51
|
Kumar R, Rojas IG, Edgerton M. Candida albicans Sap6 Initiates Oral Mucosal Inflammation via the Protease Activated Receptor PAR2. Front Immunol 2022; 13:912748. [PMID: 35844627 PMCID: PMC9277060 DOI: 10.3389/fimmu.2022.912748] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Candida albicans Sap6, a secreted aspartyl protease (Sap), contributes to fungal virulence in oral candidiasis. Beside its protease activity, Sap6 contains RGD (RGDRGD) motif required for its binding to host integrins. Sap6 activates immune cells to induce proinflammatory cytokines, although its ability to interact and activate human oral epithelial cells (OECs) remain unknown. Addition of purified recombinant Sap6 (rSap6) to OECs resulted in production of IL-1β and IL-8 cytokines similar to live hyphal C. albicans. OECs exposed to rSap6 showed phosphorylation of p38 and MKP1 and expression of c-Fos not found with C. albicans Δsap6, heat-inactivated Sap6, or rSap6ΔRGD . Heat inactivated rSap6 was able to induce IL-1β but not IL-8 in OECs, while rSap6ΔRGD induced IL-8 but not IL-1β suggesting parallel signaling pathways. C. albicans hyphae increased surface expression of Protease Activated Receptors PAR1, PAR2 and PAR3, while rSap6 increased PAR2 expression exclusively. Pretreatment of OECs with a PAR2 antagonist blocked rSap6-induced p38 MAPK signaling and IL-8 release, while rSap6ΔRGD had reduced MKP1 signaling and IL-1β release independent from PAR2. OECs exposed to rSap6 exhibited loss of barrier function as measured by TEER and reduction in levels of E-cadherin and occludin junctional proteins that was prevented by pretreating OECs with a PAR2 antagonist. OECs treated with PAR2 antagonist also showed reduced rSap6-mediated invasion by C. albicans cells. Thus, Sap6 may initiate OEC responses mediated both through protease activation of PAR2 and by its RGD domain. This novel role of PAR2 suggests new drug targets to block C. albicans oral infection.
Collapse
Affiliation(s)
| | | | - Mira Edgerton
- Department of Oral Biology, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
52
|
Longitudinal alterations of the gut mycobiota and microbiota on COVID-19 severity. BMC Infect Dis 2022; 22:572. [PMID: 35751044 PMCID: PMC9233337 DOI: 10.1186/s12879-022-07358-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 04/01/2022] [Indexed: 12/15/2022] Open
Abstract
Background The impact of SARS-CoV-2 infection on the gut fungal (mycobiota) and bacterial (microbiota) communities has been elucidated individually. This study analyzed both gut mycobiota and microbiota and their correlation in the COVID-19 patients with severe and mild conditions and follow-up to monitor their alterations after recovery. Methods We analyzed the gut mycobiota and microbiota by bacterial 16S and fungal ITS1 metagenomic sequencing of 40 severe patients, 38 mild patients, and 30 healthy individuals and reanalyzed those of 10 patients with severe COVID-19 approximately 6 months after discharge. Results The mycobiota of the severe and mild groups showed lower diversity than the healthy group, and in some, characteristic patterns dominated by a single fungal species, Candida albicans, were detected. Lower microbial diversity in the severe group was observed, but no differences in its diversity or community structure were detected between the mild and healthy groups. The microbiota of the severe group was characterized by an increase in Enterococcus and Lactobacillus, and a decrease in Faecalibacterium and Bacteroides. The abundance of Candida was positively correlated with that of Enterococcus in patients with COVID-19. After the recovery of severe patients, alteration of the microbiota remained, but the mycobiota recovered its diversity comparable to that of mild and healthy groups. Conclusion In mild cases, the microbiota is stable during SARS-CoV-2 infection, but in severe cases, alterations persist for 6 months after recovery. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-022-07358-7.
Collapse
|
53
|
The Role of Glycoside Hydrolases in S. gordonii and C. albicans Interactions. Appl Environ Microbiol 2022; 88:e0011622. [PMID: 35506689 DOI: 10.1128/aem.00116-22] [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] [Indexed: 02/05/2023] Open
Abstract
Candida albicans can coaggregate with Streptococcus gordonii and cocolonize in the oral cavity. Saliva provides a vital microenvironment for close interactions of oral microorganisms. However, the level of fermentable carbohydrates in saliva is not sufficient to support the growth of multiple species. Glycoside hydrolases (GHs) that hydrolyze glycoproteins are critical for S. gordonii growth in low-fermentable-carbohydrate environments such as saliva. However, whether GHs are involved in the cross-kingdom interactions between C. albicans and S. gordonii under such conditions remains unknown. In this study, C. albicans and S. gordonii were cocultured in heart infusion broth with a low level of fermentable carbohydrate. Planktonic growth, biofilm formation, cell aggregation, and GH activities of monocultures and cocultures were examined. The results revealed that the planktonic growth of cocultured S. gordonii in a low-carbohydrate environment was elevated, while that of cocultured C. albicans was reduced. The biomass of S. gordonii in dual-species biofilms was higher than that of monocultures, while that of cocultured C. albicans was decreased. GH activity was observed in S. gordonii, and elevated activity of GHs was detected in S. gordonii-C. albicans cocultures, with elevated expression of GH-related genes of S. gordonii. By screening a mutant library of C. albicans, we identified a tec1Δ/Δ mutant strain that showed reduced ability to promote the growth and GH activities of S. gordonii compared with the wild-type strain. Altogether, the findings of this study demonstrate the involvement of GHs in the cross-kingdom metabolic interactions between C. albicans and S. gordonii in an environment with low level of fermentable carbohydrates. IMPORTANCE Cross-kingdom interactions between Candida albicans and oral streptococci such as Streptococcus gordonii have been reported. However, their interactions in a low-fermentable-carbohydrate environment like saliva is not clear. The current study revealed glycoside hydrolase-related cross-kingdom communications between S. gordonii and C. albicans under the low-fermentable-carbohydrate condition. We demonstrate that C. albicans can promote the growth and metabolic activities of S. gordonii by elevating the activities of cell-wall-anchored glycoside hydrolases of S. gordonii. C. albicans gene TEC1 is critical for this cross-kingdom metabolic communication.
Collapse
|
54
|
Yadav M, Ali S, Shrode RL, Shahi SK, Jensen SN, Hoang J, Cassidy S, Olalde H, Guseva N, Paullus M, Cherwin C, Wang K, Cho T, Kamholz J, Mangalam AK. Multiple sclerosis patients have an altered gut mycobiome and increased fungal to bacterial richness. PLoS One 2022; 17:e0264556. [PMID: 35472144 PMCID: PMC9041819 DOI: 10.1371/journal.pone.0264556] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 02/13/2022] [Indexed: 12/13/2022] Open
Abstract
Trillions of microbes such as bacteria, fungi, and viruses exist in the healthy human gut microbiome. Although gut bacterial dysbiosis has been extensively studied in multiple sclerosis (MS), the significance of the fungal microbiome (mycobiome) is an understudied and neglected part of the intestinal microbiome in MS. The aim of this study was to characterize the gut mycobiome of patients with relapsing-remitting multiple sclerosis (RRMS), compare it to healthy controls, and examine its association with changes in the bacterial microbiome. We characterized and compared the mycobiome of 20 RRMS patients and 33 healthy controls (HC) using Internal Transcribed Spacer 2 (ITS2) and compared mycobiome interactions with the bacterial microbiome using 16S rRNA sequencing. Our results demonstrate an altered mycobiome in RRMS patients compared with HC. RRMS patients showed an increased abundance of Basidiomycota and decreased Ascomycota at the phylum level with an increased abundance of Candida and Epicoccum genera along with a decreased abundance of Saccharomyces compared to HC. We also observed an increased ITS2/16S ratio, altered fungal and bacterial associations, and altered fungal functional profiles in MS patients compared to HC. This study demonstrates that RRMS patients had a distinct mycobiome with associated changes in the bacterial microbiome compared to HC. There is an increased fungal to bacterial ratio as well as more diverse fungal-bacterial interactions in RRMS patients compared to HC. Our study is the first step towards future studies in delineating the mechanisms through which the fungal microbiome can influence MS disease.
Collapse
Affiliation(s)
- Meeta Yadav
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
- University of Iowa College of Dentistry, Iowa City, IA, United States of America
| | - Soham Ali
- Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Rachel L. Shrode
- Informatics Graduate Program, University of Iowa, Iowa City, IA, United States of America
| | - Shailesh K. Shahi
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Samantha N. Jensen
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States of America
| | - Jemmie Hoang
- College of Nursing University of Iowa, Iowa City, IA, United States of America
| | - Samuel Cassidy
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States of America
| | - Heena Olalde
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States of America
| | - Natalya Guseva
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Mishelle Paullus
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States of America
| | - Catherine Cherwin
- College of Nursing University of Iowa, Iowa City, IA, United States of America
| | - Kai Wang
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, United States of America
| | - Tracey Cho
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States of America
| | - John Kamholz
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States of America
| | - Ashutosh K. Mangalam
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
- Informatics Graduate Program, University of Iowa, Iowa City, IA, United States of America
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States of America
- Iowa City VA Health System, Iowa City, IA, United States of America
| |
Collapse
|
55
|
Archambault LS, Dongari-Bagtzoglou A. Probiotics for Oral Candidiasis: Critical Appraisal of the Evidence and a Path Forward. FRONTIERS IN ORAL HEALTH 2022; 3:880746. [PMID: 35495563 PMCID: PMC9046664 DOI: 10.3389/froh.2022.880746] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/23/2022] [Indexed: 11/29/2022] Open
Abstract
Oropharyngeal Candidiasis (OPC) is a mucosal fungal infection that is prevalent among patients with compromised immunity. The success of probiotics in treating chronic diseases with a microbial etiology component at other mucosal sites (i.e., gastro-intestinal, genitourinary and alveolar mucosae) has inspired research into the use of probiotics in the treatment of OPC. A growing body of research in vitro and in animal models indicates that some probiotic species and strains have inhibitory activities against Candida albicans growth, morphological switching, and biofilm formation. However, recent review and meta-analysis studies reveal a dearth of human randomized, controlled clinical trials on the efficacy of probiotics to treat or prevent OPC, while the majority of these have not based their selection of probiotic strains or the type of administration on sound pre-clinical evidence. In this mini-review, we assess the state of the field, outline some of the difficulties in translating lab results to clinical efficacy, and make recommendations for future research needed in order to move the field forward.
Collapse
Affiliation(s)
- Linda S. Archambault
- Department of Craniofacial Sciences, University of Connecticut Health Center, Farmington, CT, United States
- Center for Quantitative Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Anna Dongari-Bagtzoglou
- Department of Craniofacial Sciences, University of Connecticut Health Center, Farmington, CT, United States
- *Correspondence: Anna Dongari-Bagtzoglou
| |
Collapse
|
56
|
Hwang G. In it together: Candida-bacterial oral biofilms and therapeutic strategies. ENVIRONMENTAL MICROBIOLOGY REPORTS 2022; 14:183-196. [PMID: 35218311 PMCID: PMC8957517 DOI: 10.1111/1758-2229.13053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 05/16/2023]
Abstract
Under natural environmental settings or in the human body, the majority of microorganisms exist in complex polymicrobial biofilms adhered to abiotic and biotic surfaces. These microorganisms exhibit symbiotic, mutualistic, synergistic, or antagonistic relationships with other species during biofilm colonization and development. These polymicrobial interactions are heterogeneous, complex and hard to control, thereby often yielding worse outcomes than monospecies infections. Concerning fungi, Candida spp., in particular, Candida albicans is often detected with various bacterial species in oral biofilms. These Candida-bacterial interactions may induce the transition of C. albicans from commensal to pathobiont or dysbiotic organism. Consequently, Candida-bacterial interactions are largely associated with various oral diseases, including dental caries, denture stomatitis, periodontitis, peri-implantitis, and oral cancer. Given the severity of oral diseases caused by cross-kingdom consortia that develop hard-to-remove and highly drug-resistant biofilms, fundamental research is warranted to strategically develop cost-effective and safe therapies to prevent and treat cross-kingdom interactions and subsequent biofilm development. While studies have shed some light, targeting fungal-involved polymicrobial biofilms has been limited. This mini-review outlines the key features of Candida-bacterial interactions and their impact on various oral diseases. In addition, current knowledge on therapeutic strategies to target Candida-bacterial polymicrobial biofilms is discussed.
Collapse
Affiliation(s)
- Geelsu Hwang
- Department of Preventive and Restorative Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
- Corresponding Author: Geelsu Hwang,
| |
Collapse
|
57
|
Souza JG, Costa RC, Sampaio AA, Abdo VL, Nagay BE, Castro N, Retamal-Valdes B, Shibli JA, Feres M, Barão VA, Bertolini M. Cross-kingdom microbial interactions in dental implant-related infections: is Candida albicans a new villain? iScience 2022; 25:103994. [PMID: 35313695 PMCID: PMC8933675 DOI: 10.1016/j.isci.2022.103994] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Candida albicans, an oral fungal opportunistic pathogen, has shown the ability to colonize implant surfaces and has been frequently isolated from biofilms associated with dental implant-related infections, possibly due to its synergistic interactions with certain oral bacteria. Moreover, evidence suggests that this cross-kingdom interaction on implant can encourage bacterial growth, leading to increased fungal virulence and mucosal damage. However, the role of Candida in implant-related infections has been overlooked and not widely explored or even considered by most microbiological analyses and therapeutic approaches. Thus, we summarized the scientific evidence regarding the ability of C. albicans to colonize implant surfaces, interact in implant-related polymicrobial biofilms, and its possible role in peri-implant infections as far as biologic plausibility. Next, a systematic review of preclinical and clinical studies was conducted to identify the relevance and the gap in the existing literature regarding the role of C. albicans in the pathogenesis of peri-implant infections.
