TNBS-induced colitis. In Caco-2 and HT-29 cells, arctigenin lowered the monolayer permeability, increased TEER, reversed the abnormal expression of tight junction proteins, and restored the altered localization of F-actin induced by TNF-α and IL-1β. The specific antagonist PHTPP or shRNA of ERβ largely weakened the protective effect of arctigenin on the epithelial barrier function of Caco-2 and HT-29 cells. Molecular docking demonstrated that arctigenin had high affinity for ERβ mainly through hydrogen bonds as well as hydrophobic effects, and the protective effect of arctigenin on the intestinal barrier function was largely diminished in ERβ-mutated (ARG346 and/or GLU305) Caco-2 cells. Moreover, arctigenin-blocked TNF-α induced increase of the monolayer permeability in Caco-2 and HT-29 cells and the activation of myosin light chain kinase (MLCK)/myosin light chain (MLC) pathway in an ERβ-dependent manner. ERβ deletion in colons of mice with DSS-induced colitis resulted in a significant attenuation of the protective effect of arctigenin on the barrier integrity and colon inflammation. Arctigenin maintained the integrity of the intestinal epithelial barrier under IBD by upregulating the expression of tight junction proteins through the ERβ-MLCK/MLC pathway.
Collapse
Affiliation(s)
- Yu Tao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Mengfan Yue
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Changjun Lv
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xinming Yun
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Simiao Qiao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yulai Fang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhifeng Wei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yufeng Xia
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yue Dai
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
20
|
Farnesol contributes to intestinal epithelial barrier function by enhancing tight junctions via the JAK/STAT3 signaling pathway in differentiated Caco-2 cells. J Bioenerg Biomembr 2019; 51:403-412. [PMID: 31845097 DOI: 10.1007/s10863-019-09817-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/26/2019] [Indexed: 12/15/2022]
Abstract
Candida albicans causes mucosal diseases and secretes farnesol, a quorum-sensing molecule, which plays a vital role in suppressing the yeast-to-mycelia switch. Farnesol can also regulate immune cell function. However, how farnesol interacts with the intestinal epithelium remains unknown. Herein, we identified that farnesol promotes intestinal barrier function, by promoting transepithelial electrical resistance, reducing paracellular flux, inducing the Zonula Occludens-1 Protein (ZO-1) and occludin expression. Moreover, the JAK/STAT3 signaling pathway was activated after farnesol treatment, and inhibition of STAT3 phosphorylation by stattic remarkably suppressed the expression level of ZO-1. Additionally, chromatin immunoprecipitation assay (Chip) revealed that farnesol facilitated the transcriptional activation of STAT3 to significantly enhance the expression of ZO-1. Taken together, our findings demonstrated that farnesol facilitated intestinal epithelial barrier transcriptional regulation via activating JAK/STAT3 signaling. The involved molecules may be potentially targeted for treatment of Candida albicans invasion.
Collapse
|
21
|
Graf K, Last A, Gratz R, Allert S, Linde S, Westermann M, Gröger M, Mosig AS, Gresnigt MS, Hube B. Keeping Candida commensal: how lactobacilli antagonize pathogenicity of Candida albicans in an in vitro gut model. Dis Model Mech 2019; 12:dmm.039719. [PMID: 31413153 PMCID: PMC6765188 DOI: 10.1242/dmm.039719] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 08/02/2019] [Indexed: 12/20/2022] Open
Abstract
The intestine is the primary reservoir of Candida albicans that can cause systemic infections in immunocompromised patients. In this reservoir, the fungus exists as a harmless commensal. However, antibiotic treatment can disturb the bacterial microbiota, facilitating fungal overgrowth and favoring pathogenicity. The current in vitro gut models that are used to study the pathogenesis of C. albicans investigate the state in which C. albicans behaves as a pathogen rather than as a commensal. We present a novel in vitro gut model in which the fungal pathogenicity is reduced to a minimum by increasing the biological complexity. In this model, enterocytes represent the epithelial barrier and goblet cells limit C. albicans adhesion and invasion. Significant protection against C. albicans-induced necrotic damage was achieved by the introduction of a microbiota of antagonistic lactobacilli. We demonstrated a time-, dose- and species-dependent protective effect against C. albicans-induced cytotoxicity. This required bacterial growth, which relied on the presence of host cells, but was not dependent on the competition for adhesion sites. Lactobacillus rhamnosus reduced hyphal elongation, a key virulence attribute. Furthermore, bacterial-driven shedding of hyphae from the epithelial surface, associated with apoptotic epithelial cells, was identified as a main and novel mechanism of damage protection. However, host cell apoptosis was not the driving mechanism behind shedding. Collectively, we established an in vitro gut model that can be used to experimentally dissect commensal-like interactions of C. albicans with a bacterial microbiota and the host epithelial barrier. We also discovered fungal shedding as a novel mechanism by which bacteria contribute to the protection of epithelial surfaces.This article has an associated First Person interview with the joint first authors of the paper.
