1
|
Xu C, Yang L, Cheng T, Wang Z, Liu C, Shao J. Sodium Houttuyfonate Ameliorates DSS-induced Colitis Aggravated by Candida albicans through Dectin-1/NF-κB/miR-32-5p/NFKBIZ Axis Based on Intestinal microRNA Profiling. Inflammation 2024:10.1007/s10753-024-02091-6. [PMID: 38963571 DOI: 10.1007/s10753-024-02091-6] [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: 05/14/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Our previous research indicated that Sodium houttuyfonate (SH) can effectively ameliorate dextran sulfate sodium (DSS)-induced colitis exacerbated by Candida albicans. However, the underlying protective mechanism of SH remains unclear. Therefore, in this study, a mice colitis model was infected with C. albicans, and the total colonic miRNAs were assessed. Furthermore, the differentially expressed miRNAs were enriched, clustered, and analyzed. Moreover, based on the dual luciferase analysis of NFKBIZ modulation by miR-32-5p, the in vitro and in vivo therapeutic effects of SH on inflammatory response, fungal burden, oxidative stress, and apoptosis were assessed at transcriptional and translational levels in the presence of agonist and antagonist. A total of 1157 miRNAs were identified, 84 of which were differentially expressed. Furthermore, qRT-PCR validated that SH treatment improved 17 differentially expressed miRNAs with > fourfold upregulation or > sixfold downregulation. Similar to most differentially altered miRNA, C. albicans significantly increased Dectin-1, NF-κB, TNF-α, IL-1β, IL-17A, and decreased miR-32-5p which negatively targeted NFKBIZ. In addition, SH treatment reduced inflammatory response and fungal burden in a colitis model with C. albicans infection. Further analyses indicated that in C. albicans infected Caco2 cells, SH inhibited fungal growth, oxidative stress, and apoptosis by increasing Dectin-1, NF-κB, NFKBIZ, TNF-α, IL-1β, IL-17A, and decreasing miR-32-5p. Therefore, SH can ameliorate the severity of colitis aggravated by C. albicans via the Dectin-1/NF-κB/miR-32-5p/NFKBIZ axis.
Collapse
Affiliation(s)
- Chen Xu
- Laboratory of Anti-Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei, 230012, Anhui, P. R. China
| | - Liu Yang
- Laboratory of Anti-Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei, 230012, Anhui, P. R. China
| | - Ting Cheng
- Laboratory of Anti-Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei, 230012, Anhui, P. R. China
| | - Zixu Wang
- Laboratory of Anti-Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei, 230012, Anhui, P. R. China
| | - Chengcheng Liu
- Laboratory of Anti-Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei, 230012, Anhui, P. R. China
| | - Jing Shao
- Laboratory of Anti-Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 433 Room, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei, 230012, Anhui, P. R. China.
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei, 230012, Anhui, P. R. China.
| |
Collapse
|
2
|
Graham AS, Ben-Azu B, Tremblay MÈ, Torre P, Senekal M, Laughton B, van der Kouwe A, Jankiewicz M, Kaba M, Holmes MJ. A review of the auditory-gut-brain axis. Front Neurosci 2023; 17:1183694. [PMID: 37600010 PMCID: PMC10435389 DOI: 10.3389/fnins.2023.1183694] [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: 03/10/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Hearing loss places a substantial burden on medical resources across the world and impacts quality of life for those affected. Further, it can occur peripherally and/or centrally. With many possible causes of hearing loss, there is scope for investigating the underlying mechanisms involved. Various signaling pathways connecting gut microbes and the brain (the gut-brain axis) have been identified and well established in a variety of diseases and disorders. However, the role of these pathways in providing links to other parts of the body has not been explored in much depth. Therefore, the aim of this review is to explore potential underlying mechanisms that connect the auditory system to the gut-brain axis. Using select keywords in PubMed, and additional hand-searching in google scholar, relevant studies were identified. In this review we summarize the key players in the auditory-gut-brain axis under four subheadings: anatomical, extracellular, immune and dietary. Firstly, we identify important anatomical structures in the auditory-gut-brain axis, particularly highlighting a direct connection provided by the vagus nerve. Leading on from this we discuss several extracellular signaling pathways which might connect the ear, gut and brain. A link is established between inflammatory responses in the ear and gut microbiome-altering interventions, highlighting a contribution of the immune system. Finally, we discuss the contribution of diet to the auditory-gut-brain axis. Based on the reviewed literature, we propose numerous possible key players connecting the auditory system to the gut-brain axis. In the future, a more thorough investigation of these key players in animal models and human research may provide insight and assist in developing effective interventions for treating hearing loss.
Collapse
Affiliation(s)
- Amy S. Graham
- Imaging Sciences, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Human Biology, Division of Biomedical Engineering, University of Cape Town, Cape Town, South Africa
| | - Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Département de Médecine Moléculaire, Université Laval, Québec City, QC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Quebec City, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
| | - Peter Torre
- School of Speech, Language, and Hearing Sciences, San Diego State University, San Diego, CA, United States
| | - Marjanne Senekal
- Department of Human Biology, Division of Physiological Sciences, University of Cape Town, Cape Town, South Africa
| | - Barbara Laughton
- Family Clinical Research Unit, Department of Pediatrics and Child Health, Stellenbosch University, Cape Town, South Africa
| | - Andre van der Kouwe
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
- Department of Radiology, Harvard Medical School, Boston, MA, United States
| | - Marcin Jankiewicz
- Imaging Sciences, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Human Biology, Division of Biomedical Engineering, University of Cape Town, Cape Town, South Africa
| | - Mamadou Kaba
- Department of Pathology, Division of Medical Microbiology, University of Cape Town, Cape Town, South Africa
| | - Martha J. Holmes
- Imaging Sciences, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Human Biology, Division of Biomedical Engineering, University of Cape Town, Cape Town, South Africa
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- ImageTech, Simon Fraser University, Surrey, BC, Canada
| |
Collapse
|
3
|
D'Ambrosio F. Oral Mycobiome and COVID-19. Microorganisms 2023; 11:microorganisms11040982. [PMID: 37110405 PMCID: PMC10142434 DOI: 10.3390/microorganisms11040982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
The most common signs and symptoms of COVID-19 include fever, cough, dyspnea, conjunctivitis, diarrhea, and olfactory and gustatory disturbances [...].
Collapse
Affiliation(s)
- Francesco D'Ambrosio
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84084 Salerno, Italy
| |
Collapse
|
4
|
Chen Q, Fan Y, Zhang B, Yan C, Chen Z, Wang L, Hu Y, Huang Q, Su J, Ren J, Xu H. Specific fungi associated with response to capsulized fecal microbiota transplantation in patients with active ulcerative colitis. Front Cell Infect Microbiol 2023; 12:1086885. [PMID: 36683707 PMCID: PMC9849685 DOI: 10.3389/fcimb.2022.1086885] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/08/2022] [Indexed: 01/06/2023] Open
Abstract
Objective Fecal microbiota transplantation (FMT) is a novel microbial treatment for patients with ulcerative colitis (UC). In this study, we performed a clinical trial of capsulized FMT in UC patients to determine the association between the gut fungal community and capsulized FMT outcomes. Design This study recruited patients with active UC (N = 22) and healthy individuals (donor, N = 9) according to the criteria. The patients received capsulized FMT three times a week. Patient stool samples were collected before (week 0) and after FMT follow-up visits at weeks 1, 4, and 12. Fungal communities were analysed using shotgun metagenomic sequencing. Results According to metagenomic analysis, fungal community evenness index was greater in samples collected from patients, and the overall fungal community was clustered among the samples collected from donors. The dominant fungi in fecal samples collected from donors and patients were Ascomycota and Basidiomycota. However, capsulized FMT ameliorated microbial fungal diversity and altered fungal composition, based on metagenomic analysis of fecal samples collected before and during follow-up visits after capsulized FMT. Fungal diversity decreased in samples collected from patients who achieved remission after capsulized FMT, similar to samples collected from donors. Patients achieving remission after capsulized FMT had specific enrichment of Kazachstania naganishii, Pyricularia grisea, Lachancea thermotolerans, and Schizosaccharomyces pombe compared with patients who did not achieve remission. In addition, the relative abundance of P. grisea was higher in remission fecal samples during the follow-up visit. Meanwhile, decreased levels of pathobionts, such as Candida and Debaryomyces hansenii, were associated with remission in patients receiving capsulized FMT. Conclusion In the metagenomic analysis of fecal samples from donors and patients with UC receiving capsulized FMT, shifts in gut fungal diversity and composition were associated with capsulized FMT and validated in patients with active UC. We also identified the specific fungi associated with the induction of remission. ClinicalTrails.gov (NCT03426683).