Collapse
Affiliation(s)
- João G.S. Souza
- Department of Periodontology, Dental Research Division, Guarulhos University, Guarulhos, Sāo Paulo 07023-070, Brazil
- Dental Science School (Faculdade de Ciências Odontológicas - FCO), Montes Claros, Minas Gerais 39401-303, Brazil
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo 13414-903, Brazil
- Corresponding author
| | - Raphael C. Costa
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo 13414-903, Brazil
| | - Aline A. Sampaio
- Department of Clinic, Pathology and Dental Surgery, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Victória L. Abdo
- Department of Periodontology, Dental Research Division, Guarulhos University, Guarulhos, Sāo Paulo 07023-070, Brazil
| | - Bruna E. Nagay
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo 13414-903, Brazil
| | - Nidia Castro
- Department of Periodontology, Dental Research Division, Guarulhos University, Guarulhos, Sāo Paulo 07023-070, Brazil
| | - Belén Retamal-Valdes
- Department of Periodontology, Dental Research Division, Guarulhos University, Guarulhos, Sāo Paulo 07023-070, Brazil
| | - Jamil A. Shibli
- Department of Periodontology, Dental Research Division, Guarulhos University, Guarulhos, Sāo Paulo 07023-070, Brazil
| | - Magda Feres
- Department of Periodontology, Dental Research Division, Guarulhos University, Guarulhos, Sāo Paulo 07023-070, Brazil
| | - Valentim A.R. Barão
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo 13414-903, Brazil
- Corresponding author
| | - Martinna Bertolini
- Department of Periodontics and Preventive Dentistry, School of Dental Medicine, University of Pittsburgh, Pennsylvania 15260, USA
| |
Collapse
|
58
|
Tripathi A, Nahar A, Sharma R, Kanaskie T, Al-Hebshi N, Puri S. High iron-mediated increased oral fungal burden, oral-to-gut transmission, and changes to pathogenicity of Candida albicans in oropharyngeal candidiasis. J Oral Microbiol 2022; 14:2044110. [PMID: 35251523 PMCID: PMC8896197 DOI: 10.1080/20002297.2022.2044110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Background Aim Methods Results Conclusion
Collapse
Affiliation(s)
- Aparna Tripathi
- Oral Microbiome Research Laboratory, Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania, USA
| | - Anubhav Nahar
- Oral Microbiome Research Laboratory, Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania, USA
| | - Rishabh Sharma
- Oral Microbiome Research Laboratory, Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania, USA
| | - Trevor Kanaskie
- Oral Microbiome Research Laboratory, Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania, USA
| | - Nezar Al-Hebshi
- Oral Microbiome Research Laboratory, Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania, USA
| | - Sumant Puri
- Oral Microbiome Research Laboratory, Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
59
|
Hu W, Huang L, Zhou Z, Yin L, Tang J. Diallyl Disulfide (DADS) Ameliorates Intestinal Candida albicans Infection by Modulating the Gut microbiota and Metabolites and Providing Intestinal Protection in Mice. Front Cell Infect Microbiol 2022; 11:743454. [PMID: 35071031 PMCID: PMC8777027 DOI: 10.3389/fcimb.2021.743454] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Diallyl disulfide (DADS), a garlic extract also known as allicin, has been reported to have numerous biological activities, including anticancer, antifungal, and inflammation-inhibiting activities, among others. Although many studies have assessed whether DADS can treat Candida albicans infection in vitro, its in vivo function and the underlying mechanism are still not clear. Accumulated evidence has implicated the gut microbiota as an important factor in the colonization and invasion of C. albicans. Thus, this study aimed to identify the mechanism by which DADS ameliorates dextran sulfate (DSS)-induced intestinal C. albicans infection based on the systematic analysis of the gut microbiota and metabolomics in mice. Here, we determined the body weight, survival, colon length, histological score, and inflammatory cytokine levels in the serum and intestines of experimental mice. Fecal samples were collected for gut microbiota and metabolite analysis by 16S rRNA gene sequencing and LC-MS metabolomics, respectively. DADS significantly alleviated DSS-induced intestinal C. albicans infection and altered the gut microbial community structure and metabolic profile in the mice. The abundances of some pathogenic bacteria, such as Proteobacteria, Escherichia-Shigella, and Streptococcus, were notably decreased after treatment with DADS. In contrast, SCFA-producing bacteria, namely, Ruminiclostridium, Oscillibacter, and Ruminococcaceae_UCG-013, greatly increased in number. The perturbance of metabolites in infectious mice was improved by DADS, with increases in secondary bile acids, arachidonic acid, indoles and their derivatives, which were highly related to the multiple differentially altered metabolic pathways, namely, bile secretion, arachidonic acid metabolism, and tryptophan metabolism. This study indicated that DADS could modulate gut microbiota and metabolites and protect the gut barrier to alleviate DSS-induced intestinal C. albicans infection in mice. Moreover, this work might also provide novel insight into the treatment of C. albicans infection using DADS.
Collapse
Affiliation(s)
| | | | | | | | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| |
Collapse
|
60
|
Niemiec BA, Gawor J, Tang S, Prem A, Krumbeck JA. The mycobiome of the oral cavity in healthy dogs and dogs with periodontal disease. Am J Vet Res 2022; 83:42-49. [PMID: 34727047 DOI: 10.2460/ajvr.20.11.0200] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To investigate the mycobiome of the oral cavity in healthy dogs and dogs with various stages of periodontal disease. ANIMALS 51 dogs without periodontal disease (n = 12) or with mild (10), moderate (19), or severe (10) periodontal disease. PROCEDURES The whole maxillary arcade of each dog was sampled with a sterile swab, and swabs were submitted for next-generation DNA sequencing targeting the internal transcribed spacer 2 region with a commercial sequencing platform. RESULTS Fungi were detected in all samples, with a total of 320 fungal species from 135 families detected in the data set. No single fungal species was found in all samples. The 3 most frequently found fungal species were Cladosporium sp (46/51 samples), Malassezia restricta (44/51 samples), and Malassezia arunalokei (36/51 samples). Certain fungi, specifically those of the family Didymellaceae, the family Irpicaceae, and the order Pleosporales, were significantly associated with different stages of periodontitis. Mycobial analysis indicated that Cladosporium sp could be considered part of the core oral cavity mycobiome. CONCLUSIONS AND CLINICAL RELEVANCE Results highlighted that fungi are present in the oral cavity of dogs and are characterized by substantial species diversity, with different fungal communities associated with various stages of periodontal disease. The next-generation DNA sequencing used in the present study revealed substantially more species of fungi than previous culture-based studies.
Collapse
|
61
|
Jiang W, Deng Z, Dai X, Zhao W. PANoptosis: A New Insight Into Oral Infectious Diseases. Front Immunol 2022; 12:789610. [PMID: 34970269 PMCID: PMC8712492 DOI: 10.3389/fimmu.2021.789610] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/25/2021] [Indexed: 12/11/2022] Open
Abstract
The oral microbiome, one of the most complex and intensive microbial ecosystems in the human body, comprises bacteria, archaea, fungi, protozoa, and viruses. Dysbiosis of the oral microbiome is the initiating factor that leads to oral infectious diseases. Infection is a sophisticated biological process involving interplay between the pathogen and the host, which often leads to activation of programmed cell death. Studies suggest that pyroptosis, apoptosis, and necroptosis are involved in multiple oral infectious diseases. Further understanding of crosstalk between cell death pathways has led to pyroptosis, apoptosis, and necroptosis being integrated into a single term: PANoptosis. PANoptosis is a multifaceted agent of the immune response that has important pathophysiological relevance to infectious diseases, autoimmunity, and cancer. As such, it plays an important role in innate immune cells that detect and eliminate intracellular pathogens. In addition to the classical model of influenza virus-infected and Yersinia-infected macrophages, other studies have expanded the scope of PANoptosis to include other microorganisms, as well as potential roles in cell types other than macrophages. In this review, we will summarize the pathophysiological mechanisms underlying inflammation and tissue destruction caused by oral pathogens. We present an overview of different pathogens that may induce activation of PANoptosis, along with the functional consequences of PANoptosis in the context of oral infectious diseases. To advance our understanding of immunology, we also explore the strategies used by microbes that enable immune evasion and replication within host cells. Improved understanding of the interplay between the host and pathogen through PANoptosis will direct development of therapeutic strategies that target oral infectious diseases.
Collapse
Affiliation(s)
- Weiyi Jiang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zilong Deng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xingzhu Dai
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wanghong Zhao
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
62
|
Wang B, He X, Lu F, Li Y, Wang Y, Zhang M, Huang Y, Xia J. Candida Isolates From Blood and Other Normally Sterile Foci From ICU Patients: Determination of Epidemiology, Antifungal Susceptibility Profile and Evaluation of Associated Risk Factors. Front Public Health 2021; 9:779590. [PMID: 34858938 PMCID: PMC8632017 DOI: 10.3389/fpubh.2021.779590] [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: 09/19/2021] [Accepted: 10/19/2021] [Indexed: 12/29/2022] Open
Abstract
Background: The clinical diagnosis and therapy for ICU patients with invasive candidiasis are challenged by the changes of Candida community composition and antimicrobial resistance. The epidemiology and drug sensitivity of candidiasis in ICU as well as its risk factors and drug resistance mechanism were investigated. Methods: In the present study, 115 patients in ICU were recruited from June 2019 through July 2020. Among them, 83 Candida isolates were identified with MALDI-TOF mass spectrometry. The susceptibility to antifungals was measured by microdilution method. The molecular mechanisms of azole-resistant Candida tropicalis were explored by sequencing, and their outcomes were explicitly documented. Results: Candida glabrata and C. tropicalis were the predominant non-C. albicans Candida. The specimen sources were mainly urine, bronchoalveolar lavage fluid and blood. The age, length of hospitalization, tracheotomy, diabetes and concomitant bacterial infection were the main risk factors for candidiasis. The majority of Candida species exhibited susceptibility to antifungals. However, certain C. tropicalis were frequently resistant to azoles. The polymorphism of the ERG11 in C. tropicalis was likely associated with azole resistance. Conclusion: The multiple risk factors for candidiasis in ICU patients need to be considered. Certain C. tropicalis exhibit resistance to azoles likely due to the ERG11 gene polymorphism.
Collapse
Affiliation(s)
- Bo Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xinlong He
- Department of Pathogen Biology, School of Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental and Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
| | - Feng Lu
- Department of Pathogen Biology, School of Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental and Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
| | - Yajuan Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuerong Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Min Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ying Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jinxing Xia
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
63
|
Fu Y, Wang W, Zeng Q, Wang T, Qian W. Antibiofilm Efficacy of Luteolin Against Single and Dual Species of Candida albicans and Enterococcus faecalis. Front Microbiol 2021; 12:715156. [PMID: 34721318 PMCID: PMC8555412 DOI: 10.3389/fmicb.2021.715156] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/08/2021] [Indexed: 12/04/2022] Open
Abstract
Candida albicans and Enterococcus faecalis biofilm-associated infections have been a huge challenge to the medical community. However, the efficacy of natural products against mixed biofilms of C. albicans and E. faecalis still remains largely unexploited. The aim of this study was to evaluate the efficacy of luteolin against planktonic cell growth, adhesion, and biofilm formation of C. albicans and E. faecalis in single and mixed cultures in vitro. The results showed that the minimum inhibitory concentrations of luteolin against planktonic cells of C. albicans, E. faecalis, and mixed cultures were 32 and 64 μg ml–1, respectively. The results displayed that a remarkable variation in biofilm biomass, viability, structure, and composition of single and dual-species biofilms formed by mono- and dual-species biofilms of C. albicans and E. faecalis in the presence of luteolin was confirmed by mainly crystal violet staining assay (CVSA), optical microscope, field emission scanning electron microscope (FESEM), and confocal laser scanning microscope (CLSM). The tolerance of luteolin-treated single- and dual-species biofilms to antibiotics was found to obviously decrease, and the loss of biofilm matrix components (mainly polysaccharides and proteins) was revealed by CLSM. Moreover, luteolin was effective at inactivating biofilm cells, as well as destructing preformed biofilm structures by single and dual species by CVSA, FESEM, and CLSM. Collectively, these data indicate the potential of luteolin as a promising antibiofilm agent for the therapeutic management of biofilm-related infections induced by single and dual species of C. albicans and E. faecalis.
Collapse
Affiliation(s)
- Yuting Fu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Wenjing Wang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Qiao Zeng
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Ting Wang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Weidong Qian
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| |
Collapse
|
64
|
Crosstalk between the oral microbiota, mucosal immunity, and the epithelial barrier regulates oral mucosal disease pathogenesis. Mucosal Immunol 2021; 14:1247-1258. [PMID: 34040155 DOI: 10.1038/s41385-021-00413-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 04/26/2021] [Accepted: 05/04/2021] [Indexed: 02/07/2023]
Abstract
Oral mucosal disease (OMD), which is also called soft tissue oral disease, is described as a series of disorders or conditions affecting the mucosa and soft tissue in the oral cavity. Its etiology is unclear, but emerging evidence has implicated the influence of the composition of the oral mucosa and saliva-resident microbiota. In turn, this dysbiosis effects the immune response balance and epithelial barrier function, followed by the occurrence and progression of OMD. In addition, oral microbial dysbiosis is diverse in different types of diseases and different disease progressions, suggesting that key causal pathogens may exist in various oral pathologies. This narrative literature review primarily discusses the most recent findings focusing on how microbial dysbiosis communicates with mucosal adaptive immune cells and the epithelial barrier in the context of five representative OMDs, including oral candidiasis (OC), oral lichen planus (OLP), recurrent aphthous ulcer (RAU), oral leukoplakia (OLK), and oral squamous cell carcinoma (OSCC), to provide new insight into the pathogenetic mechanisms of OMDs.