Collapse
Affiliation(s)
- Katja Graf
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Antonia Last
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Rena Gratz
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Stefanie Allert
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Susanne Linde
- Center for Electron Microscopy Jena University Hospital, Ziegelmühlenweg 1, 07743 Jena, Germany
| | - Martin Westermann
- Center for Electron Microscopy Jena University Hospital, Ziegelmühlenweg 1, 07743 Jena, Germany
| | - Marko Gröger
- Center for Sepsis Control and Care (CSCC), University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Alexander S Mosig
- Center for Sepsis Control and Care (CSCC), University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany.,Institute of Biochemistry II, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Mark S Gresnigt
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany .,Friedrich Schiller University, Fürstengraben 1, 07743 Jena, Germany
| |
Collapse
|
22
|
Candida albicans Elicits Pro-Inflammatory Differential Gene Expression in Intestinal Peyer's Patches. Mycopathologia 2019; 184:461-478. [PMID: 31230200 DOI: 10.1007/s11046-019-00349-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 06/05/2019] [Indexed: 12/17/2022]
Abstract
The details of how gut-associated lymphoid tissues such as Peyer's patches (PPs) in the small intestine play a role in immune surveillance, microbial differentiation and the mucosal barrier protection in response to fungal organisms such as Candida albicans are still unclear. We particularly focus on PPs as they are the immune sensors and inductive sites of the gut that influence inflammation and tolerance. We have previously demonstrated that CD11c+ phagocytes that include dendritic cells and macrophages are located in the sub-epithelial dome within PPs sample C. albicans. To gain insight on how specific cells within PPs sense and respond to the sampling of fungi, we gavaged naïve mice with C. albicans strains ATCC 18804 and SC5314 as well as Saccharomyces cerevisiae. We measured the differential gene expression of sorted CD45+ B220+ B-cells, CD3+ T-cells and CD11c+ DCs within the first 24 h post-gavage using nanostring nCounter® technology. The results reveal that at 24 h, PP phagocytes were the cell type that displayed differential gene expression. These phagocytes were able to sample C. albicans and discriminate between strains. In particular, strain ATCC 18804 upregulated fungal-specific pro-inflammatory genes pertaining to innate and adaptive immune responses. Interestingly, PP CD11c+ phagocytes also differentially expressed genes in response to C. albicans that were important in the protection of the mucosal barrier. These results highlight that the mucosal barrier not only responds to C. albicans, but also aids in the protection of the host.
Collapse
|
23
|
Basmaciyan L, Bon F, Paradis T, Lapaquette P, Dalle F. " Candida Albicans Interactions With The Host: Crossing The Intestinal Epithelial Barrier". Tissue Barriers 2019; 7:1612661. [PMID: 31189436 PMCID: PMC6619947 DOI: 10.1080/21688370.2019.1612661] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 02/08/2023] Open
Abstract
Formerly a commensal organism of the mucosal surfaces of most healthy individuals, Candida albicans is an opportunistic pathogen that causes infections ranging from superficial to the more life-threatening disseminated infections, especially in the ever-growing population of vulnerable patients in the hospital setting. In these situations, the fungus takes advantage of its host following a disturbance in the host defense system and/or the mucosal microbiota. Overwhelming evidence suggests that the gastrointestinal tract is the main source of disseminated C. albicans infections. Major risk factors for disseminated candidiasis include damage to the mucosal intestinal barrier, immune dysfunction, and dysbiosis of the resident microbiota. A better understanding of C. albicans' interaction with the intestinal epithelial barrier will be useful for designing future therapies to avoid systemic candidiasis. In this review, we provide an overview of the current knowledge regarding the mechanisms of pathogenicity that allow the fungus to reach and translocate the gut barrier.