Collapse
Affiliation(s)
- Qiongyun Chen
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China,Institute for Microbial Ecology, School of Medicine, Xiamen University, Xiamen, China
| | - Yanyun Fan
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Bangzhou Zhang
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China,Institute for Microbial Ecology, School of Medicine, Xiamen University, Xiamen, China
| | - Changsheng Yan
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China,Institute for Microbial Ecology, School of Medicine, Xiamen University, Xiamen, China
| | - Zhangran Chen
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China,Institute for Microbial Ecology, School of Medicine, Xiamen University, Xiamen, China
| | - Lin Wang
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yiqun Hu
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Qingwen Huang
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jingling Su
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jianlin Ren
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China,Institute for Microbial Ecology, School of Medicine, Xiamen University, Xiamen, China,Xiamen Key Laboratory of Intestinal Microbiome and Human Health, Zhongshan Hospital of Xiamen University, Xiamen, China,Department of Digestive Disease, School of Medicine, Xiamen University, Xiamen, China,*Correspondence: Jianlin Ren, ; Hongzhi Xu,
| | - Hongzhi Xu
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China,Institute for Microbial Ecology, School of Medicine, Xiamen University, Xiamen, China,Xiamen Key Laboratory of Intestinal Microbiome and Human Health, Zhongshan Hospital of Xiamen University, Xiamen, China,Department of Digestive Disease, School of Medicine, Xiamen University, Xiamen, China,*Correspondence: Jianlin Ren, ; Hongzhi Xu,
| |
Collapse
|
5
|
Dong Y, Yao J, Deng Q, Li X, He Y, Ren X, Zheng Y, Song R, Zhong X, Ma J, Shan D, Lv F, Wang X, Yuan R, She G. Relationship between gut microbiota and rheumatoid arthritis: A bibliometric analysis. Front Immunol 2023; 14:1131933. [PMID: 36936921 PMCID: PMC10015446 DOI: 10.3389/fimmu.2023.1131933] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 02/14/2023] [Indexed: 03/05/2023] Open
Abstract
Introduction Rheumatoid arthritis (RA) is a multifactorial autoimmune disease. Recently, growing evidence demonstrates that gut microbiota (GM) plays an important role in RA. But so far, no bibliometric studies pertaining to GM in RA have ever been published. This study attempts to depict the knowledge framework in this field from a holistic and systematic perspective based on the bibliometric analysis. Methods Literature related to the involvement of GM in RA was searched and picked from the Web of Science Core Collection (WOSCC) database. The annual output, cooperation, hotspots, research status and development trend of this field were analyzed by bibliometric software (VOSviewer and Bibliometricx). Results 255 original research articles and 204 reviews were included in the analysis. The articles in this field that can be retrieved in WOSCC were first published in 2004 and increased year by year since then. 2013 is a growth explosion point. China and the United States are the countries with the most contributions, and Harvard University is the affiliation with the most output. Frontiers in Immunology (total citations = 603) is the journal with the most publications and the fastest growth rate. eLife is the journal with the most citations (total citations = 1248). Scher, Jose U. and Taneja, Veena are the most productive and cited authors. The research in this field is mainly distributed in the evidence, mechanism and practical application of GM participating in RA through the analysis of keywords and documents. There is sufficient evidence to prove the close relationship between GM and RA, which lays the foundation for this field. This extended two colorful and tender branches of mechanism research and application exploration, which have made some achievements but still have broad exploration space. Recently, the keywords "metabolites", "metabolomics", "acid", "b cells", "balance", "treg cells", "probiotic supplementation" appeared most frequently, which tells us that research on the mechanism of GM participating in RA and exploration of its application are the hotspots in recent years. Discussion Taken together, these results provide a data-based and objective introduction to the GM participating in RA, giving readers a valuable reference to help guide future research.
Collapse
Affiliation(s)
- Ying Dong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jianling Yao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Qingyue Deng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xianxian Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yingyu He
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xueyang Ren
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuan Zheng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ruolan Song
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiangjian Zhong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jiamu Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Dongjie Shan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Fang Lv
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiuhuan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Ruijuan Yuan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Ruijuan Yuan, ; Gaimei She,
| | - Gaimei She
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Ruijuan Yuan, ; Gaimei She,
| |
Collapse
|
6
|
Wang M, Zhang Y, Li C, Chang W, Zhang L. The relationship between gut microbiota and COVID-19 progression: new insights into immunopathogenesis and treatment. Front Immunol 2023; 14:1180336. [PMID: 37205106 PMCID: PMC10185909 DOI: 10.3389/fimmu.2023.1180336] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/17/2023] [Indexed: 05/21/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has posed a global health crisis. Increasing evidence underlines the key role of competent immune responses in resisting SARS-CoV-2 infection and manifests the disastrous consequence of host immune dysregulation. Elucidating the mechanisms responsible for deregulated host immunity in COVID-19 may provide a theoretical basis for further research on new treatment modalities. Gut microbiota comprises trillions of microorganisms colonizing the human gastrointestinal tract and has a vital role in immune homeostasis and the gut-lung crosstalk. Particularly, SARS-CoV-2 infection can lead to the disruption of gut microbiota equilibrium, a condition called gut dysbiosis. Due to its regulatory effect on host immunity, gut microbiota has recently received considerable attention in the field of SARS-CoV-2 immunopathology. Imbalanced gut microbiota can fuel COVID-19 progression through production of bioactive metabolites, intestinal metabolism, enhancement of the cytokine storm, exaggeration of inflammation, regulation of adaptive immunity and other aspects. In this review, we provide an overview of the alterations in gut microbiota in COVID-19 patients, and their effects on individuals' susceptibility to viral infection and COVID-19 progression. Moreover, we summarize currently available data on the critical role of the bidirectional regulation between intestinal microbes and host immunity in SARS-CoV-2-induced pathology, and highlight the immunomodulatory mechanisms of gut microbiota contributing to COVID-19 pathogenesis. In addition, we discuss the therapeutic benefits and future perspectives of microbiota-targeted interventions including faecal microbiota transplantation (FMT), bacteriotherapy and traditional Chinese medicine (TCM) in COVID-19 treatment.
Collapse
Affiliation(s)
- Man Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
- *Correspondence: Man Wang, ; Chunmei Li,
| | - Yuan Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Chunmei Li
- Department of Radiology, Qingdao Municipal Hospital, Qingdao, China
- *Correspondence: Man Wang, ; Chunmei Li,
| | - Wenguang Chang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Lei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
7
|
Mao X, Ma J, Jiao C, Tang N, Zhao X, Wang D, Zhang Y, Ye Z, Xu C, Jiang J, Wu S, Cui X, Zhang H, Qiu X. Faecalibacterium prausnitzii Attenuates DSS-Induced Colitis by Inhibiting the Colonization and Pathogenicity of Candida albicans. Mol Nutr Food Res 2021; 65:e2100433. [PMID: 34558816 DOI: 10.1002/mnfr.202100433] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/12/2021] [Indexed: 11/12/2022]
Abstract
SCOPE Intestinal commensal microbiota interactions play critical roles in the inflammatory bowel disease (IBD) development. Candida albicans (CA) can aggravate intestinal inflammation; however, whether Faecalibacterium prausnitzii (FP) can antagonize CA is unknown. METHODS AND RESULTS CA are co-cultured with bacteria (FP and Escherichia coli (EC)), bacterial supernatant, and bacterial medium, respectively. Then, the CA hyphae-specific genes' expression and CA cells' morphology are investigated. The Nod-like receptor pyrin-containing protein 6 (NLRP6) inflammasome, inflammatory cytokines, and antimicrobial peptides (AMPs) production are evaluated in intestinal epithelial cells pre-treated with bacteria, bacterial med, and bacterial supernatant and exposed without or with CA. Both bacteria significantly prohibit CA numbers, while only FP and FP supernatant prohibit the transformation and virulence factors (extracellular phospholipase, secreted aspartyl proteinase, and hemolysin) secretion of CA in a co-culture system compared with media controls. Further, FP and FP supernatant promote the production of the NLRP6 inflammasome, interleukin (IL)-1β, IL-18, and antibacterial peptides (β-defensin (BD)-2 and BD-3) and inhibit in vitro and in vivo CA growth and pathogenicity, and alleviate DSS-colitis in mice, while EC do not show the similar effect. CONCLUSION FP improve intestinal inflammation by inhibiting CA reproduction, colonization, and pathogenicity and inducing AMP secretion in the gut. This study uncovers new relationships between intestinal microbes and fungi in IBD patients.