Collapse
|
65
|
Lyu X, Zheng H, Wang X, Zhang H, Gao L, Xun Z, Zhang Q, He X, Hua H, Yan Z, Chen F. Oral Microbiota Composition and Function Changes During Chronic Erythematous Candidiasis. Front Cell Infect Microbiol 2021; 11:691092. [PMID: 34490138 PMCID: PMC8418087 DOI: 10.3389/fcimb.2021.691092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 07/12/2021] [Indexed: 01/14/2023] Open
Abstract
Oral microbiota is constantly changing with the host state, whereas the oral microbiome of chronic erythematous candidiasis remains poorly understood. The aim of this study was to compare oral microbial signatures and functional profiling between chronic erythematous candidiasis and healthy subjects. Using shotgun metagenomic sequencing, we analyzed the microbiome in 12 chronic erythematous candidiasis, 12 healthy subjects, and 2 chronic erythematous candidiasis cured by antifungal therapy. We found that the salivary microbiota of chronic erythematous candidiasis was significantly different from that of healthy subjects. Among them, Rothia mucilaginosa and Streptococcus mitis were the most abundant disease-enriched species (Mann-Whitney U-test, P < 0.05). In addition, co-occurrence network analysis showed that C. albicans formed densely connected modules with oral bacterial species and was mainly positive connected to Streptococcus species. Furthermore, we investigated the functional potentials of the microbiome and identified a set of microbial marker genes associated with chronic erythematous candidiasis. Some of these genes enriching in chronic erythematous candidiasis are involved in eukaryotic ribosome, putative glutamine transport system, and cytochrome bc1 complex respiratory unit. Altogether, this study revealed the changes of oral microbial composition, the co-occurrence between C. albicans and oral bacteria, as well as the changes of microbial marker genes during chronic erythematous candidiasis, which provides evidence of oral microbiome as a target for the treatment and prevention of chronic erythematous candidiasis.
Collapse
Affiliation(s)
- Xin Lyu
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Hui Zheng
- Central Laboratory, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Xu Wang
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Heyu Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Lu Gao
- Central Laboratory, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Zhe Xun
- Central Laboratory, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Qian Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Xuesong He
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, United States
| | - Hong Hua
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Zhimin Yan
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Feng Chen
- Central Laboratory, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, China
| |
Collapse
|
66
|
Last A, Maurer M, S. Mosig A, S. Gresnigt M, Hube B. In vitro infection models to study fungal-host interactions. FEMS Microbiol Rev 2021; 45:fuab005. [PMID: 33524102 PMCID: PMC8498566 DOI: 10.1093/femsre/fuab005] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 01/14/2021] [Indexed: 12/14/2022] Open
Abstract
Fungal infections (mycoses) affect over a billion people per year. Approximately, two million of these infections are life-threatening, especially for patients with a compromised immune system. Fungi of the genera Aspergillus, Candida, Histoplasma and Cryptococcus are opportunistic pathogens that contribute to a substantial number of mycoses. To optimize the diagnosis and treatment of mycoses, we need to understand the complex fungal-host interplay during pathogenesis, the fungal attributes causing virulence and how the host resists infection via immunological defenses. In vitro models can be used to mimic fungal infections of various tissues and organs and the corresponding immune responses at near-physiological conditions. Furthermore, models can include fungal interactions with the host-microbiota to mimic the in vivo situation on skin and mucosal surfaces. This article reviews currently used in vitro models of fungal infections ranging from cell monolayers to microfluidic 3D organ-on-chip (OOC) platforms. We also discuss how OOC models can expand the toolbox for investigating interactions of fungi and their human hosts in the future.
Collapse
Affiliation(s)
- Antonia Last
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knoell Institute, Beutenbergstrasse 11a, 07745, Jena, Germany
| | - Michelle Maurer
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
- Institute of Biochemistry II, Jena University Hospital, Nonnenplan 2,07743, Jena, Germany
| | - Alexander S. Mosig
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
- Institute of Biochemistry II, Jena University Hospital, Nonnenplan 2,07743, Jena, Germany
| | - Mark S. Gresnigt
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knoell Institute, Beutenbergstrasse 11a, 07745, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knoell Institute, Beutenbergstrasse 11a, 07745, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Neugasse 24, 07743, Jena, Germany
| |
Collapse
|
67
|
Villar CC, Dongari-Bagtzoglou A. Fungal diseases: Oral dysbiosis in susceptible hosts. Periodontol 2000 2021; 87:166-180. [PMID: 34463992 DOI: 10.1111/prd.12378] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The oral cavity is colonized by a large number of microorganisms that are referred to collectively as the oral microbiota. These indigenous microorganisms have evolved in symbiotic relationships with the oral mucosal immune system and are involved in maintaining homeostasis in the oral cavity. Although Candida species are commonly found in the healthy oral cavity without causing infection, these fungi can become pathogenic. Recents advances indicate that the development of oral candidiasis is driven both by Candida albicans overgrowth in a dysbiotic microbiome and by disturbances in the host's immune system. Perturbation of the oral microbiota triggered by host-extrinsic (ie, medications), host-intrinsic (ie, host genetics), and microbiome-intrinsic (ie, microbial interactions) factors may increase the risk of oral candidiasis. In this review, we provide an overview of the oral mycobiome, with a particular focus on the interactions of Candida albicans with some of the most common oral bacteria and the oral mucosal immune system. Also, we present a summary of our current knowledge of the host-intrinsic and host-extrinsic factors that can predispose to oral candidiasis.
Collapse
Affiliation(s)
- Cristina Cunha Villar
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Anna Dongari-Bagtzoglou
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, Farmington, CT, USA
| |
Collapse
|
68
|
Bertolini M, Vazquez Munoz R, Archambault L, Shah S, Souza JGS, Costa RC, Thompson A, Zhou Y, Sobue T, Dongari-Bagtzoglou A. Mucosal Bacteria Modulate Candida albicans Virulence in Oropharyngeal Candidiasis. mBio 2021; 12:e0193721. [PMID: 34399623 PMCID: PMC8406182 DOI: 10.1128/mbio.01937-21] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/09/2021] [Indexed: 01/12/2023] Open
Abstract
Oropharyngeal candidiasis (OPC) is the most prevalent oral infection in immunocompromised patients, primarily associated with Candida albicans. Increasing evidence points to a significant role of mucosal bacteria on the transition of C. albicans from commensal to pathogenic. In this work, we hypothesized that changes in the abundance or composition of the mucosal bacterial microbiota induced by dietary sucrose during the development of OPC can modulate C. albicans virulence. C. albicans burdens and mucosal lesions were evaluated in a mouse cortisone immunosuppression model amended with sucrose. We also analyzed the mucosal bacterial composition using 16S rRNA gene sequencing and culture methods. In immunocompetent mice, sucrose significantly increased total bacterial burdens and reduced alpha diversity, by increasing the relative abundance of mitis group streptococci. In immunocompromised mice, C. albicans infection was associated with a significantly reduced bacterial alpha diversity due to an increase in the relative abundance of enterococci. When exposed to dietary sucrose, these mice had reduced C. albicans burdens and reduced bacterial alpha diversity, associated with an increase in the relative abundance of Lactobacillus. SparCC correlation networks showed a significant negative correlation between Lactobacillus and Enterococcus in all Candida-infected mice. Depletion of lactobacilli with antibiotic treatment partially restored C. albicans burdens in mice receiving sucrose. In coculture in vitro experiments, mouse oral Lactobacillus johnsonii isolates inhibited growth of Enterococcus faecalis isolates and C. albicans. These results support the hypothesis that the sucrose-induced attenuation of C. albicans virulence was a result of changes in the mucosal bacterial microbiome characterized by a reduction in enterococci and an increase in lactobacilli. IMPORTANCE By comparing Candida albicans virulence and the mucosal bacterial composition in a mouse oral infection model, we were able to dissect the effects of the host environment (immunosuppression), infection with C. albicans, and local modulating factors (availability of sucrose as a carbon source) on the mucosal bacterial microbiome and its role on fungal virulence. We showed that changes in endogenous microbial communities in response to sucrose can lead to attenuation of fungal disease. We also showed that Lactobacillus johnsonii may curtail Candida virulence both by inhibiting its growth and by inhibiting the growth of potentially synergistic bacteria such as enterococci. Our results support the concept that Candida pathogenesis should be viewed in the contexts of both a susceptible host and a mucosal bacterial microbiota conducive to virulence.
Collapse
Affiliation(s)
- M. Bertolini
- Department of Oral Health and Diagnostic Sciences, UConn Health, Farmington, Connecticut, USA
| | - R. Vazquez Munoz
- Department of Oral Health and Diagnostic Sciences, UConn Health, Farmington, Connecticut, USA
| | - L. Archambault
- Department of Oral Health and Diagnostic Sciences, UConn Health, Farmington, Connecticut, USA
| | - S. Shah
- Department of Computer Science and Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - J. G. S. Souza
- Dental Research Division, Guarulhos University, Guarulhos, SP, Brazil
- Dental Science School (Faculdade de Ciências Odontológicas [FCO]), Montes Claros, MG, Brazil
| | - R. C. Costa
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, SP, Brazil
| | - A. Thompson
- Department of Oral Health and Diagnostic Sciences, UConn Health, Farmington, Connecticut, USA
| | - Y. Zhou
- Department of Medicine, UConn Health, Connecticut, Farmington, Connecticut, USA
| | - T. Sobue
- Department of Oral Health and Diagnostic Sciences, UConn Health, Farmington, Connecticut, USA
| | - A. Dongari-Bagtzoglou
- Department of Oral Health and Diagnostic Sciences, UConn Health, Farmington, Connecticut, USA
| |
Collapse
|
69
|
Vazquez-Munoz R, Dongari-Bagtzoglou A. Anticandidal Activities by Lactobacillus Species: An Update on Mechanisms of Action. FRONTIERS IN ORAL HEALTH 2021; 2:689382. [PMID: 35048033 PMCID: PMC8757823 DOI: 10.3389/froh.2021.689382] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022] Open
Abstract
Lactobacilli are among the most studied bacteria in the microbiome of the orodigestive and genitourinary tracts. As probiotics, lactobacilli may provide various benefits to the host. These benefits include regulating the composition of the resident microbiota, preventing - or even potentially reverting- a dysbiotic state. Candida albicans is an opportunistic pathogen that can influence and be influenced by other members of the mucosal microbiota and, under immune-compromising conditions, can cause disease. Lactobacillus and Candida species can colonize the same mucosal sites; however, certain Lactobacillus species display antifungal activities that can contribute to low Candida burdens and prevent fungal infection. Lactobacilli can produce metabolites with direct anticandidal function or enhance the host defense mechanisms against fungi. Most of the Lactobacillus spp. anticandidal mechanisms of action remain underexplored. This work aims to comprehensively review and provide an update on the current knowledge regarding these anticandidal mechanisms.
Collapse
Affiliation(s)
- Roberto Vazquez-Munoz
- Department of Oral Health and Diagnostic Sciences, University of Connecticut Health Center, Farmington, CT, United States
| | - Anna Dongari-Bagtzoglou
- Department of Oral Health and Diagnostic Sciences, University of Connecticut Health Center, Farmington, CT, United States
| |
Collapse
|
70
|
Risky Business: Oropharyngeal Bacteria Linked to Risk for Invasive Fungal Infection. Infect Immun 2021; 89:e0017421. [PMID: 33972370 DOI: 10.1128/iai.00174-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This work combines a clinical investigation with a mouse model of fungal infection to study the role of bacterial microbiota in fungal invasion. The investigators identified a dysbiosis in the oropharyngeal mucosa that was associated with a high risk for invasive infection in hematologic oncology patients. This study builds on our current understanding that the pathogenesis of fungal infections has to be studied in the context of a specific host background and a site-specific bacterial microbiota.
Collapse
|
71
|
Pharyngeal Microbial Signatures Are Predictive of the Risk of Fungal Pneumonia in Hematologic Patients. Infect Immun 2021; 89:e0010521. [PMID: 33782152 DOI: 10.1128/iai.00105-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The ability to predict invasive fungal infections (IFI) in patients with hematological malignancies is fundamental for successful therapy. Although gut dysbiosis is known to occur in hematological patients, whether airway dysbiosis also contributes to the risk of IFI has not been investigated. Nasal and oropharyngeal swabs were collected for functional microbiota characterization in 173 patients with hematological malignancies recruited in a multicenter, prospective, observational study and stratified according to the risk of developing IFI. A lower microbial richness and evenness were found in the pharyngeal microbiota of high-risk patients that were associated with a distinct taxonomic and metabolic profile. A murine model of IFI provided biologic plausibility for the finding that loss of protective anaerobes, such as Clostridiales and Bacteroidetes, along with an apparent restricted availability of tryptophan, is causally linked to the risk of IFI in hematologic patients and indicates avenues for antimicrobial stewardship and metabolic reequilibrium in IFI.
Collapse
|
72
|
Fusco A, Savio V, Donniacuo M, Perfetto B, Donnarumma G. Antimicrobial Peptides Human Beta-Defensin-2 and -3 Protect the Gut During Candida albicans Infections Enhancing the Intestinal Barrier Integrity: In Vitro Study. Front Cell Infect Microbiol 2021; 11:666900. [PMID: 34178720 PMCID: PMC8223513 DOI: 10.3389/fcimb.2021.666900] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022] Open
Abstract
The intestinal mucosa is composed of a monolayer of epithelial cells, which is highly polarized and firmly united to each other thanks to the presence of proteins complexes, called Tight junctions (TJs). Alteration of the mucus layer and TJs causes an increase in intestinal permeability, which can lead to a microbial translocation and systemic disorders. Candida albicans, in addition to its role of commensal, is an opportunistic pathogen responsible for disseminated candidiasis, especially in immunocompromised subjects where the dysbiosis leads to damage of the intestinal mucosal barrier . In this work, we used a line of intestinal epithelial cells able to stably express the genes that encodes human beta defensin-2 (HBD-2) and -3 (HBD-3) to monitor the invasion of C. albicans in vitro. Defensins are a group of antimicrobial peptides (AMPs) found in different living organisms, and are involved in the first line of defense in the innate immune response against pathogens. The results obtained show that the presence of antimicrobial peptides improves the expression of TJs and increases the Trans Epithelial Electrical Resistence value. In addition, the invasive ability of C. albicans in transfected cells is significantly reduced, as well as the expression levels of genes involved in the apoptotic pathway. Through the study of interaction between antimicrobial peptides and microbiota we will be able in the future to better understand the mechanisms by which they exert the host defense function against intestinal pathogens.