Collapse
Affiliation(s)
- Louise Basmaciyan
- Laboratoire de Parasitologie-Mycologie, Plateforme de Biologie Hospitalo-Universitaire Gérard Mack, Dijon France
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Fabienne Bon
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Tracy Paradis
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Pierre Lapaquette
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Frédéric Dalle
- Laboratoire de Parasitologie-Mycologie, Plateforme de Biologie Hospitalo-Universitaire Gérard Mack, Dijon France
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| |
Collapse
|
24
|
Allert S, Förster TM, Svensson CM, Richardson JP, Pawlik T, Hebecker B, Rudolphi S, Juraschitz M, Schaller M, Blagojevic M, Morschhäuser J, Figge MT, Jacobsen ID, Naglik JR, Kasper L, Mogavero S, Hube B. Candida albicans-Induced Epithelial Damage Mediates Translocation through Intestinal Barriers. mBio 2018; 9:e00915-18. [PMID: 29871918 PMCID: PMC5989070 DOI: 10.1128/mbio.00915-18] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 05/03/2018] [Indexed: 01/31/2023] Open
Abstract
Life-threatening systemic infections often occur due to the translocation of pathogens across the gut barrier and into the bloodstream. While the microbial and host mechanisms permitting bacterial gut translocation are well characterized, these mechanisms are still unclear for fungal pathogens such as Candida albicans, a leading cause of nosocomial fungal bloodstream infections. In this study, we dissected the cellular mechanisms of translocation of C. albicans across intestinal epithelia in vitro and identified fungal genes associated with this process. We show that fungal translocation is a dynamic process initiated by invasion and followed by cellular damage and loss of epithelial integrity. A screen of >2,000 C. albicans deletion mutants identified genes required for cellular damage of and translocation across enterocytes. Correlation analysis suggests that hypha formation, barrier damage above a minimum threshold level, and a decreased epithelial integrity are required for efficient fungal translocation. Translocation occurs predominantly via a transcellular route, which is associated with fungus-induced necrotic epithelial damage, but not apoptotic cell death. The cytolytic peptide toxin of C. albicans, candidalysin, was found to be essential for damage of enterocytes and was a key factor in subsequent fungal translocation, suggesting that transcellular translocation of C. albicans through intestinal layers is mediated by candidalysin. However, fungal invasion and low-level translocation can also occur via non-transcellular routes in a candidalysin-independent manner. This is the first study showing translocation of a human-pathogenic fungus across the intestinal barrier being mediated by a peptide toxin.IMPORTANCECandida albicans, usually a harmless fungus colonizing human mucosae, can cause lethal bloodstream infections when it manages to translocate across the intestinal epithelium. This can result from antibiotic treatment, immune dysfunction, or intestinal damage (e.g., during surgery). However, fungal processes may also contribute. In this study, we investigated the translocation process of C. albicans using in vitro cell culture models. Translocation occurs as a stepwise process starting with invasion, followed by epithelial damage and loss of epithelial integrity. The ability to secrete candidalysin, a peptide toxin deriving from the hyphal protein Ece1, is key: C. albicans hyphae, secreting candidalysin, take advantage of a necrotic weakened epithelium to translocate through the intestinal layer.
Collapse
Affiliation(s)
- Stefanie Allert
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
| | - Toni M Förster
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
| | | | - Jonathan P Richardson
- Mucosal & Salivary Biology Division, Dental Institute, King's College London, London, United Kingdom
| | - Tony Pawlik
- Research Group Microbial Immunology, Hans-Knöll-Institute, Jena, Germany
| | - Betty Hebecker
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
- Research Group Microbial Immunology, Hans-Knöll-Institute, Jena, Germany
- Aberdeen Fungal Group, MRC Centre for Medical Mycology, University of Aberdeen, Aberdeen, United Kingdom
| | - Sven Rudolphi
- Research Group Microbial Immunology, Hans-Knöll-Institute, Jena, Germany
| | - Marc Juraschitz
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
| | - Martin Schaller
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Mariana Blagojevic
- Mucosal & Salivary Biology Division, Dental Institute, King's College London, London, United Kingdom
| | - Joachim Morschhäuser
- Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Marc Thilo Figge
- Research Group Applied Systems Biology, Hans-Knöll-Institute, Jena, Germany
- Faculty of Biological Sciences, Friedrich-Schiller-University Jena, Jena, Germany
| | - Ilse D Jacobsen
- Research Group Microbial Immunology, Hans-Knöll-Institute, Jena, Germany
- Institute of Microbiology, Friedrich-Schiller-University Jena, Jena, Germany
| | - Julian R Naglik
- Mucosal & Salivary Biology Division, Dental Institute, King's College London, London, United Kingdom
| | - Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
| | - Selene Mogavero