Collapse
Affiliation(s)
- Xiaqiong Mao
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingjing Ma
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chunhua Jiao
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Nana Tang
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaojing Zhao
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Di Wang
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Zhang
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ziping Ye
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chenjing Xu
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingyue Jiang
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shasha Wu
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiufang Cui
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongjie Zhang
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyun Qiu
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
8
|
Zuo T, Wu X, Wen W, Lan P. Gut Microbiome Alterations in COVID-19. GENOMICS PROTEOMICS & BIOINFORMATICS 2021; 19:679-688. [PMID: 34560321 PMCID: PMC8478109 DOI: 10.1016/j.gpb.2021.09.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 12/20/2022]
Abstract
Since the outset of the coronavirus disease 2019 (COVID-19) pandemic, the gut microbiome in COVID-19 has garnered substantial interest, given its significant roles in human health and pathophysiology. Accumulating evidence is unveiling that the gut microbiome is broadly altered in COVID-19, including the bacterial microbiome, mycobiome, and virome. Overall, the gut microbial ecological network is significantly weakened and becomes sparse in patients with COVID-19, together with a decrease in gut microbiome diversity. Beyond the existence of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), the gut microbiome of patients with COVID-19 is also characterized by enrichment of opportunistic bacteria, fungi, and eukaryotic viruses, which are also associated with disease severity and presentation. Meanwhile, a multitude of symbiotic bacteria and bacteriophages are decreased in abundance in patients with COVID-19. Such gut microbiome features persist in a significant subset of patients with COVID-19 even after disease resolution, coinciding with ‘long COVID’ (also known as post-acute sequelae of COVID-19). The broadly-altered gut microbiome is largely a consequence of SARS-CoV-2infection and its downstream detrimental effects on the systemic host immunity and the gut milieu. The impaired host immunity and distorted gut microbial ecology, particularly loss of low-abundance beneficial bacteria and blooms of opportunistic fungi including Candida, may hinder the reassembly of the gut microbiome post COVID-19. Future investigation is necessary to fully understand the role of the gut microbiome in host immunity against SARS-CoV-2 infection, as well as the long-term effect of COVID-19 on the gut microbiome in relation to the host health after the pandemic.
Collapse
Affiliation(s)
- Tao Zuo
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China; Center for Fecal Microbiota Transplantation Research, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China.
| | - Xiaojian Wu
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China; Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China.
| | - Weiping Wen
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510080, China; Guangzhou Key Laboratory of Otorhinolaryngology, Guangzhou 510080, China; Department of Otorhinolaryngology, Head and Neck Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China.
| | - Ping Lan
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China; Center for Fecal Microbiota Transplantation Research, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China; Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China.
| |
Collapse
|
9
|
Mining the capacity of human-associated microorganisms to trigger rheumatoid arthritis-A systematic immunoinformatics analysis of T cell epitopes. PLoS One 2021; 16:e0253918. [PMID: 34185818 PMCID: PMC8241107 DOI: 10.1371/journal.pone.0253918] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/15/2021] [Indexed: 12/14/2022] Open
Abstract
Autoimmune diseases, often triggered by infection, affect ~5% of the worldwide population. Rheumatoid Arthritis (RA)–a painful condition characterized by the chronic inflammation of joints—comprises up to 20% of known autoimmune pathologies, with the tendency of increasing prevalence. Molecular mimicry is recognized as the leading mechanism underlying infection-mediated autoimmunity, which assumes sequence similarity between microbial and self-peptides driving the activation of autoreactive lymphocytes. T lymphocytes are leading immune cells in the RA-development. Therefore, deeper understanding of the capacity of microorganisms (both pathogens and commensals) to trigger autoreactive T cells is needed, calling for more systematic approaches. In the present study, we address this problem through a comprehensive immunoinformatics analysis of experimentally determined RA-related T cell epitopes against the proteomes of Bacteria, Fungi, and Viruses, to identify the scope of organisms providing homologous antigenic peptide determinants. By this, initial homology screening was complemented with de novo T cell epitope prediction and another round of homology search, to enable: i) the confirmation of homologous microbial peptides as T cell epitopes based on the predicted binding affinity to RA-related HLA polymorphisms; ii) sequence similarity inference for top de novo T cell epitope predictions to the RA-related autoantigens to reveal the robustness of RA-triggering capacity for identified (micro/myco)organisms. Our study reveals a much larger repertoire of candidate RA-triggering organisms, than previously recognized, providing insights into the underestimated role of Fungi in autoimmunity and the possibility of a more direct involvement of bacterial commensals in RA-pathology. Finally, our study pinpoints Endoplasmic reticulum chaperone BiP as the most potent (most likely mimicked) RA-related autoantigen, opening an avenue for identifying the most potent autoantigens in a variety of different autoimmune pathologies, with possible implications in the design of next-generation therapeutics aiming to induce self-tolerance by affecting highly reactive autoantigens.
Collapse
|
10
|
García-Gamboa R, Kirchmayr MR, Gradilla-Hernández MS, Pérez-Brocal V, Moya A, González-Avila M. The intestinal mycobiota and its relationship with overweight, obesity and nutritional aspects. J Hum Nutr Diet 2021; 34:645-655. [PMID: 33586805 DOI: 10.1111/jhn.12864] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND The fungal community of the gastrointestinal tract has recently become of interest, and knowledge of its relationship with the development of obesity is scarce. The present study aimed to evaluate the cultivable fungal fraction from the microbiota and to analyze its relationship with obesity. METHODS Samples were taken from 99 participants with normal weight, overweight and obesity (n = 31, 34 and 34, respectively) and were cultivated in selective medium, and the cultivable yeasts were identified by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Anthropometric and biochemical measures were also evaluated. RESULTS Eutrophic, overweight and obese groups presented concentrations of 1.6, 2.16 and 2.19 log10 colony-forming units g-1 yeast, respectively. Ascomycota and Basidiomycota were the two identified phyla. At the genus level, Candida spp. showed a relatively high prevalence, and 10 different species were detected: Candida glabrata, Candida orthopsilosis, Candida lambica, Candida kefyr, Candida albicans, Candida krusei, Candida valida, Candida parapsilosis, Candida utilis and Candida humilis (with relative abundances of 71.72%, 5.05%, 21.21%, 6.06%, 29.29%, 27.27%, 8.08%, 16.16%, 1.01% and 2.02%, respectively). CONCLUSIONS The obese group presented a higher prevalence of Candida albicans. Furthermore, Candida albicans, Candida kefyr and Rhodotorula mucilaginosa showed a high positive correlation with obesity, weight gain and fat mass and showed a negative correlation with high-density lipoprotein and lean mass, parameters related to weight loss.
Collapse
Affiliation(s)
- Ricardo García-Gamboa
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, Guadalajara, Mexico
| | - Manuel R Kirchmayr
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, Guadalajara, Mexico
| | | | - Vicente Pérez-Brocal
- Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), València, Spain.,Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Andrés Moya
- Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), València, Spain.,Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Integrative Systems Biology Institute (I2SysBio) Universitat de València and Consejo Superior de Investigaciones Científicas (CSIC), València, Spain
| | - Marisela González-Avila
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, Guadalajara, Mexico
| |
Collapse
|
11
|
Mahapatro M, Erkert L, Becker C. Cytokine-Mediated Crosstalk between Immune Cells and Epithelial Cells in the Gut. Cells 2021; 10:cells10010111. [PMID: 33435303 PMCID: PMC7827439 DOI: 10.3390/cells10010111] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/28/2020] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Cytokines are small proteins that are secreted by a vast majority of cell types in the gut. They not only establish cell-to-cell interactions and facilitate cellular signaling, but also regulate both innate and adaptive immune responses, thereby playing a central role in genetic, inflammatory, and infectious diseases of the gut. Both, immune cells and gut epithelial cells, play important roles in intestinal disease development. The epithelium is located in between the mucosal immune system and the gut microbiome. It not only establishes an efficient barrier against gut microbes, but it also signals information from the gut lumen and its composition to the immune cell compartment. Communication across the epithelial cell layer also occurs in the other direction. Intestinal epithelial cells respond to immune cell cytokines and their response influences and shapes the microbial community within the gut lumen. Thus, the epithelium should be seen as a translator or a moderator between the microbiota and the mucosal immune system. Proper communication across the epithelium seems to be a key to gut homeostasis. Indeed, current genome-wide association studies for intestinal disorders have identified several disease susceptibility loci, which map cytokine signatures and their related signaling genes. A thorough understanding of this tightly regulated cytokine signaling network is crucial. The main objective of this review was to shed light on how cytokines can orchestrate epithelial functions such as proliferation, cell death, permeability, microbe interaction, and barrier maintenance, thereby safeguarding host health. In addition, cytokine-mediated therapy for inflammation and cancer are discussed.