Collapse
Affiliation(s)
- Alessandra Fusco
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Vittoria Savio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Donniacuo
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Brunella Perfetto
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giovanna Donnarumma
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
73
|
Maia CMDA, Pasetto S, Nonaka CFW, Costa EMMDB, Murata RM. Yeast-Host Interactions: Anadenanthera colubrina Modulates Virulence Factors of C. albicans and Inflammatory Response In Vitro. Front Pharmacol 2021; 12:629778. [PMID: 34168555 PMCID: PMC8217765 DOI: 10.3389/fphar.2021.629778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 05/25/2021] [Indexed: 01/08/2023] Open
Abstract
Oral candidiasis is one of the most common fungal infections in humans. Its incidence has increased widely, as well as the antifungal resistance, demanding for the search for novel antifungal therapeutic agents. Anadenanthera colubrina (Vell.) Brenan is a plant species that has been proven to possess pharmacological effects, including antifungal and anti-inflammatory activities. This study evaluated in vitro the effects of standardized A. colubrina extract on virulence factors of Candida albicans and its regulation on immune response through C. albicans-host interaction. Antifungal activity was evaluated by Broth Microdilution Method against reference Candida strains (C. albicans, C. glabrata, C. tropicalis; C. dubliniensis). Anti-biofilm effect was performed on C. albicans mature biofilm and quantified by CFU/mL/g of biofilm dry weight. Proleotlytic enzymatic activities of proteinase and phospholipase were assessed by Azocasein and Phosphatidylcholine assays, respectively. Cytotoxicity effect was determined by Cell Titer Blue Viability Assay on Human Gingival Fibroblasts. Co-cultured model was used to analyze C. albicans coexisting with HGF by Scanning Electron Microscopy and fluorescence microscopies; gene expression was assessed by RT-PCR of C. albicans enzymes (SAP-1, PLB-1) and of host inflammatory cytokines (IL-6, IL-8, IL-1β, IL-10). Cytokines secretion was analysed by Luminex. The extract presented antifungal effect with MIC<15.62 μg/ml against Candida strains. Biofilm and proteolytic activity were significant reduced at 312.4 μg/ml (20 × 15.62 μg/ml) extract concentration. Cell viability was maintained higher than 70% in concentrations up to 250 μg/ml (LD50 = 423.3 μg/ml). Co-culture microscopies demonstrated a substantial decreased in C. albicans growth and minimal toxicity against host cells. Gene expressions of SAP-1/PLB-1 were significantly down-regulated and host immune response was modulated by a significant decreased on IL-6 and IL-8 cytokines secretion. A. colubrina had antifungal activity on Candida strains, antibiofilm, and anti-proteolytic enzyme effects against C. albicans. Presented low cytotoxicity to the host cells and modulatory effects on the host immune response.
Collapse
Affiliation(s)
- Carolina Medeiros de Almeida Maia
- Department of Dentistry, Postgraduate Program in Dentistry, State University of Paraiba, Campina Grande, Brazil
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, NC, United States
| | - Silvana Pasetto
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, NC, United States
| | | | | | - Ramiro Mendonça Murata
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, NC, United States
| |
Collapse
|
74
|
Herrera G, Vega L, Patarroyo MA, Ramírez JD, Muñoz M. Gut microbiota composition in health-care facility-and community-onset diarrheic patients with Clostridioides difficile infection. Sci Rep 2021; 11:10849. [PMID: 34035404 PMCID: PMC8149855 DOI: 10.1038/s41598-021-90380-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 05/04/2021] [Indexed: 02/04/2023] Open
Abstract
The role of gut microbiota in the establishment and development of Clostridioides difficile infection (CDI) has been widely discussed. Studies showed the impact of CDI on bacterial communities and the importance of some genera and species in recovering from and preventing infection. However, most studies have overlooked important components of the intestinal ecosystem, such as eukaryotes and archaea. We investigated the bacterial, archaea, and eukaryotic intestinal microbiota of patients with health-care-facility- or community-onset (HCFO and CO, respectively) diarrhea who were positive or negative for CDI. The CDI-positive groups (CO/+, HCFO/+) showed an increase in microorganisms belonging to Bacteroidetes, Firmicutes, Proteobacteria, Ascomycota, and Opalinata compared with the CDI-negative groups (CO/-, HCFO/-). Patients with intrahospital-acquired diarrhea (HCFO/+, HCFO/-) showed a marked decrease in bacteria beneficial to the intestine, and there was evidence of increased Archaea and Candida and Malassezia species compared with the CO groups (CO/+, CO/-). Characteristic microbiota biomarkers were established for each group. Finally, correlations between bacteria and eukaryotes indicated interactions among the different kingdoms making up the intestinal ecosystem. We showed the impact of CDI on microbiota and how it varies with where the infection is acquired, being intrahospital-acquired diarrhea one of the most influential factors in the modulation of bacterial, archaea, and eukaryotic populations. We also highlight interactions between the different kingdoms of the intestinal ecosystem, which need to be evaluated to improve our understanding of CDI pathophysiology.
Collapse
Affiliation(s)
- Giovanny Herrera
- Centro de Investigaciones en Microbiología y Biotecnología - UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Laura Vega
- Centro de Investigaciones en Microbiología y Biotecnología - UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Manuel Alfonso Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
- Microbiology Department, Faculty of Medicine, Universidad Nacional de Colombia, 111321, Bogotá D.C., Colombia
- Health Sciences Division, Main Campus, Universidad Santo Tomás, 110231, Bogotá D.C., Colombia
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología - UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Marina Muñoz
- Centro de Investigaciones en Microbiología y Biotecnología - UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
75
|
d'Enfert C, Kaune AK, Alaban LR, Chakraborty S, Cole N, Delavy M, Kosmala D, Marsaux B, Fróis-Martins R, Morelli M, Rosati D, Valentine M, Xie Z, Emritloll Y, Warn PA, Bequet F, Bougnoux ME, Bornes S, Gresnigt MS, Hube B, Jacobsen ID, Legrand M, Leibundgut-Landmann S, Manichanh C, Munro CA, Netea MG, Queiroz K, Roget K, Thomas V, Thoral C, Van den Abbeele P, Walker AW, Brown AJP. The impact of the Fungus-Host-Microbiota interplay upon Candida albicans infections: current knowledge and new perspectives. FEMS Microbiol Rev 2021; 45:fuaa060. [PMID: 33232448 PMCID: PMC8100220 DOI: 10.1093/femsre/fuaa060] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Candida albicans is a major fungal pathogen of humans. It exists as a commensal in the oral cavity, gut or genital tract of most individuals, constrained by the local microbiota, epithelial barriers and immune defences. Their perturbation can lead to fungal outgrowth and the development of mucosal infections such as oropharyngeal or vulvovaginal candidiasis, and patients with compromised immunity are susceptible to life-threatening systemic infections. The importance of the interplay between fungus, host and microbiota in driving the transition from C. albicans commensalism to pathogenicity is widely appreciated. However, the complexity of these interactions, and the significant impact of fungal, host and microbiota variability upon disease severity and outcome, are less well understood. Therefore, we summarise the features of the fungus that promote infection, and how genetic variation between clinical isolates influences pathogenicity. We discuss antifungal immunity, how this differs between mucosae, and how individual variation influences a person's susceptibility to infection. Also, we describe factors that influence the composition of gut, oral and vaginal microbiotas, and how these affect fungal colonisation and antifungal immunity. We argue that a detailed understanding of these variables, which underlie fungal-host-microbiota interactions, will present opportunities for directed antifungal therapies that benefit vulnerable patients.
Collapse
Affiliation(s)
- Christophe d'Enfert
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Ann-Kristin Kaune
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Leovigildo-Rey Alaban
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Sayoni Chakraborty
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Neugasse 25, 07743 Jena, Germany
| | - Nathaniel Cole
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Margot Delavy
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Daria Kosmala
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Benoît Marsaux
- ProDigest BV, Technologiepark 94, B-9052 Gent, Belgium
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links, 9000 Ghent, Belgium
| | - Ricardo Fróis-Martins
- Immunology Section, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, Zurich 8057, Switzerland
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Moran Morelli
- Mimetas, Biopartner Building 2, J.H. Oortweg 19, 2333 CH Leiden, The Netherlands
| | - Diletta Rosati
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Marisa Valentine
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Zixuan Xie
- Gut Microbiome Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Yoan Emritloll
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Peter A Warn
- Magic Bullet Consulting, Biddlecombe House, Ugbrook, Chudleigh Devon, TQ130AD, UK
| | - Frédéric Bequet
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
| | - Marie-Elisabeth Bougnoux
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Stephanie Bornes
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF0545, 20 Côte de Reyne, 15000 Aurillac, France
| | - Mark S Gresnigt
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Bernhard Hube
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Ilse D Jacobsen
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Mélanie Legrand
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Salomé Leibundgut-Landmann
- Immunology Section, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, Zurich 8057, Switzerland
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Chaysavanh Manichanh
- Gut Microbiome Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Carol A Munro
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Karla Queiroz
- Mimetas, Biopartner Building 2, J.H. Oortweg 19, 2333 CH Leiden, The Netherlands
| | - Karine Roget
- NEXBIOME Therapeutics, 22 allée Alan Turing, 63000 Clermont-Ferrand, France
| | - Vincent Thomas
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
| | - Claudia Thoral
- NEXBIOME Therapeutics, 22 allée Alan Turing, 63000 Clermont-Ferrand, France
| | | | - Alan W Walker
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Alistair J P Brown
- MRC Centre for Medical Mycology, Department of Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| |
Collapse
|
76
|
Alterations in the gut microbiota and metabolic profiles coincide with intestinal damage in mice with a bloodborne Candida albicans infection. Microb Pathog 2021; 154:104826. [PMID: 33689815 DOI: 10.1016/j.micpath.2021.104826] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 12/25/2022]
Abstract
Candida albicans is an opportunistic fungus that can threaten life especially in patients with candidemia. The morbidity and mortality of candidemia originating from a central venous catheter (CVC) and illicit intravenous drug use (IVDU) are increasing. However, the mechanism underlying the bloodborne C. albicans infection remains unclear. Herein, we evaluated the gut microbiome, metabolites and intestinal mucosa by constructing the mouse models with candidemia. Model mice were injected with C. albicans via tail vein. Control mice underwent sham procedures. We observed basic life characteristics, intestinal damage-related alterations using hematoxylin and eosin (H&E) staining, intestinal tight junction protein levels, and intestinal permeability in these mice. Fecal samples were analyzed by performing 16S rRNA gene sequencing of the microbiota and LC-MS metabolomics to reveal the perturbations in intestinal flora and metabolism exacerbating intestinal damage. Weight loss, a decreased survival rate, C. albicans infection spread, and colonic epithelial damage occurred in the model group. Furthermore, the intestinal flora abundance was reduced. Several probiotics, such as Lactobacillus, and butyrate-producing bacteria, including Roseburia, Lachnospiraceae, and Clostridia, were depleted, and some pathogenic bacteria, such as Escherichia-Shigella and Proteus, belonging to the Proteobacteria phylum, and the inflammation mediators Ruminococcus and Parabacteroides were enriched in model mice. Multiple differentially altered metabolic pathways were observed and mainly related to bile acid, arachidonic acid, bile secretion, and arachidonic acid metabolism. This study illustrated the effects of a bloodborne C. albicans on the intestinal microbiota, metabolites, and intestinal barrier, which may provide new insights into tests or treatments for candidemia originating from CVC or IVDU.
Collapse
|
77
|
Zhong M, Xiong Y, Zhao J, Gao Z, Ma J, Wu Z, Song Y, Hong X. Candida albicans disorder is associated with gastric carcinogenesis. Theranostics 2021; 11:4945-4956. [PMID: 33754037 PMCID: PMC7978306 DOI: 10.7150/thno.55209] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/29/2021] [Indexed: 12/18/2022] Open
Abstract
Background: Bacterial infection is associated with gastric carcinogenesis. However, the relationship between nonbacterial components and gastric cancer (GC) has not been fully explored. We aimed to characterize the fungal microbiome in GC. Methods: We performed ITS rDNA gene analysis in cancer lesions and adjacent noncancerous tissues of 45 GC cases from Shenyang, China. Obtaining the OTUs and combining effective grouping, we carried out species identifications, alpha and beta diversity analyses, and FUNGuild functional annotation. Moreover, differences were compared and tested between groups to better investigate the composition and ecology of fungi associated with GC and find fungal indicators. Results: We observed significant gastric fungal imbalance in GC. Principal component analysis revealed separate clusters for the GC and control groups, and Venn diagram analysis indicated that the GC group showed a lower OTU abundance than the control. At the genus level, the abundances of 15 fungal biomarkers distinguished the GC group from the control, of which Candida (p = 0.000246) and Alternaria (p = 0.00341) were enriched in GC, while Saitozyma (p = 0.002324) and Thermomyces (p = 0.009158) were decreased. Combining the results of Welch's t test and Wilcoxon rank sum test, Candida albicans (C. albicans) was significantly elevated in GC. The species richness Krona pie chart further revealed that C. albicans occupied 22% and classified GC from the control with an area under the receiver operating curve (AUC) of 0.743. Random forest analysis also confirmed that C. albicans could serve as a biomarker with a certain degree of accuracy. Moreover, compared with that of the control, the alpha diversity index was significantly reduced in the GC group. The Jaccard distance index and the Bray abundance index of the PCoA clarified separate clusters between the GC and control groups at the species level (p = 0.00051). Adonis (PERMANOVA) analysis and ANOVA showed that there were significant differences in fungal structure among groups (p = 0.001). Finally, FUNGuild functional classification predicted that saprotrophs were the most abundant taxa in the GC group. Conclusions: This study revealed GC-associated mycobiome imbalance characterized by an altered fungal composition and ecology and demonstrated that C. albicans can be a fungal biomarker for GC. With the significant increase of C. albicans in GC, the abundance of Fusicolla acetilerea, Arcopilus aureus, Fusicolla aquaeductuum were increased, while Candida glabrata, Aspergillus montevidensis, Saitozyma podzolica and Penicillium arenicola were obviously decreased. In addition, C. albicans may mediate GC by reducing the diversity and richness of fungi in the stomach, contributing to the pathogenesis of GC.