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
- Institute of Microbiology, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
25
|
Binding of Candida albicans to Human CEACAM1 and CEACAM6 Modulates the Inflammatory Response of Intestinal Epithelial Cells. mBio 2017; 8:mBio.02142-16. [PMID: 28292985 PMCID: PMC5350469 DOI: 10.1128/mbio.02142-16] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Candida albicans colonizes human mucosa, including the gastrointestinal tract, as a commensal. In immunocompromised patients, C. albicans can breach the intestinal epithelial barrier and cause fatal invasive infections. Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1; CD66a), CEACAM5 (CEA), and CEACAM6 (CD66c) are immunomodulatory receptors expressed on human mucosa and are recruited by bacterial and viral pathogens. Here we show for the first time that a fungal pathogen (i.e., C. albicans) also binds directly to the extracellular domain of human CEACAM1, CEACAM3, CEACAM5, and CEACAM6. Binding was specific for human CEACAMs and mediated by the N-terminal IgV-like domain. In enterocytic C2BBe1 cells, C. albicans caused a transient tyrosine phosphorylation of CEACAM1 and induced higher expression of membrane-bound CEACAM1 and soluble CEACAM6. Lack of the CEACAM1 receptor after short hairpin RNA (shRNA) knockdown abolished CXCL8 (interleukin-8) secretion by C2BBe1 cells in response to C. albicans In CEACAM1-competent cells, the addition of recombinant soluble CEACAM6 reduced the C. albicans-induced CXCL8 secretion.IMPORTANCE The present study demonstrates for the first time that fungal pathogens can be recognized by at least four members of the immunomodulatory CEACAM receptor family: CEACAM1, -3, -5, and -6. Three of the four receptors (i.e., CEACAM1, -5, and -6) are expressed in mucosal cells of the intestinal tract, where they are implicated in immunomodulation and control of tissue homeostasis. Importantly, the interaction of the major fungal pathogen in humans Candida albicans with CEACAM1 and CEACAM6 resulted in an altered epithelial immune response. With respect to the broad impact of CEACAM receptors on various aspects of the innate and the adaptive immune responses, in particular epithelial, neutrophil, and T cell behavior, understanding the role of CEACAMs in the host response to fungal pathogens might help to improve management of superficial and systemic fungal infections.
Collapse
|
26
|
Guthke R, Gerber S, Conrad T, Vlaic S, Durmuş S, Çakır T, Sevilgen FE, Shelest E, Linde J. Data-based Reconstruction of Gene Regulatory Networks of Fungal Pathogens. Front Microbiol 2016; 7:570. [PMID: 27148247 PMCID: PMC4840211 DOI: 10.3389/fmicb.2016.00570] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 04/05/2016] [Indexed: 12/17/2022] Open
Abstract
In the emerging field of systems biology of fungal infection, one of the central roles belongs to the modeling of gene regulatory networks (GRNs). Utilizing omics-data, GRNs can be predicted by mathematical modeling. Here, we review current advances of data-based reconstruction of both small-scale and large-scale GRNs for human pathogenic fungi. The advantage of large-scale genome-wide modeling is the possibility to predict central (hub) genes and thereby indicate potential biomarkers and drug targets. In contrast, small-scale GRN models provide hypotheses on the mode of gene regulatory interactions, which have to be validated experimentally. Due to the lack of sufficient quantity and quality of both experimental data and prior knowledge about regulator–target gene relations, the genome-wide modeling still remains problematic for fungal pathogens. While a first genome-wide GRN model has already been published for Candida albicans, the feasibility of such modeling for Aspergillus fumigatus is evaluated in the present article. Based on this evaluation, opinions are drawn on future directions of GRN modeling of fungal pathogens. The crucial point of genome-wide GRN modeling is the experimental evidence, both used for inferring the networks (omics ‘first-hand’ data as well as literature data used as prior knowledge) and for validation and evaluation of the inferred network models.
Collapse
Affiliation(s)
- Reinhard Guthke
- Research Group Systems Biology and Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute Jena, Germany
| | - Silvia Gerber
- Research Group Systems Biology and Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute Jena, Germany
| | - Theresia Conrad
- Research Group Systems Biology and Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute Jena, Germany
| | - Sebastian Vlaic
- Research Group Systems Biology and Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute Jena, Germany
| | - Saliha Durmuş
- Computational Systems Biology Group, Department of Bioengineering, Gebze Technical University Kocaeli, Turkey
| | - Tunahan Çakır
- Computational Systems Biology Group, Department of Bioengineering, Gebze Technical University Kocaeli, Turkey
| | - F E Sevilgen
- Department of Computer Engineering, Gebze Technical University Kocaeli, Turkey
| | - Ekaterina Shelest
- Research Group Systems Biology and Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute Jena, Germany
| | - Jörg Linde
- Research Group Systems Biology and Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute Jena, Germany
| |
Collapse
|