Collapse
|
12
|
Smit B, Kuballa A, Coulson S, Katouli M. Interaction of Candida albicans with human gut epithelium in the presence of Live Biotherapeutic Products (LBPs). MICROBIOLOGY AUSTRALIA 2021. [DOI: 10.1071/ma21035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Candida albicans is a semi-ubiquitous pathobiont that is known to significantly impact human health and wellbeing, causing a significant financial strain on the medical system. Due to increasing antifungal resistance, there is a growing need for novel fungal therapeutics to treat diseases caused by this fungus. The development and use of Live Biotherapeutic Products (LBPs) is an innovative and novel approach to potentially treating Candidiasis and other comorbidities associated with C. albicans infection. To evaluate their anti-pathogenic efficacy, it is necessary to understand the underlying mechanisms involved, via the use of biomimetic cell models. In this study, six LBPs were chosen to investigate their competitive inhibitory effect against C. albicans using a co-culture of Caco-2 cells and mucous-secreting HT29-MTX cells to mimic human gut epithelium. The LBP strains were supplied by Servatus Biopharmaceuticals and identified as SVT 01D1, SVT 04P1, SVT 05P2, SVT 06B1, SVT 07R1 and SVT 08Z1. Five out of the six LBPs showed a significant reduction in the adhesion of C. albicans and all six LBPs reduced C. albicans invasion in the co-culture cells to varying degrees. There was no significant difference between co-inoculation of C. albicans with the LBPs or pre-inoculation of LBPs before the addition of C. albicans. The potential of these LBPs as novel anti-fungal therapeutics for the treatment of C. albicans diseases can be further documented in clinical trials.
Collapse
|
13
|
Xu S, Wang X, Nageen Y, Pecoraro L. Analysis of gut-associated fungi from Chinese mitten crab Eriocheir sinensis. ALL LIFE 2021. [DOI: 10.1080/26895293.2021.1939171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Shihan Xu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, People’s Republic of China
| | - Xiao Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, People’s Republic of China
| | - Yumna Nageen
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, People’s Republic of China
| | - Lorenzo Pecoraro
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, People’s Republic of China
| |
Collapse
|
14
|
He ZJ, Liang YX, Cai LY. Advances in the Interaction between Intestinal Microbiota and COVID-19. EXPLORATORY RESEARCH AND HYPOTHESIS IN MEDICINE 2020; 000:1-8. [DOI: 10.14218/erhm.2020.00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
15
|
Zuo T, Zhan H, Zhang F, Liu Q, Tso EYK, Lui GCY, Chen N, Li A, Lu W, Chan FKL, Chan PKS, Ng SC. Alterations in Fecal Fungal Microbiome of Patients With COVID-19 During Time of Hospitalization until Discharge. Gastroenterology 2020; 159:1302-1310.e5. [PMID: 32598884 PMCID: PMC7318920 DOI: 10.1053/j.gastro.2020.06.048] [Citation(s) in RCA: 226] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infects intestinal cells, and might affect the intestinal microbiota. We investigated changes in the fecal fungal microbiomes (mycobiome) of patients with SARS-CoV-2 infection during hospitalization and on recovery. METHODS We performed deep shotgun metagenomic sequencing analysis of fecal samples from 30 patients with coronavirus disease 2019 (COVID-19) in Hong Kong, from February 5 through May 12, 2020. Fecal samples were collected 2 to 3 times per week from time of hospitalization until discharge. We compared fecal mycobiome compositions of patients with COVID-19 with those from 9 subjects with community-acquired pneumonia and 30 healthy individuals (controls). We assessed fecal mycobiome profiles throughout time of hospitalization until clearance of SARS-CoV-2 from nasopharyngeal samples. RESULTS Patients with COVID-19 had significant alterations in their fecal mycobiomes compared with controls, characterized by enrichment of Candia albicans and a highly heterogeneous mycobiome configuration, at time of hospitalization. Although fecal mycobiomes of 22 patients with COVID-19 did not differ significantly from those of controls during times of hospitalization, 8 of 30 patients with COVID-19 had continued significant differences in fecal mycobiome composition, through the last sample collected. The diversity of the fecal mycobiome of the last sample collected from patients with COVID-19 was 2.5-fold higher than that of controls (P < .05). Samples collected at all timepoints from patients with COVID-19 had increased proportions of opportunistic fungal pathogens, Candida albicans, Candida auris, and Aspergillus flavus compared with controls. Two respiratory-associated fungal pathogens, A. flavus and Aspergillus niger, were detected in fecal samples from a subset of patients with COVID-19, even after clearance of SARS-CoV-2 from nasopharyngeal samples and resolution of respiratory symptoms. CONCLUSIONS In a pilot study, we found heterogeneous configurations of the fecal mycobiome, with enrichment of fungal pathogens from the genera Candida and Aspergillus, during hospitalization of 30 patients with COVID-19 compared with controls. Unstable gut mycobiomes and prolonged dysbiosis persisted in a subset of patients with COVID-19 up to 12 days after nasopharyngeal clearance of SARS-CoV-2. Studies are needed to determine whether alterations in intestinal fungi contribute to or result from SARS-CoV-2 infection, and the effects of these changes in disease progression.
Collapse
Affiliation(s)
- Tao Zuo
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China; State Key Laboratory for Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Hui Zhan
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China; State Key Laboratory for Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Fen Zhang
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China; State Key Laboratory for Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Qin Liu
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China; State Key Laboratory for Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Eugene Y K Tso
- Department of Medicine and Geriatrics, United Christian Hospital, Hong Kong, China
| | - Grace C Y Lui
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Nan Chen
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Amy Li
- State Key Laboratory for Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Wenqi Lu
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Francis K L Chan
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China; State Key Laboratory for Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Paul K S Chan
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Department of Microbiology, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Siew C Ng
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China; State Key Laboratory for Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
16
|
Arabi Monfared A, Ayatollahi Mousavi SA, Zomorodian K, Mehrabani D, Iraji A, Moein MR. Trachyspermum ammi aromatic water: A traditional drink with considerable anti- Candida activity. Curr Med Mycol 2020; 6:1-8. [PMID: 33834136 PMCID: PMC8018821 DOI: 10.18502/cmm.6.3.3979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 05/22/2020] [Accepted: 07/28/2020] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Aromatic waters (AWs) are therapeutic distillates, which harbor both essential oil and water-soluble components of a plant. Due to the dispersion of the light amount of essence through the AWs, they have their specific pleasant smell, taste, and medicinal properties. In Iranian traditional medicine, Trachyspermum ammi AW is used to treat gastrointestinal disorders. The present study was conducted to determine the chemical composition of the essential oil extracted from T. ammi AW and its antifungal activities against Candida species. MATERIALS AND METHODS The composition of the essential oil extracted from T. ammi AW was analyzed by gas chromatography-mass spectrometry. In addition, the evaluation of the antifungal activity of AW against Candida species was performed using broth microdilution methods as recommended by the Clinical Laboratory Standard Institute. Moreover, the biofilm formation inhibition, antioxidant properties, and experimental activity of AW were determined in an animal model. RESULTS According to the results, thymol (78.08%) was the major compound of EO, followed by carvacrol (8.20%) and carvotanacetone (6.50%). Furthermore, T. ammi AW exhibited antifungal activities against the examined fungi and inhibited the biofilm formation of C. albicans at a concentration of up to 0.25 V/V. Histopathological analyses revealed that Candida colonization declined in the mice following the administration ofT. ammi AW in a therapeutic trial. CONCLUSION It seems that the presence of phenolic monoterpenes in AW has resulted in antifungal effects. Pleasant odor and antioxidant properties are extra bonuses to the antimicrobial effects of this plant. Based on the findings, AW might have the potential to be used in the management of alimentary candidiasis or oral hygienic products.