Collapse
Affiliation(s)
- Mengya Zhong
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, Xiamen, Fujian, China
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yubo Xiong
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, Xiamen, Fujian, China
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jiabao Zhao
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, Xiamen, Fujian, China
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhi Gao
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key laboratory of Biological Targeting Diagnosis and Therapy Research, Guangxi Medical University, Nanning, Guangxi, China
| | - Jingsong Ma
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, Xiamen, Fujian, China
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhengxin Wu
- School of Medicine, Guangxi University, Nanning, Guangxi, China
| | - Yongxi Song
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xuehui Hong
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, Xiamen, Fujian, China
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
78
|
Andremont A, Cervesi J, Bandinelli PA, Vitry F, de Gunzburg J. Spare and repair the gut microbiota from antibiotic-induced dysbiosis: state-of-the-art. Drug Discov Today 2021; 26:2159-2163. [PMID: 33639249 DOI: 10.1016/j.drudis.2021.02.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/02/2021] [Accepted: 02/16/2021] [Indexed: 02/08/2023]
Abstract
Homeostasis of the intestinal microbiota is currently recognized as a major contributor to human health. Furthermore, intestinal dysbiosis is associated with a multitude of consequences, including intestinal colonization by antibiotic-resistant or pathogenic bacteria, such as Clostridioides difficile, and reduced efficacy of promising anticancer immunotherapies. By far, the most immediate and drastic exposure leading to dysbiosis is antibiotic treatment. Many attempts have been made to prevent or repair antibiotic-associated dysbiosis. Here, we review these innovations and the difficulties associated with their development.
Collapse
Affiliation(s)
- Antoine Andremont
- Da Volterra, Paris, France; Université de Paris, IAME, INSERM, Paris, France.
| | | | | | | | | |
Collapse
|
79
|
Understanding Human Microbiota Offers Novel and Promising Therapeutic Options against Candida Infections. Pathogens 2021; 10:pathogens10020183. [PMID: 33572162 PMCID: PMC7915436 DOI: 10.3390/pathogens10020183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 01/20/2021] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
Human fungal pathogens particularly of Candida species are one of the major causes of hospital acquired infections in immunocompromised patients. The limited arsenal of antifungal drugs to treat Candida infections with concomitant evolution of multidrug resistant strains further complicates the management of these infections. Therefore, deployment of novel strategies to surmount the Candida infections requires immediate attention. The human body is a dynamic ecosystem having microbiota usually involving symbionts that benefit from the host, but in turn may act as commensal organisms or affect positively (mutualism) or negatively (pathogenic) the physiology and nourishment of the host. The composition of human microbiota has garnered a lot of recent attention, and despite the common occurrence of Candida spp. within the microbiota, there is still an incomplete picture of relationships between Candida spp. and other microorganism, as well as how such associations are governed. These relationships could be important to have a more holistic understanding of the human microbiota and its connection to Candida infections. Understanding the mechanisms behind commensalism and pathogenesis is vital for the development of efficient therapeutic strategies for these Candida infections. The concept of host-microbiota crosstalk plays critical roles in human health and microbiota dysbiosis and is responsible for various pathologies. Through this review, we attempted to analyze the types of human microbiota and provide an update on the current understanding in the context of health and Candida infections. The information in this article will help as a resource for development of targeted microbial therapies such as pre-/pro-biotics and microbiota transplant that has gained advantage in recent times over antibiotics and established as novel therapeutic strategy.
Collapse
|
80
|
Pan CH, Lo HJ, Yan JY, Hsiao YJ, Hsueh JW, Lin DW, Lin TH, Wu SH, Chen YC. Candida albicans Colonizes and Disseminates to the Gastrointestinal Tract in the Presence of the Microbiota in a Severe Combined Immunodeficient Mouse Model. Front Microbiol 2021; 11:619878. [PMID: 33488563 PMCID: PMC7819875 DOI: 10.3389/fmicb.2020.619878] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 11/23/2020] [Indexed: 01/08/2023] Open
Abstract
Candida albicans is the leading cause of candidemia or other invasive candidiasis. Gastrointestinal colonization has been considered as the primary source of candidemia. However, few established mouse models that mimic this infection route are available. In the present study, we established a mouse model of disseminated candidiasis developed through the translocation of Candida from the gut. In this study, we developed a novel C. albicans GI colonization and dissemination animal model by using severe combined immunodeficient Rag2–/–IL2γc–/– (Rag2γc) mice, which lack functional T, B, NK cells, and IL2γc-dependent signaling. Rag2γc mice were highly susceptible to C. albicans gastrointestinal infection even in the presence of the gut microbiota. Within 4 weeks post infection, Rag2γc mice showed dose-dependent weight loss and disseminated candidiasis in more than 58% (7/12) of moribund mice. Histological analysis demonstrated abundant hyphae penetrating the mucosa, with significant neutrophilic infiltration in mice infected with wild-type C. albicans but not a filamentation-defective mutant. In moribund Rag2γc mice, the necrotic lesions and disrupted epithelial cells were associated with C. albicans hyphae. Notably, removal of the gut microbiota by antibiotics exacerbated the severity of fungal infection in Rag2γc mice, as demonstrated by elevated fungal burdens and accelerated weight loss and death. Furthermore, higher fungal burden and IL-1β expression were prominently noted in the stomach of Rag2γc mice. In fact, a significant increase in circulating proinflammatory cytokines, including IL-6, TNF-α, and IL-10, indicative of a septic response, was evident in infected Rag2γc mice. Additionally, Rag2γc mice exhibited significantly lower levels of IL-22 but not IFN-γ or IL-17A than wild-type B6 mice, suggesting that IL-22 plays a role in C. albicans gastrointestinal infection. Collectively, our analysis of the Rag2γc mouse model revealed features of C. albicans gastrointestinal colonization and dissemination without the interference from antibiotics or chemotherapeutic agents, thus offering a new investigative tool for delineating the pathogenesis of C. albicans and its cross-talk with the gut microbiota.
Collapse
Affiliation(s)
- Chien-Hsiung Pan
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung City, Taiwan.,Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsiu-Jung Lo
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Jia-Ying Yan
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Yu-Ju Hsiao
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Jun-Wei Hsueh
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Di-Wei Lin
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Tsung-Han Lin
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Sze-Hsien Wu
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Yee-Chun Chen
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli, Taiwan.,Department of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
81
|
Souza JGS, Bertolini MM, Costa RC, Nagay BE, Dongari-Bagtzoglou A, Barão VAR. Targeting implant-associated infections: titanium surface loaded with antimicrobial. iScience 2021; 24:102008. [PMID: 33490916 PMCID: PMC7811145 DOI: 10.1016/j.isci.2020.102008] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Implant devices have = proven a successful treatment modality in reconstructive surgeries. However, increasing rates of peri-implant diseases demand further examination of their pathogenesis. Polymicrobial biofilm formation on titanium surfaces has been considered the main risk factor for inflammatory processes on tissues surrounding implant devices, which often lead to implant failure. To overcome microbial accumulation on titanium surfaces biofilm targeting strategies have been developed to modify the surface and incorporate antimicrobial coatings. Because antibiotics are widely used to treat polymicrobial infections, these agents have recently started to be incorporated on titanium surface. This review discusses the biofilm formation on titanium dental implants and key factors to be considered in therapeutic and preventative strategies. Moreover, a systematic review was conducted on coatings developed for titanium surfaces using different antibiotics. This review will also shed light on potential alternative strategies aiming to reduce microbial loads and control polymicrobial infection on implanted devices.
Collapse
Affiliation(s)
- João Gabriel Silva Souza
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo 13414-903, Brazil
- Dental Research Division, Guarulhos University, Guarulhos, SP 07023-070, Brazil
- Dentistry Science School (Faculdade de Ciências Odontológicas - FCO), Montes Claros, Minas Gerais, 39401-303, Brazil
| | - Martinna Mendonça Bertolini
- Department of Oral Health and Diagnostic Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Raphael Cavalcante Costa
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo 13414-903, Brazil
| | - Bruna Egumi Nagay
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo 13414-903, Brazil
| | - Anna Dongari-Bagtzoglou
- Department of Oral Health and Diagnostic Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Valentim Adelino Ricardo Barão
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo 13414-903, Brazil
| |
Collapse
|
82
|
Markey L, Pugliese A, Tian T, Roy F, Lee K, Kumamoto CA. Decreased Ecological Resistance of the Gut Microbiota in Response to Clindamycin Challenge in Mice Colonized with the Fungus Candida albicans. mSphere 2021; 6:e00982-20. [PMID: 33472981 PMCID: PMC7845615 DOI: 10.1128/msphere.00982-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/18/2020] [Indexed: 02/06/2023] Open
Abstract
The mammalian gut microbiota is a complex community of microorganisms which typically exhibits remarkable stability. As the gut microbiota has been shown to affect many aspects of host health, the molecular keys to developing and maintaining a "healthy" gut microbiota are highly sought after. Yet, the qualities that define a microbiota as healthy remain elusive. We used the ability to resist change in response to antibiotic disruption, a quality we refer to as ecological resistance, as a metric for the health of the bacterial microbiota. Using a mouse model, we found that colonization with the commensal fungus Candida albicans decreased the ecological resistance of the bacterial microbiota in response to the antibiotic clindamycin such that increased microbiota disruption was observed in C. albicans-colonized mice compared to that in uncolonized mice. C. albicans colonization resulted in decreased alpha diversity and small changes in abundance of bacterial genera prior to clindamycin challenge. Strikingly, co-occurrence network analysis demonstrated that C. albicans colonization resulted in sweeping changes to the co-occurrence network structure, including decreased modularity and centrality and increased density. Thus, C. albicans colonization resulted in changes to the bacterial microbiota community and reduced its ecological resistance.IMPORTANCECandida albicans is the most common fungal member of the human gut microbiota, yet its ability to interact with and affect the bacterial gut microbiota is largely uncharacterized. Previous reports showed limited changes in microbiota composition as defined by bacterial species abundance as a consequence of C. albicans colonization. We also observed only a few bacterial genera that were significantly altered in abundance in C. albicans-colonized mice; however, C. albicans colonization significantly changed the structure of the bacterial microbiota co-occurrence network. Additionally, C. albicans colonization changed the response of the bacterial microbiota ecosystem to a clinically relevant perturbation, challenge with the antibiotic clindamycin.
Collapse
Affiliation(s)
- Laura Markey
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Antonia Pugliese
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Theresa Tian
- Department of Chemical and Biological Engineering, Tufts University School of Engineering, Medford, Massachusetts, USA
| | - Farrah Roy
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Kyongbum Lee
- Department of Chemical and Biological Engineering, Tufts University School of Engineering, Medford, Massachusetts, USA
| | - Carol A Kumamoto
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
83
|
Authier H, Salon M, Rahabi M, Bertrand B, Blondeau C, Kuylle S, Holowacz S, Coste A. Oral Administration of Lactobacillus helveticus LA401 and Lactobacillus gasseri LA806 Combination Attenuates Oesophageal and Gastrointestinal Candidiasis and Consequent Gut Inflammation in Mice. J Fungi (Basel) 2021; 7:57. [PMID: 33467443 PMCID: PMC7830595 DOI: 10.3390/jof7010057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/08/2021] [Accepted: 01/13/2021] [Indexed: 12/17/2022] Open
Abstract
Candida albicans is an opportunistic pathogen that causes mucosal gastrointestinal (GI) candidiasis tightly associated with gut inflammatory status. The emergence of drug resistance, the side effects of currently available antifungals and the high frequency of recurrent candidiasis indicate that new and improved therapeutics are needed. Probiotics have been suggested as a useful alternative for the management of candidiasis. We demonstrated that oral administration of Lactobacillus gasseri LA806 alone or combined with Lactobacillus helveticus LA401 in Candida albicans-infected mice decrease the Candida colonization of the oesophageal and GI tract, highlighting a protective role for these strains in C. albicans colonization. Interestingly, the probiotic combination significantly modulates the composition of gut microbiota towards a protective profile and consequently dampens inflammatory and oxidative status in the colon. Moreover, we showed that L. helveticus LA401 and/or L. gasseri LA806 orient macrophages towards a fungicidal phenotype characterized by a C-type lectin receptors signature composed of Dectin-1 and Mannose receptor. Our findings suggest that the use of the LA401 and LA806 combination might be a promising strategy to manage GI candidiasis and the inflammation it causes by inducing the intrinsic antifungal activities of macrophages. Thus, the probiotic combination is a good candidate for managing GI candidiasis by inducing fungicidal functions in macrophages while preserving the GI integrity by modulating the microbiota and inflammation.