Collapse
Affiliation(s)
- Ali Arabi Monfared
- Department of Medical Mycology and Parasitology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyyed Amin Ayatollahi Mousavi
- Department of Medical Mycology and Parasitology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Kamiar Zomorodian
- Basic Sciences in Infectious Diseases Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Davood Mehrabani
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aida Iraji
- Basic Sciences in Infectious Diseases Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahmood Reza Moein
- Department of Pharmacognosy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
17
|
Pareek S, Kurakawa T, Das B, Motooka D, Nakaya S, Rongsen-Chandola T, Goyal N, Kayama H, Dodd D, Okumura R, Maeda Y, Fujimoto K, Nii T, Ogawa T, Iida T, Bhandari N, Kida T, Nakamura S, Nair GB, Takeda K. Comparison of Japanese and Indian intestinal microbiota shows diet-dependent interaction between bacteria and fungi. NPJ Biofilms Microbiomes 2019; 5:37. [PMID: 31885873 PMCID: PMC6925221 DOI: 10.1038/s41522-019-0110-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/22/2019] [Indexed: 02/07/2023] Open
Abstract
The bacterial species living in the gut mediate many aspects of biological processes such as nutrition and activation of adaptive immunity. In addition, commensal fungi residing in the intestine also influence host health. Although the interaction of bacterium and fungus has been shown, its precise mechanism during colonization of the human intestine remains largely unknown. Here, we show interaction between bacterial and fungal species for utilization of dietary components driving their efficient growth in the intestine. Next generation sequencing of fecal samples from Japanese and Indian adults revealed differential patterns of bacterial and fungal composition. In particular, Indians, who consume more plant polysaccharides than Japanese, harbored increased numbers of Prevotella and Candida. Candida spp. showed strong growth responses to the plant polysaccharide arabinoxylan in vitro. Furthermore, the culture supernatants of Candida spp. grown with arabinoxylan promoted rapid proliferation of Prevotella copri. Arabinose was identified as a potential growth-inducing factor in the Candida culture supernatants. Candida spp. exhibited a growth response to xylose, but not to arabinose, whereas P. copri proliferated in response to both xylose and arabinose. Candida spp., but not P. copri, colonized the intestine of germ-free mice. However, P. copri successfully colonized mouse intestine already harboring Candida. These findings demonstrate a proof of concept that fungal members of gut microbiota can facilitate a colonization of the intestine by their bacterial counterparts, potentially mediated by a dietary metabolite.
Collapse
Affiliation(s)
- Siddhika Pareek
- 1Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871 Japan.,2WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871 Japan.,3Core Research for Evolutional Science and Technology, Japan Agency for Medical Research and Development, Tokyo, 100-0004 Japan
| | - Takashi Kurakawa
- 1Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871 Japan.,2WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871 Japan
| | - Bhabatosh Das
- 4Molecular Genetics Laboratory, Center for Human Microbial Ecology, Translational Health Science and Technology Institute, Faridabad, 121001 India
| | - Daisuke Motooka
- 5Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, 565-0871 Japan
| | - Shuuichi Nakaya
- 6Global Applications Development Center, Shimadzu Corp, Kyoto, 604-8511 Japan
| | | | - Nidhi Goyal
- 7Centre for Health Research and Development, Society for Applied Studies, New Delhi, 110016 India
| | - Hisako Kayama
- 1Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871 Japan.,2WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871 Japan.,3Core Research for Evolutional Science and Technology, Japan Agency for Medical Research and Development, Tokyo, 100-0004 Japan
| | - Dylan Dodd
- 8Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Ryu Okumura
- 1Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871 Japan.,2WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871 Japan.,3Core Research for Evolutional Science and Technology, Japan Agency for Medical Research and Development, Tokyo, 100-0004 Japan
| | - Yuichi Maeda
- 1Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871 Japan.,2WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871 Japan.,9Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Kosuke Fujimoto
- 1Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871 Japan.,9Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Takuro Nii
- 1Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871 Japan.,2WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871 Japan.,9Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Takao Ogawa
- 1Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871 Japan.,2WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871 Japan.,9Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Tetsuya Iida
- 5Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, 565-0871 Japan.,10Department of Bacterial Infections, Research Institute for Microbial Diseases, Osaka University, Osaka, 565-0871 Japan
| | - Nita Bhandari
- 7Centre for Health Research and Development, Society for Applied Studies, New Delhi, 110016 India
| | - Toshiyuki Kida
- 11Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, 565-0871 Japan
| | - Shota Nakamura
- 5Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, 565-0871 Japan
| | - G Balakrish Nair
- 4Molecular Genetics Laboratory, Center for Human Microbial Ecology, Translational Health Science and Technology Institute, Faridabad, 121001 India
| | - Kiyoshi Takeda
- 1Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871 Japan.,2WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871 Japan.,3Core Research for Evolutional Science and Technology, Japan Agency for Medical Research and Development, Tokyo, 100-0004 Japan
| |
Collapse
|
18
|
Trained Innate Immunity and Its Implications for Mucosal Immunity and Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1197:11-26. [PMID: 31732931 DOI: 10.1007/978-3-030-28524-1_2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The long-standing dogma that immunological memory is the exclusive prerogative of the adaptive immune system has been challenged by emerging evidence that innate immunity can also maintain memory of past events. Such immunological imprinting takes two forms, trained innate immunity and tolerance. Trained immunity involves metabolic and epigenetic adaptations in innate immune cells and their progenitors in the bone marrow upon exposure to certain microbial and/or inflammatory stimuli so that the "trained" cells would be poised to respond much faster and stronger to a subsequent challenge (e.g., a new infection that is not necessarily the same as the earlier one). Conversely, tolerance leads to attenuated immune responses to secondary stimuli. This review focuses on trained immunity and discusses evidence for its existence from lower organisms to humans, its mechanistic underpinnings, and its translational ramifications. Although trained immunity can be considered as an evolutionarily conserved beneficial response against reinfections, in the setting of modern societies with high prevalence of chronic mucosal and systemic inflammatory diseases, trained immunity could also promote maladaptive immune responses that aggravate pathology. Thus, depending on context, innate immune memory could be therapeutically manipulated using defined agonists to either promote innate immune responses (particularly useful for the treatment of infections or chemotherapy-induced myelosuppression) or suppress excessive inflammation in inflammatory and autoimmune diseases.
Collapse
|
19
|
Gürsoy S, Koçkar T, Atik SU, Önal Z, Önal H, Adal E. Autoimmunity and intestinal colonization by Candida albicans in patients with type 1 diabetes at the time of the diagnosis. KOREAN JOURNAL OF PEDIATRICS 2018; 61:217-220. [PMID: 30032588 PMCID: PMC6106689 DOI: 10.3345/kjp.2018.61.7.217] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 03/05/2018] [Indexed: 11/27/2022]
Abstract
PURPOSE Type 1 diabetes mellitus (T1DM) is a chronic and immune-mediated disease, which is characterized by the progressive destruction of pancreatic beta cells. T1DM precipitates in genetically susceptible individuals through environmental factors. In this study, we aimed to evaluate the impact of autoimmunity and intestinal colonization of Candida albicans on the development of T1DM. METHODS Forty-two patients newly diagnosed with T1DM and 42 healthy subjects were included in this monocentric study. The basic and clinical characteristics of the patients were recorded. T1DM-, thyroid-, and celiac-associated antibodies were evaluated. Stool cultures for C. albicans were performed to assess whether or not gut integrity was impaired in patients with T1DM. RESULTS The evaluation of T1DM- and thyroid-associated antibodies showed that the prevalences of islet cell antibodies and antithyroperoxidase positivity were higher in the study patients than in the patients in the control group. Furthermore, the direct examination and culture of fresh stool samples revealed that 50% of the patients with T1DM and 23.8% of the control subjects had fungi (C. albicans). CONCLUSION Through this study, we suggest that the presence of intestinal C. albicans colonization at the time of the diagnosis of T1DM may indicate impairment of normal intestinal microbiota. We also suggest that there may be a tendency of T1DM in patients with a high prevalence of intestinal C. albicans.
Collapse
Affiliation(s)
- Semra Gürsoy
- Department of Pediatrics, Kanuni Sultan Suleyman Training and Research Hospital, Istanbul, Turkey
| | - Tuba Koçkar
- Department of Pediatrics, Kanuni Sultan Suleyman Training and Research Hospital, Istanbul, Turkey
| | - Sezen Ugan Atik
- Department of Pediatrics, Kanuni Sultan Suleyman Training and Research Hospital, Istanbul, Turkey
| | - Zerrin Önal
- Department of Pediatric Gastroenterology, Kanuni Sultan Suleyman Training and Research Hospital, Istanbul, Turkey
| | - Hasan Önal
- Department of Pediatric Metabolic Disease, Kanuni Sultan Suleyman Training and Research Hospital, Istanbul, Turkey
| | - Erdal Adal
- Department of Pediatric Endocrinology and Metabolism, Kanuni Sultan Suleyman Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
20
|
Li J, Chen D, Yu B, He J, Zheng P, Mao X, Yu J, Luo J, Tian G, Huang Z, Luo Y. Fungi in Gastrointestinal Tracts of Human and Mice: from Community to Functions. MICROBIAL ECOLOGY 2018; 75:821-829. [PMID: 29110065 DOI: 10.1007/s00248-017-1105-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/30/2017] [Indexed: 05/10/2023]
Abstract
Fungi are often ignored in studies on gut microbes because of their low level of presence (making up only 0.1% of the total microorganisms) in the gastrointestinal tract (GIT) of monogastric animals. Recent studies using novel technologies such as next generation sequencing have expanded our understanding on the importance of intestinal fungi in humans and animals. Here, we provide a comprehensive review on the fungal community, the so-called mycobiome, and their functions from recent studies in humans and mice. In the GIT of humans, fungi belonging to the phyla Ascomycota, Basidiomycota and Chytridiomycota are predominant. The murine intestines harbor a more diverse assemblage of fungi. Diet is one of the major factors influencing colonization of fungi in the GIT. Presence of the genus Candida is positively associated with dietary carbohydrates, but are negatively correlated with dietary amino acids, proteins, and fatty acids. However, the relationship between diet and the fungal community (and functions), as well as the underlying mechanisms remains unclear. Dysbiosis of intestinal fungi can cause invasive infections and inflammatory bowel diseases (IBD). However, it is not clear whether dysbiosis of the mycobiome is a cause, or a result of IBD. Compared to non-inflamed intestinal mucosa, the abundance and diversity of fungi is significantly increased in the inflamed mucosa. The commonly observed commensal fungal species Candida albicans might contribute to occurrence and development of IBD. Limited studies show that Candida albicans might interact with immune cells of the host intestines through the pathways associated with Dectin-1, Toll-like receptor 2 (TLR2), and TLR4. This review is expected to provide new thoughts for future studies on intestinal fungi and for new therapies to fungal infections in the GIT of human and animals.
Collapse
Affiliation(s)
- Jiayan Li
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Jun He
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Ping Zheng
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Xiangbing Mao
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Jie Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Junqiu Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Gang Tian
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Yuheng Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China.
| |
Collapse
|
21
|
Laurence M, Asquith M, Rosenbaum JT. Spondyloarthritis, Acute Anterior Uveitis, and Fungi: Updating the Catterall-King Hypothesis. Front Med (Lausanne) 2018; 5:80. [PMID: 29675414 PMCID: PMC5895656 DOI: 10.3389/fmed.2018.00080] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/09/2018] [Indexed: 12/12/2022] Open
Abstract
Spondyloarthritis is a common type of arthritis which affects mostly adults. It consists of idiopathic chronic inflammation of the spine, joints, eyes, skin, gut, and prostate. Inflammation is often asymptomatic, especially in the gut and prostate. The HLA-B*27 allele group, which presents intracellular peptides to CD8+ T cells, is by far the strongest risk factor for spondyloarthritis. The precise mechanisms and antigens remain unknown. In 1959, Catterall and King advanced a novel hypothesis explaining the etiology of spondyloarthritis: an as-yet-unrecognized sexually acquired microbe would be causing all spondyloarthritis types, including acute anterior uveitis. Recent studies suggest an unrecognized sexually acquired fungal infection may be involved in prostate cancer and perhaps multiple sclerosis. This warrants reanalyzing the Catterall-King hypothesis based on the current literature. In the last decade, many links between spondyloarthritis and fungal infections have been found. Antibodies against the fungal cell wall component mannan are elevated in spondyloarthritis. Functional polymorphisms in genes regulating the innate immune response against fungi have been associated with spondyloarthritis (CARD9 and IL23R). Psoriasis and inflammatory bowel disease, two common comorbidities of spondyloarthritis, are both strongly associated with fungi. Evidence reviewed here lends credence to the Catterall-King hypothesis and implicates a common fungal etiology in prostate cancer, benign prostatic hyperplasia, multiple sclerosis, psoriasis, inflammatory bowel disease, and spondyloarthritis. However, the evidence available at this time is insufficient to definitely confirm this hypothesis. Future studies investigating the microbiome in relation to these conditions should screen specimens for fungi in addition to bacteria. Future clinical studies of spondyloarthritis should consider antifungals which are effective in psoriasis and multiple sclerosis, such as dimethyl fumarate and nystatin.
Collapse
Affiliation(s)
| | - Mark Asquith
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, United States
| | - James T Rosenbaum
- Department of Ophthalmology, Oregon Health and Science University, Portland, OR, United States.,Department of Medicine, Oregon Health and Science University, Portland, OR, United States.,Department of Cell Biology, Oregon Health and Science University, Portland, OR, United States.,Legacy Devers Eye Institute, Portland, OR, United States
| |
Collapse
|
22
|
Maekawa T, Ishijima AS, Ida M, Izumo T, Ono Y, Shibata H, Abe S. Prophylactic Effect of Lactobacillus pentosus strain S-PT84 on Candida Infection and Gastric Inflammation in a Murine Gastrointestinal Candidiasis Model [Errata]. Med Mycol J 2017; 57:E81-E92. [PMID: 27904074 DOI: 10.3314/mmj.16-00012e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We previously showed a prophylactic effect of Lactobacillus pentosus strain S-PT84 against oral candidiasis in mice. In the present study, we evaluated the protective effect of S-PT84 against Candida infection of the gastrointestinal tract. As the first step, we used an in vitro assay to compare the inhibitory effects of several lactobacilli (S-PT84 and Lactobacillus pentosus type strain JCM1558T, Lactobacillus gasseri type strain JCM1131T and Lactobacillus casei type strain JCM1134T) on mycelial growth of Candida albicans. S-PT84 directly adhered to Candida cells and showed the strongest growth-inhibitory activity among the tested Lactobacillus strains. In the second experiment, we used an in vivo assay to evaluate the effect of S-PT84 ingestion on severity score of stomach lesion and gastric inflammation in a mouse model of gastrointestinal candidiasis. The severity scores were significantly improved by oral administration of S-PT84 (6 mg/ 200 μL), consistent with decreased coverage of stomach lesions by patchy whitish plaques. The attenuation of stomach lesion severity by S-PT84 was more pronounced than that obtained with L. gasseri type strain JCM1131T, consistent with the results of the above in vitro study. Histological analysis also indicated that S-PT84 prevented the adhesion of C. albicans to the stomach surface and suppressed stomach inflammation caused by neutrophil infiltration. Furthermore, S-PT84 also suppressed the vascular permeability observed in Candida-infected stomach. These results suggest that oral administration of S-PT84 might be effective not only in inhibiting Candida infection but also in preventing gastric inflammation induced by Candida infection.
Collapse
|
23
|
Motooka D, Fujimoto K, Tanaka R, Yaguchi T, Gotoh K, Maeda Y, Furuta Y, Kurakawa T, Goto N, Yasunaga T, Narazaki M, Kumanogoh A, Horii T, Iida T, Takeda K, Nakamura S. Fungal ITS1 Deep-Sequencing Strategies to Reconstruct the Composition of a 26-Species Community and Evaluation of the Gut Mycobiota of Healthy Japanese Individuals. Front Microbiol 2017; 8:238. [PMID: 28261190 PMCID: PMC5309391 DOI: 10.3389/fmicb.2017.00238] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 02/03/2017] [Indexed: 12/18/2022] Open
Abstract
The study of mycobiota remains relatively unexplored due to the lack of sufficient available reference strains and databases compared to those of bacterial microbiome studies. Deep sequencing of Internal Transcribed Spacer (ITS) regions is the de facto standard for fungal diversity analysis. However, results are often biased because of the wide variety of sequence lengths in the ITS regions and the complexity of high-throughput sequencing (HTS) technologies. In this study, a curated ITS database, ntF-ITS1, was constructed. This database can be utilized for the taxonomic assignment of fungal community members. We evaluated the efficacy of strategies for mycobiome analysis by using this database and characterizing a mock fungal community consisting of 26 species representing 15 genera using ITS1 sequencing with three HTS platforms: Illumina MiSeq (MiSeq), Ion Torrent Personal Genome Machine (IonPGM), and Pacific Biosciences (PacBio). Our evaluation demonstrated that PacBio's circular consensus sequencing with greater than 8 full-passes most accurately reconstructed the composition of the mock community. Using this strategy for deep-sequencing analysis of the gut mycobiota in healthy Japanese individuals revealed two major mycobiota types: a single-species type composed of Candida albicans or Saccharomyces cerevisiae and a multi-species type. In this study, we proposed the best possible processing strategies for the three sequencing platforms, of which, the PacBio platform allowed for the most accurate estimation of the fungal community. The database and methodology described here provide critical tools for the emerging field of mycobiome studies.