Collapse
Affiliation(s)
- Hélène Authier
- UMR 152 Pharma-Dev, Université de Toulouse, IRD, UPS, 31432 Toulouse, France; (M.S.); (M.R.); (B.B.)
| | - Marie Salon
- UMR 152 Pharma-Dev, Université de Toulouse, IRD, UPS, 31432 Toulouse, France; (M.S.); (M.R.); (B.B.)
| | - Mouna Rahabi
- UMR 152 Pharma-Dev, Université de Toulouse, IRD, UPS, 31432 Toulouse, France; (M.S.); (M.R.); (B.B.)
| | - Bénédicte Bertrand
- UMR 152 Pharma-Dev, Université de Toulouse, IRD, UPS, 31432 Toulouse, France; (M.S.); (M.R.); (B.B.)
| | | | | | | | - Agnès Coste
- UMR 152 Pharma-Dev, Université de Toulouse, IRD, UPS, 31432 Toulouse, France; (M.S.); (M.R.); (B.B.)
| |
Collapse
|
84
|
Candida Administration Worsens Uremia-Induced Gut Leakage in Bilateral Nephrectomy Mice, an Impact of Gut Fungi and Organismal Molecules in Uremia. mSystems 2021; 6:6/1/e01187-20. [PMID: 33436518 PMCID: PMC7901485 DOI: 10.1128/msystems.01187-20] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The impact of gut fungi and (1→3)-β-d-glucan (BG), a major fungal cell wall component, on uremia was explored by Candida albicans oral administration in bilateral nephrectomy (BiNx) mice because of the prominence of C. albicans in the human intestine but not in mice. As such, BiNx with Candida administration (BiNx-Candida) enhanced intestinal injury (colon cytokines and apoptosis), gut leakage (fluorescein isothiocyanate [FITC]-dextran assay, endotoxemia, serum BG, and bacteremia), systemic inflammation, and liver injury at 48 h postsurgery compared with non-Candida BiNx mice. Interestingly, uremia-induced enterocyte apoptosis was severe enough for gut translocation of viable bacteria, as indicated by culture positivity for bacteria in blood, mesenteric lymph nodes (MLNs), and other organs, which was more severe in BiNx-Candida than in non-Candida BiNx mice. Candida induced alterations in the gut microbiota of BiNx mice as indicated by (i) the higher fungal burdens in the feces of BiNx-Candida mice than in sham-Candida mice by culture methods and (ii) increased Bacteroides with decreased Firmicutes and reduced bacterial diversity in the feces of BiNx-Candida mice compared with non-Candida BiNx mice by fecal microbiome analysis. In addition, lipopolysaccharide plus BG (LPS+BG), compared with each molecule alone, induced high supernatant cytokine levels, which were enhanced by uremic mouse serum in both hepatocytes (HepG2 cells) and macrophages (RAW264.7 cells). Moreover, LPS+BG, but not each molecule alone, reduced the glycolysis capacity and mitochondrial function in HepG2 cells as determined by extracellular flux analysis. Additionally, a probiotic, Lactobacillus rhamnosus L34 (L34), attenuated disease severity only in BiNx-Candida mice but not in non-Candida BiNx mice, as indicated by liver injury and serum cytokines through the attenuation of gut leakage, the fecal abundance of fungi, and fecal bacterial diversity but not fecal Gram-negative bacteria. In conclusion, Candida enhanced BiNx severity through the worsening of gut leakage and microbiota alterations that resulted in bacteremia, endotoxemia, and glucanemia.IMPORTANCE The impact of fungi in the intestine on acute uremia was demonstrated by the oral administration of Candida albicans in mice with the removal of both kidneys. Because fungi in the mouse intestine are less abundant than in humans, a Candida-administered mouse model has more resemblance to patient conditions. Accordingly, acute uremia, without Candida, induced intestinal mucosal injury, which resulted in the translocation of endotoxin, a major molecule of gut bacteria, from the intestine into blood circulation. In acute uremia with Candida, intestinal injury was more severe due to fungi and the alteration in intestinal bacteria (increased Bacteroides with decreased Firmicutes), leading to the gut translocation of both endotoxin from gut bacteria and (1→3)-β-d-glucan from Candida, which synergistically enhanced systemic inflammation in acute uremia. Both pathogen-associated molecules were delivered to the liver and induced hepatocyte inflammatory responses with a reduced energy production capacity, resulting in acute uremia-induced liver injury. In addition, Lactobacillus rhamnosus attenuated intestinal injury through reduced gut Candida and improved intestinal bacterial conditions.
Collapse
|
85
|
Valand N, Girija UV. Candida Pathogenicity and Interplay with the Immune System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:241-272. [PMID: 34661898 DOI: 10.1007/978-3-030-67452-6_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Candida species are opportunistic fungal pathogens that are part of the normal skin and mucosal microflora. Overgrowth of Candida can cause infections such as thrush or life-threatening invasive candidiasis in immunocompromised patients. Though Candida albicans is highly prevalent, several non-albicans species are also isolated from nosocomial infections. Candida sp. are over presented in the gut of people with Crohn's disease and certain types of neurological disorders, with hyphal form and biofilms being the most virulent states. In addition, Candida uses several secreted and cell surface molecules such as pH related antigen 1, High affinity glucose transporter, Phosphoglycerate mutase 1 and lipases to establish pathogenicity. A strong innate immune response is elicited against Candida via dendritic cells, neutrophils and macrophages. All three complement pathways are also activated. Production of proinflammatory cytokines IL-10 and IL-12 signal differentiation of CD4+ cells into Th1 and Th2 cells, whereas IL-6, IL-17 and IL-23 induce Th17 cells. Importance of T-lymphocytes is reflected in depleted T-cell count patients being more prone to Candidiasis. Anti- Candida antibodies also play a role against candidiasis using various mechanisms such as targeting virulent enzymes and exhibiting direct candidacidal activity. However, the significance of antibody response during infection remains controversial. Furthermore, some of the Candida strains have evolved molecular strategies to evade the sophisticated host attack by proteolysis of components of immune system and interfering with immune signalling pathways. Emergence of several non-albicans species that are resistant to current antifungal agents makes treatment more difficult. Therefore, deeper insight into interactions between Candida and the host immune system is required for discovery of novel therapeutic options.
Collapse
Affiliation(s)
- Nisha Valand
- Leicester School of Allied Health and Life sciences, Faculty of Health and Life Sciences, De Montfort University, Leicester, UK
| | - Umakhanth Venkatraman Girija
- Leicester School of Allied Health and Life sciences, Faculty of Health and Life Sciences, De Montfort University, Leicester, UK.
| |
Collapse
|
86
|
Diet Rich in Simple Sugars Promotes Pro-Inflammatory Response via Gut Microbiota Alteration and TLR4 Signaling. Cells 2020; 9:cells9122701. [PMID: 33339337 PMCID: PMC7766268 DOI: 10.3390/cells9122701] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/05/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Diet is a strong modifier of microbiome and mucosal microenvironment in the gut. Recently, components of western-type diets have been associated with metabolic and immune diseases. Here, we studied how high-sugar diet (HSD) consumption influences gut mucosal barrier and immune response under steady state conditions and in a mouse model of acute colitis. We found that HSD significantly increased gut permeability, spleen weight, and neutrophil levels in spleens of healthy mice. Subsequent dextran sodium sulfate administration led to severe colitis. In colon, HSD significantly promoted neutrophil infiltration and increased the levels of IL-6, IL-1β, and TNF-α. Moreover, HSD-fed mice had significantly higher abundance of pathobionts, such as Escherichia coli and Candida, in fecal samples. Although germ-free mice colonized with microbiota of conventionally reared mice that consumed different diets had equally severe colitis, mice colonized with HSD microbiota showed markedly increased infiltration of neutrophils to the gut. The induction of colitis in Toll-like receptor 4 (TLR4)-deficient HSD-fed mice led to significantly milder colitis than in wild-type mice. In conclusion, our results suggested a significant role of HSD in disruption of barrier integrity and balanced mucosal and systemic immune response. In addition, these processes seemed to be highly influenced by resident potentially pathogenic microbiota or metabolites via the TLR4 signaling pathway.
Collapse
|
87
|
Hyposalivation, oral health, and Candida colonization in independent dentate elders. PLoS One 2020; 15:e0242832. [PMID: 33237956 PMCID: PMC7688165 DOI: 10.1371/journal.pone.0242832] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Hyposalivation is an important problem in elders and could interfere with several oral functions and microbial ecology. While the number of independent elders who retain more natural teeth increases worldwide, few studies examined hyposalivation in this population. Thus, this study aims to examine relationships between hyposalivation, oral health conditions and oral Candida colonization in independent dentate elders and evaluate factors associated with salivary flow and Candida carriage. We conducted a cross-sectional study in fifty-three dentate elders (≥65 years old with at least 4 pairs of posterior occlusal contacts) with no, or well-controlled, systemic conditions. Participants were interviewed for medical history, subjective dry mouth symptoms, oral hygiene practices and denture information. Unstimulated and stimulated salivary flow rates, objective dry mouth signs, gingival, tongue-coating, and root-caries indices were recorded. Stimulated saliva was cultured on Sabouraud-dextrose agar for Candida counts. Candida species were identified using chromogenic Candida agar and polymerase chain reaction. Statistical significance level was set at p<0.05. The results showed that hyposalivation was associated with higher gingival and tongue-coating indices (p = 0.003 and 0.015, respectively), but not root-caries index. Hyposalivation was also associated with higher prevalence of oral Candida colonization (p = 0.010; adjusted OR = 4.36, 95% confidence interval = 1.29–14.72). These two indices and Candida load were negatively correlated with unstimulated and stimulated salivary flow rates. Interestingly, non-albicans Candida species were more prevalent in denture wearers (p = 0.017). Hence, hyposalivation is a risk factor for poorer oral health and oral Candida colonization in independent dentate elders. Because of its potential adverse effects on oral and systemic health, hyposalivation should be carefully monitored in elders.
Collapse
|
88
|
González-Sanmiguel J, Schuh CMAP, Muñoz-Montesino C, Contreras-Kallens P, Aguayo LG, Aguayo S. Complex Interaction between Resident Microbiota and Misfolded Proteins: Role in Neuroinflammation and Neurodegeneration. Cells 2020; 9:E2476. [PMID: 33203002 PMCID: PMC7697492 DOI: 10.3390/cells9112476] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/02/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD) and Creutzfeldt-Jakob disease (CJD) are brain conditions affecting millions of people worldwide. These diseases are associated with the presence of amyloid-β (Aβ), alpha synuclein (α-Syn) and prion protein (PrP) depositions in the brain, respectively, which lead to synaptic disconnection and subsequent progressive neuronal death. Although considerable progress has been made in elucidating the pathogenesis of these diseases, the specific mechanisms of their origins remain largely unknown. A body of research suggests a potential association between host microbiota, neuroinflammation and dementia, either directly due to bacterial brain invasion because of barrier leakage and production of toxins and inflammation, or indirectly by modulating the immune response. In the present review, we focus on the emerging topics of neuroinflammation and the association between components of the human microbiota and the deposition of Aβ, α-Syn and PrP in the brain. Special focus is given to gut and oral bacteria and biofilms and to the potential mechanisms associating microbiome dysbiosis and toxin production with neurodegeneration. The roles of neuroinflammation, protein misfolding and cellular mediators in membrane damage and increased permeability are also discussed.
Collapse
Affiliation(s)
| | - Christina M. A. P. Schuh
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile; (C.M.A.P.S.); (P.C.-K.)
| | - Carola Muñoz-Montesino
- Department of Physiology, Universidad de Concepción, Concepción 4070386, Chile; (J.G.-S.); (C.M.-M.)
| | - Pamina Contreras-Kallens
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile; (C.M.A.P.S.); (P.C.-K.)
| | - Luis G. Aguayo
- Department of Physiology, Universidad de Concepción, Concepción 4070386, Chile; (J.G.-S.); (C.M.-M.)
- Program on Neuroscience, Psychiatry and Mental Health, Universidad de Concepción, Concepción 4070386, Chile
| | - Sebastian Aguayo
- School of Dentistry, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| |
Collapse
|
89
|
Krishnamoorthy AL, Lemus AA, Solomon AP, Valm AM, Neelakantan P. Interactions between Candida albicans and Enterococcus faecalis in an Organotypic Oral Epithelial Model. Microorganisms 2020; 8:E1771. [PMID: 33187237 PMCID: PMC7696566 DOI: 10.3390/microorganisms8111771] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/06/2020] [Accepted: 11/10/2020] [Indexed: 12/23/2022] Open
Abstract
Candida albicans as an opportunistic pathogen exploits the host immune system and causes a variety of life-threatening infections. The polymorphic nature of this fungus gives it tremendous advantage to breach mucosal barriers and cause oral and disseminated infections. Similar to C. albicans, Enterococcus faecalis is a major opportunistic pathogen, which is of critical concern in immunocompromised patients. There is increasing evidence that E. faecalis co-exists with C. albicans in the human body in disease samples. While the interactive profiles between these two organisms have been studied on abiotic substrates and mouse models, studies on their interactions on human oral mucosal surfaces are non-existent. Here, for the first time, we comprehensively characterized the interactive profiles between laboratory and clinical isolates of C. albicans (SC5314 and BF1) and E. faecalis (OG1RF and P52S) on an organotypic oral mucosal model. Our results demonstrated that the dual species biofilms resulted in profound surface erosion and significantly increased microbial invasion into mucosal compartments, compared to either species alone. Notably, several genes of C. albicans involved in tissue adhesion, hyphal formation, fungal invasion, and biofilm formation were significantly upregulated in the presence of E. faecalis. By contrast, E. faecalis genes involved in quorum sensing, biofilm formation, virulence, and mammalian cell invasion were downregulated. This study highlights the synergistic cross-kingdom interactions between E. faecalis and C. albicans in mucosal tissue invasion.
Collapse
Affiliation(s)
- Akshaya Lakshmi Krishnamoorthy
- Faculty of Dentistry, The University of Hong Kong, Pok Fu Lam, Hong Kong;
- Quorum Sensing Laboratory, Center of Research in Infectious Diseases, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India;
| | - Alex A. Lemus
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA; (A.A.L.); (A.M.V.)