Collapse
Affiliation(s)
- Daisuke Motooka
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University Suita, Japan
| | - Kosuke Fujimoto
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka UniversitySuita, Japan; Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Reiko Tanaka
- Division of Bio-resources, Medical Mycology Research Center, Chiba University Chiba, Japan
| | - Takashi Yaguchi
- Division of Bio-resources, Medical Mycology Research Center, Chiba University Chiba, Japan
| | - Kazuyoshi Gotoh
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka UniversitySuita, Japan; Department of Bacteriology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama UniversityOkayama, Japan
| | - Yuichi Maeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka UniversitySuita, Japan; Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Yoki Furuta
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University Suita, Japan
| | - Takashi Kurakawa
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University Suita, Japan
| | - Naohisa Goto
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University Suita, Japan
| | - Teruo Yasunaga
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University Suita, Japan
| | - Masashi Narazaki
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Graduate School of Medicine, Osaka University Suita, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Graduate School of Medicine, Osaka University Suita, Japan
| | - Toshihiro Horii
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University Suita, Japan
| | - Tetsuya Iida
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University Suita, Japan
| | - Kiyoshi Takeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University Suita, Japan
| | - Shota Nakamura
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University Suita, Japan
| |
Collapse
|
24
|
Maekawa T, Ishijima AS, Ida M, Izumo T, Ono Y, Shibata H, Abe S. Prophylactic Effect of Lactobacillus pentosus strain S-PT84 on Candida Infection and Gastric Inflammation in a Murine Gastrointestinal Candidiasis Model. Med Mycol J 2016; 57:E81-E92. [PMID: 27904056 DOI: 10.3314/mmj.16-00012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We previously showed a prophylactic effect of Lactobacillus pentosus strain S-PT84 against oral candidiasis in mice. In the present study, we evaluated the protective effect of S-PT84 against Candida infection of the gastrointestinal tract. As the first step, we used an in vitro assay to compare the inhibitory effects of several lactobacilli (S-PT84 and Lactobacillus pentosus type strain JCM1558T, Lactobacillus gasseri type strain JCM1131T and Lactobacillus casei type strain JCM1134T) on mycelial growth of Candida albicans. S-PT84 directly adhered to Candida cells and showed the strongest growth-inhibitory activity among the tested Lactobacillus strains. In the second experiment, we used an in vivo assay to evaluate the effect of S-PT84 ingestion on severity score of stomach lesion and gastric inflammation in a mouse model of gastrointestinal candidiasis. The severity scores were significantly improved by oral administration of S-PT84 (6 mg/ 200 μL), consistent with decreased coverage of stomach lesions by patchy whitish plaques. The attenuation of stomach lesion severity by S-PT84 was more pronounced than that obtained with L. gasseri type strain JCM1131T, consistent with the results of the above in vitro study. Histological analysis also indicated that S-PT84 prevented the adhesion of C. albicans to the stomach surface and suppressed stomach inflammation caused by neutrophil infiltration. Furthermore, S-PT84 also suppressed the vascular permeability observed in Candida-infected stomach. These results suggest that oral administration of S-PT84 might be effective not only in inhibiting Candida infection but also in preventing gastric inflammation induced by Candida infection.
Collapse
|
25
|
Carrière J, Darfeuille-Michaud A, Nguyen HTT. Infectious etiopathogenesis of Crohn’s disease. World J Gastroenterol 2014; 20:12102-12117. [PMID: 25232246 PMCID: PMC4161797 DOI: 10.3748/wjg.v20.i34.12102] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 04/18/2014] [Accepted: 05/26/2014] [Indexed: 02/07/2023] Open
Abstract
Important advances during the last decade have been made in understanding the complex etiopathogenesis of Crohn’s disease (CD). While many gaps in our knowledge still exist, it has been suggested that the etiology of CD is multifactorial including genetic, environmental and infectious factors. The most widely accepted theory states that CD is caused by an aggressive immune response to infectious agents in genetically predisposed individuals. The rise of genome-wide association studies allowed the identification of loci and genetic variants in several components of host innate and adaptive immune responses to microorganisms in the gut, highlighting an implication of intestinal microbiota in CD etiology. Moreover, numerous independent studies reported a dysbiosis, i.e., a modification of intestinal microbiota composition, with an imbalance between the abundance of beneficial and harmful bacteria. Although microorganisms including viruses, yeasts, fungi and bacteria have been postulated as potential CD pathogens, based on epidemiological, clinicopathological, genetic and experimental evidence, their precise role in this disease is not clearly defined. This review summarizes the current knowledge of the infectious agents associated with an increased risk of developing CD. Therapeutic approaches to modulate the intestinal dysbiosis and to target the putative CD-associated pathogens, as well as their potential mechanisms of action are also discussed.
Collapse
|
26
|
Soyucen E, Gulcan A, Aktuglu-Zeybek AC, Onal H, Kiykim E, Aydin A. Differences in the gut microbiota of healthy children and those with type 1 diabetes. Pediatr Int 2014; 56:336-43. [PMID: 24475780 DOI: 10.1111/ped.12243] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Revised: 08/06/2013] [Accepted: 10/28/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND Intestinal barriers, intestinal flora, and mucosal immunity are the main factors responsible for the development of various allergic and autoimmune diseases. The aim of this study was to investigate the relationship between the intestinal flora of children and the presence of type 1 diabetes, and to determine if gut microbiota could partly explain the etiology of the disease. METHODS Fecal flora analysis was done using quantitative cultures on selective and non-selective media with different thermal and atmospheric conditions for bacterial and fungal growth. The study group consisted of 35 patients (16 female, 19 male; mean age, 10.73 ± 4.16 years), who had been followed by the University of Istanbul, Cerrahpasa Medical Faculty, Department of Pediatrics, and were newly diagnosed with type 1 diabetes. The control group consisted of 35 healthy subjects (15 female, 20 male; mean age, 9.96 ± 4.09 years), who were randomly selected and had similar demographics. RESULTS Bifidobacterium colonization was lower in patients with type 1 diabetes compared to the control group, whereas Candida albicans and Enterobacteriaceae other than Echerichia coli colonization was increased. CONCLUSION A decrease in beneficial anaerobic bacteria levels and a concomitant increase in Enterobacteriaceae other than E. coli and C. albicans colonization may lead to a disturbance in the ecological balance of intestinal flora, which could be a triggering factor in type 1 diabetes etiology.
Collapse
Affiliation(s)
- Erdogan Soyucen
- Department of Pediatric Metabolic Disease, Akdeniz University Medical Faculty, Antalya
| | | | | | | | | | | |
Collapse
|
27
|
Sakurai A, Yamaguchi N, Sonoyama K. Cell Wall Polysaccharides of Candida albicans Induce Mast Cell Degranulation in the Gut. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2012; 31:67-70. [PMID: 24936351 PMCID: PMC4034280 DOI: 10.12938/bmfh.31.67] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Accepted: 02/16/2012] [Indexed: 02/06/2023]
Abstract
We investigated Candida albicans-induced mast cell degranulation in vitro and in vivo. Cell wall fraction but not culture supernatant and cell membrane fraction prepared from hyphally grown C. albicans induced β-hexosaminidase release in RBL-2H3 cells. Cell wall mannan and soluble β-glucan fractions also induced β-hexosaminidase release. Histological examination of mouse forestomach showed that C. albicans gut colonization induces mast cell degranulation. However, intragastric administration of cell wall fraction failed to induce mast cell degranulation. We propose that cell wall polysaccharides are responsible for mast cell degranulation in the C. albicans-colonized gut.