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Center of Research in Infectious Diseases, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India;
| | - Alex M. Valm
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA; (A.A.L.); (A.M.V.)
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| | | |
Collapse
|
90
|
Domnich M, Riedesel J, Pylaeva E, Kürten CHL, Buer J, Lang S, Jablonska J. Oral Neutrophils: Underestimated Players in Oral Cancer. Front Immunol 2020; 11:565683. [PMID: 33162980 PMCID: PMC7582090 DOI: 10.3389/fimmu.2020.565683] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 09/10/2020] [Indexed: 01/03/2023] Open
Abstract
The composition of the oral milieu reflects oral health. Saliva provides an environment for multiple microorganisms, and contains soluble factors and immune cells. Neutrophils, which rapidly react on the changes in the microenvironment, are a major immune cell population in saliva and thus may serve as a biomarker for oral pathologies. This review focuses on salivary neutrophils in the oral cavity, their phenotype changes in physiological and pathological conditions, as well as on factors regulating oral neutrophil amount, activation and functionality, with special emphasis on oral cancer and its risk factors.
Collapse
Affiliation(s)
- Maksim Domnich
- Department of Otorhinolaryngology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jana Riedesel
- Department of Otorhinolaryngology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ekaterina Pylaeva
- Department of Otorhinolaryngology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Cornelius H. L. Kürten
- Department of Otorhinolaryngology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jan Buer
- Institute of Medical Microbiology, University of Duisburg-Essen, Essen, Germany
| | - Stephan Lang
- Department of Otorhinolaryngology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jadwiga Jablonska
- Department of Otorhinolaryngology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
91
|
Arastehfar A, Carvalho A, Nguyen MH, Hedayati MT, Netea MG, Perlin DS, Hoenigl M. COVID-19-Associated Candidiasis (CAC): An Underestimated Complication in the Absence of Immunological Predispositions? J Fungi (Basel) 2020; 6:jof6040211. [PMID: 33050019 PMCID: PMC7712987 DOI: 10.3390/jof6040211] [Citation(s) in RCA: 169] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 12/20/2022] Open
Abstract
The recent global pandemic of COVID-19 has predisposed a relatively high number of patients to acute respiratory distress syndrome (ARDS), which carries a risk of developing super-infections. Candida species are major constituents of the human mycobiome and the main cause of invasive fungal infections, with a high mortality rate. Invasive yeast infections (IYIs) are increasingly recognized as s complication of severe COVID-19. Despite the marked immune dysregulation in COVID-19, no prominent defects have been reported in immune cells that are critically required for immunity to Candida. This suggests that relevant clinical factors, including prolonged ICU stays, central venous catheters, and broad-spectrum antibiotic use, may be key factors causing COVID-19 patients to develop IYIs. Although data on the comparative performance of diagnostic tools are often lacking in COVID-19 patients, a combination of serological and molecular techniques may present a promising option for the identification of IYIs. Clinical awareness and screening are needed, as IYIs are difficult to diagnose, particularly in the setting of severe COVID-19. Echinocandins and azoles are the primary antifungal used to treat IYIs, yet the therapeutic failures exerted by multidrug-resistant Candida spp. such as C. auris and C. glabrata call for the development of new antifungal drugs with novel mechanisms of action.
Collapse
Affiliation(s)
- Amir Arastehfar
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA;
- Correspondence: (A.A.); (A.C.); (M.H.); Tel./Fax: +1-201-880-3100 (A.A.); +351-253-604811 (A.C.); +1-619-543-5605 (M.H.)
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Guimarães/Braga, Portugal
- Correspondence: (A.A.); (A.C.); (M.H.); Tel./Fax: +1-201-880-3100 (A.A.); +351-253-604811 (A.C.); +1-619-543-5605 (M.H.)
| | - M. Hong Nguyen
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Mohammad Taghi Hedayati
- Invasive Fungi Research Center, Department of Medical Mycology, School of Medicine, Mazandaran University of Medical Sciences, Sari 4815733971, Iran;
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, 6500HB Nijmegen, The Netherlands;
- Department of Genomics & Immunoregulation, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - David S. Perlin
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA;
| | - Martin Hoenigl
- Clinical and Translational Fungal-Working Group, University of California San Diego, La Jolla, CA 92093, USA
- Section of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA
- Correspondence: (A.A.); (A.C.); (M.H.); Tel./Fax: +1-201-880-3100 (A.A.); +351-253-604811 (A.C.); +1-619-543-5605 (M.H.)
| |
Collapse
|
92
|
Ashammakhi N, Nasiri R, Barros NRD, Tebon P, Thakor J, Goudie M, Shamloo A, Martin MG, Khademhosseini A. Gut-on-a-chip: Current progress and future opportunities. Biomaterials 2020; 255:120196. [PMID: 32623181 PMCID: PMC7396314 DOI: 10.1016/j.biomaterials.2020.120196] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/11/2020] [Accepted: 06/09/2020] [Indexed: 12/21/2022]
Abstract
Organ-on-a-chip technology tries to mimic the complexity of native tissues in vitro. Important progress has recently been made in using this technology to study the gut with and without microbiota. These in vitro models can serve as an alternative to animal models for studying physiology, pathology, and pharmacology. While these models have greater physiological relevance than two-dimensional (2D) cell systems in vitro, endocrine and immunological functions in gut-on-a-chip models are still poorly represented. Furthermore, the construction of complex models, in which different cell types and structures interact, remains a challenge. Generally, gut-on-a-chip models have the potential to advance our understanding of the basic interactions found within the gut and lay the foundation for future applications in understanding pathophysiology, developing drugs, and personalizing medical treatments.
Collapse
Affiliation(s)
- Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA.
| | - Rohollah Nasiri
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA; Department of Mechanical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran
| | - Natan Roberto de Barros
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA.
| | - Peyton Tebon
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA
| | - Jai Thakor
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA
| | - Marcus Goudie
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA
| | - Amir Shamloo
- Department of Mechanical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran
| | - Martin G Martin
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA; Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA; Terasaki Institute for Biomedical Innovation, Los Angeles, CA, USA.
| |
Collapse
|
93
|
Diaz PI, Dongari-Bagtzoglou A. Critically Appraising the Significance of the Oral Mycobiome. J Dent Res 2020; 100:133-140. [PMID: 32924741 DOI: 10.1177/0022034520956975] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recent efforts to understand the oral microbiome have focused on its fungal component. Since fungi occupy a low proportion of the oral microbiome biomass, mycobiome studies rely on sequencing of internal transcribed spacer (ITS) amplicons. ITS-based studies usually detect hundreds of fungi in oral samples. Here, we review the oral mycobiome, critically appraising the significance of such large fungal diversity. When harsh lysis methods are used to extract DNA, 2 oral mycobiome community types (mycotypes) are evident, each dominated by only 1 genus, either Candida or Malassezia. The rest of the diversity in ITS surveys represents low-abundance fungi possibly acquired from the environment and ingested food. So far, Candida is the only genus demonstrated to reach a significant biomass in the oral cavity and clearly shown to be associated with a distinct oral ecology. Candida thrives in the presence of lower oral pH and is enriched in caries, with mechanistic studies in animal models suggesting it participates in the disease process by synergistically interacting with acidogenic bacteria. Candida serves as the main etiological agent of oral mucosal candidiasis, in which a Candida-bacteriome partnership plays a key role. The function of other potential oral colonizers, such as lipid-dependent Malassezia, is still unclear, with further studies needed to establish whether Malassezia are metabolically active oral commensals. Low-abundance oral mycobiome members acquired from the environment may be viable in the oral cavity, and although they may not play a significant role in microbiome communities, they could serve as opportunistic pathogens in immunocompromised hosts. We suggest that further work is needed to ascertain the significance of oral mycobiome members beyond Candida. ITS-based surveys should be complemented with other methods to determine the in situ biomass and metabolic state of fungi thought to play a role in the oral environment.
Collapse
Affiliation(s)
- P I Diaz
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, State University of New York, Buffalo, NY, USA.,UB Microbiome Center, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - A Dongari-Bagtzoglou
- Division of Periodontology, Department of Oral Health and Diagnostic Sciences, School of Dental Medicine, UConn Health, Farmington, CT, USA
| |
Collapse
|
94
|
Souza E Silva VCD, Oliveira VDC, Sousa ÁFLD, Bim FL, Macedo AP, Andrade DD, Watanabe E. Prevalence and susceptibility profile of Candida spp. isolated from patients in cancer therapy. Arch Oral Biol 2020; 119:104906. [PMID: 32947164 DOI: 10.1016/j.archoralbio.2020.104906] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVE This study determined the prevalence of Candida spp. in the saliva of cancer patients. Furthermore, we assessed the antimicrobial activity of mouthwashes against the isolated strains and its susceptibility to amphotericin B and fluconazole. METHODS Thirty-four cancer patients undergoing radiotherapy, chemotherapy alone or combined treatment were investigated for oral Candida spp. colonization and compared in regard to mucositis presence. The maximum inhibitory dilution was used to assess the antimicrobial activity of Periogard®, Cepacol® Cool Ice and 0.12 % Chlorhexidine Digluconate mouthwashes against the isolates. In parallel, susceptibility to amphotericin B and fluconazole was determined by agar-based E-test. Data did not adhere to normal distribution as inferred by the Shapiro-Wilk test and statistical analysis was conducted by non-parametric McNemar test (α0.05). RESULTS Twenty-seven participants (79.4 %) were male, 19 (55.9 %) had mucositis and 9 (26.5 %) were colonized by Candida spp. 12 different strains of Candida spp. were isolated, being Candida albicans the most prevalent strain. Risk of Candida spp. colonization was increased by almost twofold among the participants with mucositis (odds ratio: 1.84; 95 % confidence interval: 0.37-9.07). Mouthwash Cepacol® Cool Ice presented better antimicrobial activity against Candida spp. while 0.12 % Chlorhexidine exhibited the worst activity. All strains were sensitive to amphotericin B, and 2 non-albicans strains were dose-dependent sensitive to fluconazole. CONCLUSION Considering the increased risk of colonization byCandida spp. in patients with mucositis, and the emergence of antifungal drug resistance, the antiseptics use could benefit the maintenance of cancer patient's oral health.
Collapse
Affiliation(s)
- Vanessa Castro de Souza E Silva
- Human Exposome and Infectious Diseases Network (HEID), School of Nursing of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Viviane de Cássia Oliveira
- Human Exposome and Infectious Diseases Network (HEID), School of Nursing of Ribeirão Preto, University of São Paulo, São Paulo, Brazil; Department of Dental Materials and Prostheses, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Álvaro Francisco Lopes de Sousa
- Human Exposome and Infectious Diseases Network (HEID), School of Nursing of Ribeirão Preto, University of São Paulo, São Paulo, Brazil; Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Portugal
| | - Felipe Lazarini Bim
- Human Exposome and Infectious Diseases Network (HEID), School of Nursing of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Ana Paula Macedo
- Department of Dental Materials and Prostheses, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Denise de Andrade
- Human Exposome and Infectious Diseases Network (HEID), School of Nursing of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Evandro Watanabe
- Human Exposome and Infectious Diseases Network (HEID), School of Nursing of Ribeirão Preto, University of São Paulo, São Paulo, Brazil; Department of Restorative Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
95
|
Silva RVDR, Costa MI, Jarros IC, Del Bel Cury AA, Sidhu SK, Negri M, Pascotto RC. Effect of Silicon dioxide coating of acrylic resin surfaces on Candida albicans adhesion. Braz Oral Res 2020; 34:e110. [PMID: 32876123 DOI: 10.1590/1807-3107bor-2020.vol34.0110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/10/2020] [Indexed: 12/29/2022] Open
Abstract
Acrylic resin has been used in the manufacture of prostheses, however, in the oral cavity, this material starts to retain microorganisms capable of causing gingival inflammation due its porosities. The aim of this study was to evaluate the influence of the use of silicon dioxide as a coating layer applied onto acrylic resin, on the adhesion of Candida albicans (Ca). After the incubation period in Sabouraud Dextrose Broth, a total of 1 ml of the Ca suspension was added to plate wells, each well containing a specimen of acrylic resin. The adhesion ability of Ca on acrylic resin was determined by counting colonies. Three groups (n = 6) of acrylic resin were assessed: with polishing (RP); without polishing (RW); with polishing and coating layer of silicon dioxide (RPC). Ca deposited on the surface of the acrylic resin was also observed using Scanning Electron Microscopy (SEM). Statistical assessment by Kruskal-Wallis and Student-Newman-Keuls Method were done (α = 2%). There was significant difference among the groups. The RPC group showed the lowest growth, with an average of 5.59 Log CFU/cm 2 ; there was a statistically significant difference in relation to group RW, which presented a growth of 6.07 Log CFU/cm 2 and to group RP with 5.91 Log CFU/cm 2 (p < 000.1). SEM images demonstrated that in the RP and RPC group, the surface of the resin had greater regularity, and smaller number of microorganisms. The application of silicon dioxide coating on acrylic resin appears to be a promising alternative, and its use can help in reducing the adhesion of Ca in prostheses.
Collapse
Affiliation(s)
| | - Maiara Ignácio Costa
- Division of Medical Mycology, Department of Clinical Analysis, Universidade Estadual de Maringá, Maringá, PR, Brazil
| | - Isabele Carrilho Jarros
- Division of Medical Mycology, Department of Clinical Analysis, Universidade Estadual de Maringá, Maringá, PR, Brazil
| | - Altair Antoninha Del Bel Cury
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas, Piracicaba, SP, Brazil
| | - Sharanbir Kaur Sidhu
- Centre for Oral Bioengineering, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Melyssa Negri
- Division of Medical Mycology, Department of Clinical Analysis, Universidade Estadual de Maringá, Maringá, PR, Brazil
| | | |
Collapse
|
96
|
Recognition of Candida albicans and Role of Innate Type 17 Immunity in Oral Candidiasis. Microorganisms 2020; 8:microorganisms8091340. [PMID: 32887412 PMCID: PMC7563233 DOI: 10.3390/microorganisms8091340] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/01/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023] Open
Abstract
Candida albicans is an opportunistic pathogenic fungus considered to be a common member of the human microflora. Similar to some other opportunistic microbes, C. albicans can invade and benefit from its host when the immune status of that host is weakened. Most often this happens to immunocompromised individuals, leading to the infection of oral and vaginal mucosae or the systemic spread of the pathogen throughout the entire body. Oropharyngeal candidiasis (OPC) occurs in up to 90 percent of patients with acquired immunodeficiency syndrome (AIDS), making it the most frequent opportunistic infection for this group. Upon first signs of fungal invasion, a range of host signaling activates in order to eliminate the threat. Epithelial and myeloid type cells detect C. albicans mainly through receptor tyrosine kinases and pattern-recognition receptors. This review provides an overview of downstream signaling resulting in an adequate immune response through the activation of various transcription factors. The study discusses recent advances in research of the interleukin-17 (IL-17) producing innate cells, including natural T helper 17 (nTh17) cells, γδ T cells, invariant natural killer T (iNKT) cells and type 3 innate lymphoid cells (ILC3) that are involved in response to oral C. albicans infections.
Collapse
|
97
|
Ferrer MD, López-López A, Nicolescu T, Perez-Vilaplana S, Boix-Amorós A, Dzidic M, Garcia S, Artacho A, Llena C, Mira A. Topic Application of the Probiotic Streptococcus dentisani Improves Clinical and Microbiological Parameters Associated With Oral Health. Front Cell Infect Microbiol 2020; 10:465. [PMID: 32984080 PMCID: PMC7488176 DOI: 10.3389/fcimb.2020.00465] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 07/28/2020] [Indexed: 01/04/2023] Open
Abstract
Streptococcus dentisani 7746, isolated from dental plaque of caries-free individuals, has been shown to have several beneficial effects in vitro which could contribute to promote oral health, including an antimicrobial activity against oral pathogens by the production of bacteriocins and a pH buffering capacity through ammonia production. Previous work has shown that S. dentisani was able to colonize the oral cavity for 2–4 weeks after application. The aim of the present work was to evaluate its clinical efficacy by a randomized, double-blind, placebo-controlled parallel group study. Fifty nine volunteers were enrolled in the study and randomly assigned to a treatment or placebo group. The treatment consisted of a bucco-adhesive gel application (2.5 109 cfu/dose) with a dental splint for 5 min every 48 h, for a period of 1 month (i.e., 14 doses). Dental plaque and saliva samples were collected at baseline, 15 and 30 days after first application, and 15 days after the end of treatment. At baseline, there was a significant correlation between S. dentisani levels and frequency of toothbrushing. Salivary flow, a major factor influencing oral health, was significantly higher in the probiotic group at day 15 compared with the placebo (4.4 and 3.4 ml/5 min, respectively). In the probiotic group, there was a decrease in the amount of dental plaque and in gingival inflammation, but no differences were observed in the placebo group. The probiotic group showed a significant increase in the levels of salivary ammonia and calcium. Finally, Illumina sequencing of plaque samples showed a beneficial shift in bacterial composition at day 30 relative to baseline, with a reduction of several cariogenic organisms and the key players in plaque formation, probably as a result of bacteriocins production. Only 58% of the participants in the probiotic group showed increased plaque levels of S. dentisani at day 30 and 71% by day 45, indicating that the benefits of S. dentisani application could be augmented by improving colonization efficiency. In conclusion, the application of S. dentisani 7746 improved several clinical and microbiological parameters associated with oral health, supporting its use as a probiotic to prevent tooth decay.
Collapse
Affiliation(s)
- María D Ferrer
- Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO), Valencia, Spain
| | - Aranzazu López-López
- Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO), Valencia, Spain
| | - Teodora Nicolescu
- Clínica Odontológica, Fundació Lluís Alcanyis, Universitat de València, Valencia, Spain
| | | | - Alba Boix-Amorós
- Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO), Valencia, Spain
| | - Majda Dzidic
- Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO), Valencia, Spain
| | - Sandra Garcia
- Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO), Valencia, Spain
| | - Alejandro Artacho
- Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO), Valencia, Spain
| | - Carmen Llena
- Clínica Odontológica, Fundació Lluís Alcanyis, Universitat de València, Valencia, Spain
| | - Alex Mira
- Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO), Valencia, Spain
| |
Collapse
|
98
|
Kumamoto CA, Gresnigt MS, Hube B. The gut, the bad and the harmless: Candida albicans as a commensal and opportunistic pathogen in the intestine. Curr Opin Microbiol 2020; 56:7-15. [PMID: 32604030 PMCID: PMC7744392 DOI: 10.1016/j.mib.2020.05.006] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/04/2020] [Accepted: 05/11/2020] [Indexed: 12/26/2022]
Abstract
Candida albicans is a regular member of the intestinal microbiota in the majority of the human population. This underscores C. albicans' adaptation to life in the intestine without inducing competitive interactions with other microbes, or immune responses detrimental to its survival. However, specific conditions such as a dysbalanced microbiome, a suppression of the immune system, and an impaired intestinal barrier can predispose for invasive, mostly nosocomial, C. albicans infections. Colonization of the intestine and translocation through the intestinal barrier are fundamental aspects of the processes preceding life-threatening systemic candidiasis. Insights into C. albicans' commensal lifestyle and translocation can thus help us to understand how patients develop candidiasis, and may provide leads for therapeutic strategies aimed at preventing infection. In this review, we discuss the commensal lifestyle of C. albicans in the intestine, the role of morphology for commensalism, the influence of diet, and the interactions with bacteria of the microbiota.
Collapse
Affiliation(s)
- Carol A Kumamoto
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02111, USA
| | - Mark S Gresnigt
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knoell-Institute, Beutenbergstraße 11a, 07745 Jena, Germany; Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11a 07745, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knoell-Institute, Beutenbergstraße 11a, 07745 Jena, Germany; Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University, 07743 Jena, Germany.
| |
Collapse
|
99
|
Ge Y, Pan M, Zhang C, Wang C, Ma K, Yan G, Wang T, Wu D, Shao J. Paeonol alleviates dextran sodium sulfate induced colitis involving Candida albicans-associated dysbiosis. Med Mycol 2020; 59:335-344. [PMID: 32598443 DOI: 10.1093/mmy/myaa053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/21/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel disease (IBD), which consists of ulcerative colitis (UC) and Crohn's disease (CD), is a chronic inflammatory disorder of the gastrointestinal tract. Occurrence and development of UC have been associated with multiple potential causative factors, which include fungal dysbiosis. Growing evidence reveals that Candida albicans-associated dysbiosis is correlated with clinical deterioration in UC. Paeonol (PAE) is a commonly used traditional medicine with multiple reported properties including effective alleviation of UC. In this study, a murine UC model was established by colonizing mice with additional C. albicans via gavage prior to dextran sodium sulfate (DSS) administration. Effects of PAE treatment were also assessed at initiation and in preestablished C. albicans-associated colitis. The results showed that C. albicans supplementation could aggravate disease activity index (DAI), compromise mucosal integrity, exacerbate fecal and tissue fungal burdens, increase serum β-glucan and anti-Saccharomyces cerevisiae antibody (ASCA) levels, promote serum and colonic tissue pro-inflammatory cytokine secretion (tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, and IL-8) and decrease the anti-inflammatory cytokine IL-10 level. It also stimulated Dectin-1, TLR2 and TLR4 as well as expression of their downstream effector NF-κB in colonic tissue. After PAE treatment, the adverse impacts of C. albicans on colitis were relieved, via decreased receptor-associated local and systemic inflammation. Our study suggests that PAE should be a candidate for treatment of fungal dysbiosis-associated UC and may act through the Dectin-1/NF-κB pathway in collaboration with TLR2 and TLR4. LAY SUMMARY Candida albicans is believed to be an important stimulator in ulcerative colitice (UC) development. Suppressing the growth of intestinal C. albicans can be contributory to the amelioration of UC. Paeonol (PAE) is a commonly used traditional medicine with multiple biological functions. In this study, we observed that PAE could alleviate symptoms in mice UC model accompanying with burden reduction of C. albicans. Therefore, we suppose that PAE can be a candidate in the treatment of C. albicans-associated UC.
Collapse
Affiliation(s)
- Yuzhu Ge
- Laboratory of Infection and Tumor, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 436 Room, Zhijing Building, No. 1 Qianjiang Road, Xinzhan District, Hefei 230012, Anhui, China
| | - Min Pan
- Laboratory of Infection and Tumor, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 436 Room, Zhijing Building, No. 1 Qianjiang Road, Xinzhan District, Hefei 230012, Anhui, China
| | - Chuanfeng Zhang
- Laboratory of Infection and Tumor, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 436 Room, Zhijing Building, No. 1 Qianjiang Road, Xinzhan District, Hefei 230012, Anhui, China
| | - Changzhong Wang
- Laboratory of Infection and Tumor, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 436 Room, Zhijing Building, No. 1 Qianjiang Road, Xinzhan District, Hefei 230012, Anhui, China.,Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Zhijing Building, No. 1 Qianjiang Road, Xinzhan District, Hefei 230012, Anhui, China.,Key Laboratory of Xin'An Medicine, Ministry of Education, Anhui Academy of Chinese Medicine, Xin'An Building, No. 103 Meishan Road, Shushan District, Hefei 230038, Anhui, China.,Anhui Provincial Key Laboratory for Chinese Herbal Compound, Anhui Academy of Chinese Medicine, Hefei 230012, China
| | - Kelong Ma
- Laboratory of Infection and Tumor, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 436 Room, Zhijing Building, No. 1 Qianjiang Road, Xinzhan District, Hefei 230012, Anhui, China.,Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Zhijing Building, No. 1 Qianjiang Road, Xinzhan District, Hefei 230012, Anhui, China.,Key Laboratory of Xin'An Medicine, Ministry of Education, Anhui Academy of Chinese Medicine, Xin'An Building, No. 103 Meishan Road, Shushan District, Hefei 230038, Anhui, China.,Anhui Provincial Key Laboratory for Chinese Herbal Compound, Anhui Academy of Chinese Medicine, Hefei 230012, China
| | - Guiming Yan
- Laboratory of Infection and Tumor, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 436 Room, Zhijing Building, No. 1 Qianjiang Road, Xinzhan District, Hefei 230012, Anhui, China.,Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Zhijing Building, No. 1 Qianjiang Road, Xinzhan District, Hefei 230012, Anhui, China.,Key Laboratory of Xin'An Medicine, Ministry of Education, Anhui Academy of Chinese Medicine, Xin'An Building, No. 103 Meishan Road, Shushan District, Hefei 230038, Anhui, China.,Anhui Provincial Key Laboratory for Chinese Herbal Compound, Anhui Academy of Chinese Medicine, Hefei 230012, China
| | - Tianming Wang
- Laboratory of Infection and Tumor, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 436 Room, Zhijing Building, No. 1 Qianjiang Road, Xinzhan District, Hefei 230012, Anhui, China.,Key Laboratory of Xin'An Medicine, Ministry of Education, Anhui Academy of Chinese Medicine, Xin'An Building, No. 103 Meishan Road, Shushan District, Hefei 230038, Anhui, China.,Anhui Provincial Key Laboratory for Chinese Herbal Compound, Anhui Academy of Chinese Medicine, Hefei 230012, China
| | - Daqiang Wu
- Laboratory of Infection and Tumor, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 436 Room, Zhijing Building, No. 1 Qianjiang Road, Xinzhan District, Hefei 230012, Anhui, China.,Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Zhijing Building, No. 1 Qianjiang Road, Xinzhan District, Hefei 230012, Anhui, China.,Key Laboratory of Xin'An Medicine, Ministry of Education, Anhui Academy of Chinese Medicine, Xin'An Building, No. 103 Meishan Road, Shushan District, Hefei 230038, Anhui, China.,Anhui Provincial Key Laboratory for Chinese Herbal Compound, Anhui Academy of Chinese Medicine, Hefei 230012, China
| | - Jing Shao
- Laboratory of Infection and Tumor, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, 436 Room, Zhijing Building, No. 1 Qianjiang Road, Xinzhan District, Hefei 230012, Anhui, China.,Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Zhijing Building, No. 1 Qianjiang Road, Xinzhan District, Hefei 230012, Anhui, China.,Key Laboratory of Xin'An Medicine, Ministry of Education, Anhui Academy of Chinese Medicine, Xin'An Building, No. 103 Meishan Road, Shushan District, Hefei 230038, Anhui, China.,Anhui Provincial Key Laboratory for Chinese Herbal Compound, Anhui Academy of Chinese Medicine, Hefei 230012, China
| |
Collapse
|
100
|
Minority report: the intestinal mycobiota in systemic infections. Curr Opin Microbiol 2020; 56:1-6. [PMID: 32599521 DOI: 10.1016/j.mib.2020.05.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/09/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022]
Abstract
Compared to bacteria, fungi often exhibit a lower abundance and a higher temporal volatility in the intestinal microbiota. Analysis of fungi in the microbiota (mycobiota) faces technical limitations with tools that were originally developed for analyzing bacteria. Dysbiotic states of the intestinal mycobiota, often associated with disruption of the healthy bacterial microbiota, are characterized by overgrowth (domination) of specific fungal taxa and loss of diversity. Intestinal domination by Candida species has been shown to be a major source of Candida bloodstream infections. Fungal dysbiosis is also linked to the development and treatment response in non-fungal infections, for example Clostridioides difficile colitis and HIV. Further research is needed to define the contribution of intestinal mycobiota to human fungal and non-fungal infections.
Collapse
|