Collapse
Affiliation(s)
- Atsuko Sakurai
- Graduate School of Life Science, Hokkaido University, Kita 9, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-8589, Japan
| | - Natsu Yamaguchi
- Graduate School of Agriculture, Hokkaido University, Kita 9, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-8589, Japan ; Present address: Faculty of Medicine, Shimane University, Izumo, Shimane 693-8501, Japan
| | - Kei Sonoyama
- Research Faculty of Agriculture, Hokkaido University, Kita 9, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-8589, Japan
| |
Collapse
|
28
|
Marijnissen RJ, Koenders MI, van de Veerdonk FL, Dulos J, Netea MG, Boots AMH, Joosten LAB, van den Berg WB. Exposure to Candida albicans polarizes a T-cell driven arthritis model towards Th17 responses, resulting in a more destructive arthritis. PLoS One 2012; 7:e38889. [PMID: 22719976 PMCID: PMC3373564 DOI: 10.1371/journal.pone.0038889] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 05/14/2012] [Indexed: 11/19/2022] Open
Abstract
Background Fungal components have been shown very effective in generating Th17 responses. We investigated whether exposure to a minute amount of C. albicans in the arthritic joint altered the local cytokine environment, leading to enhanced Th17 expansion and resulting in a more destructive arthritis. Methodology Chronic SCW arthritis was induced by repeated injection with Streptococcus pyogenes (SCW) cell wall fragments into the knee joint of C57Bl/6 mice, alone or in combination with the yeast of C. albicans or Zymosan A. During the chronic phase of the arthritis, the cytokine levels, mRNA expression and histopathological analysis of the joints were performed. To investigate the phenotype of the IL-17 producing T-cells, synovial cells were isolated and analyzed by flowcytometry. Principal Findings Intra-articular injection of either Zymosan A or C. albicans on top of the SCW injection both resulted in enhanced joint swelling and inflammation compared to the normal SCW group. However, only the addition of C. albicans during SCW arthritis resulted in severe chondrocyte death and enhanced destruction of cartilage and bone. Additionally, exposure to C. albicans led to increased IL-17 in the arthritic joint, which was accompanied by an increased synovial mRNA expression of T-bet and RORγT. Moreover, the C. albicans-injected mice had significantly more Th17 cells in the synovium, of which a large population also produced IFN-γ. Conclusion This study clearly shows that minute amounts of fungal components, like C. albicans, are very potent in interfering with the local cytokine environment in an arthritic joint, thereby polarizing arthritis towards a more destructive phenotype.
Collapse
Affiliation(s)
- Renoud J Marijnissen
- Rheumatology Research and Advanced Therapeutics, Department of Rheumatology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Romanowski K, Zaborin A, Valuckaite V, Rolfes RJ, Babrowski T, Bethel C, Olivas A, Zaborina O, Alverdy JC. Candida albicans isolates from the gut of critically ill patients respond to phosphate limitation by expressing filaments and a lethal phenotype. PLoS One 2012; 7:e30119. [PMID: 22253901 PMCID: PMC3258262 DOI: 10.1371/journal.pone.0030119] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 12/09/2011] [Indexed: 12/29/2022] Open
Abstract
Candida albicans is an opportunistic pathogen that proliferates in the intestinal tract of critically ill patients where it continues to be a major cause of infectious-related mortality. The precise cues that shift intestinal C. albicans from its ubiquitous indolent colonizing yeast form to an invasive and lethal filamentous form remain unknown. We have previously shown that severe phosphate depletion develops in the intestinal tract during extreme physiologic stress and plays a major role in shifting intestinal Pseudomonas aeruginosa to express a lethal phenotype via conserved phosphosensory-phosphoregulatory systems. Here we studied whether phosphate dependent virulence expression could be similarly demonstrated for C. albicans. C. albicans isolates from the stool of critically ill patients and laboratory prototype strains (SC5314, BWP17, SN152) were evaluated for morphotype transformation and lethality against C. elegans and mice during exposure to phosphate limitation. Isolates ICU1 and ICU12 were able to filament and kill C. elegans in a phosphate dependent manner. In a mouse model of intestinal phosphate depletion (30% hepatectomy), direct intestinal inoculation of C. albicans caused mortality that was prevented by oral phosphate supplementation. Prototype strains displayed limited responses to phosphate limitation; however, the pho4Δ mutant displayed extensive filamentation during low phosphate conditions compared to its isogenic parent strain SN152, suggesting that mutation in the transcriptional factor Pho4p may sensitize C. albicans to phosphate limitation. Extensive filamentation was also observed in strain ICU12 suggesting that this strain is also sensitized to phosphate limitation. Analysis of the sequence of PHO4 in strain ICU12, its transcriptional response to phosphate limitation, and phosphatase assays confirmed that ICU12 demonstrates a profound response to phosphate limitation. The emergence of strains of C. albicans with marked responsiveness to phosphate limitation may represent a fitness adaptation to the complex and nutrient scarce environment typical of the gut of a critically ill patient.
Collapse
Affiliation(s)
- Kathleen Romanowski
- Department of Surgery, University of Chicago, Chicago, Illinois, United States of America
| | - Alexander Zaborin
- Department of Surgery, University of Chicago, Chicago, Illinois, United States of America
| | - Vesta Valuckaite
- Department of Surgery, University of Chicago, Chicago, Illinois, United States of America
| | - Ronda J. Rolfes
- Department of Biology, Georgetown University, Washington, D. C., United States of America
| | - Trissa Babrowski
- Department of Surgery, University of Chicago, Chicago, Illinois, United States of America
| | - Cindy Bethel
- Clinical Microbiology/Immunology Laboratories, University of Chicago, Chicago, Illinois, United States of America
| | - Andrea Olivas
- Department of Surgery, University of Chicago, Chicago, Illinois, United States of America
| | - Olga Zaborina
- Department of Surgery, University of Chicago, Chicago, Illinois, United States of America
| | - John C. Alverdy
- Department of Surgery, University of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
30
|
Sugita R, Hata E, Miki A, Andoh R, Umeda C, Takemura N, Sonoyama K. Gut Colonization by Candida albicans Inhibits the Induction of Humoral Immune Tolerance to Dietary Antigen in BALB/c Mice. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2012; 31:77-84. [PMID: 24936353 PMCID: PMC4034285 DOI: 10.12938/bmfh.31.77] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 05/07/2012] [Indexed: 12/27/2022]
Abstract
We previously observed that gut colonization by Candida albicans
promoted serum antibody response to orally administered ovalbumin in mice. We therefore
postulated that C. albicans affects oral tolerance induction. The present
study tested this idea. BALB/c mice were intragastrically administered with either
C. albicans (1 × 107) or vehicle, and the colonization was
confirmed by weekly fecal cultures. Mice were further divided into two subgroups and
intragastrically administered with either ovalbumin (20 mg) or vehicle for five
consecutive days. Thereafter, all mice were intraperitoneally immunized with ovalbumin in
alum. In mice without C. albicans inoculation, ovalbumin feeding prior to
immunization significantly suppressed the increase in ovalbumin-specific IgE, IgG1 and
IgG2a in sera, suggesting oral tolerance induction. In C.
albicans-inoculated mice, however, the antibody levels were the same between
ovalbumin- and vehicle-fed mice. In contrast, ovalbumin feeding significantly suppressed
cellular immune responses, as evidenced by reduced proliferation of splenocytes
restimulated by ovalbumin ex vivo, in both C.
albicans-inoculated and uninoculated mice. Ex vivo
supplementation with neither heat-killed C. albicans nor the culture
supernatant of C. albicans enhanced the production of ovalbumin-specific
IgG1 in splenocytes restimulated by the antigen. These results suggest that gut
colonization by C. albicans inhibits the induction of humoral immune
tolerance to dietary antigen in mice, whereas C. albicans may not
directly promote antibody production. We therefore propose that C.
albicans gut colonization could be a risk factor for triggering food allergy in
susceptible individuals.
Collapse
Affiliation(s)
- Ryusuke Sugita
- Graduate School of Agriculture, Hokkaido University, Kita 9 Nishi 9, Kita-ku, Sapporo 060-8589, Japan
| | - Erina Hata
- Graduate School of Life Science, Hokkaido University, Kita 9 Nishi 9, Kita-ku, Sapporo 060-8589, Japan
| | - Atsuko Miki
- Graduate School of Life Science, Hokkaido University, Kita 9 Nishi 9, Kita-ku, Sapporo 060-8589, Japan
| | - Ryoko Andoh
- Graduate School of Agriculture, Hokkaido University, Kita 9 Nishi 9, Kita-ku, Sapporo 060-8589, Japan
| | - Chisato Umeda
- Graduate School of Agriculture, Hokkaido University, Kita 9 Nishi 9, Kita-ku, Sapporo 060-8589, Japan
| | - Naoki Takemura
- Graduate School of Agriculture, Hokkaido University, Kita 9 Nishi 9, Kita-ku, Sapporo 060-8589, Japan
| | - Kei Sonoyama
- Research Faculty of Agriculture, Hokkaido University, Kita 9 Nishi 9, Kita-ku, Sapporo 060-8589, Japan
| |
Collapse
|