1
|
Sumitomo T, Kawabata S. Respiratory tract barrier dysfunction in viral-bacterial co-infection cases. JAPANESE DENTAL SCIENCE REVIEW 2024; 60:44-52. [PMID: 38274948 PMCID: PMC10808858 DOI: 10.1016/j.jdsr.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/20/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
A preceding viral infection of the respiratory tract predisposes the host to secondary bacterial pneumonia, known as a major cause of morbidity and mortality. However, the underlying mechanism of the viral-bacterial synergy that leads to disease progression has remained elusive, thus hampering the production of effective prophylactic and therapeutic intervention options. In addition to viral-induced airway epithelial damage, which allows dissemination of bacteria to the lower respiratory tract and increases their invasiveness, dysfunction of immune defense following a viral infection has been implicated as a factor for enhanced susceptibility to secondary bacterial infections. Given the proximity of the oral cavity to the respiratory tract, where viruses enter and replicate, it is also well-established that oral health status can significantly influence the initiation, progression, and pathology of respiratory viral infections. This review was conducted to focus on the dysfunction of the respiratory barrier, which plays a crucial role in providing physical and secretory barriers as well as immune defense in the context of viral-bacterial synergy. Greater understanding of barrier response to viral-bacterial co-infections, will ultimately lead to development of effective, broad-spectrum therapeutic approaches for prevention of enhanced susceptibility to these pathogens.
Collapse
Affiliation(s)
- Tomoko Sumitomo
- Department of Oral Microbiology, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770–8504, Japan
| | - Shigetada Kawabata
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka 565–0871, Japan
| |
Collapse
|
2
|
Bravo E, Arce M, Herrera D, Sanz M. The Effect of Xanthohumol and Thymol on Candida albicans Filamentation and Its Impact on the Structure, Size, and Cell Viability of Biofilms Developed over Implant Surfaces. Cells 2024; 13:1877. [PMID: 39594625 PMCID: PMC11593281 DOI: 10.3390/cells13221877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/06/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
The aim of this in vitro study was to evaluate the effect of xanthohumol and thymol on the impact of Candida albicans on the structure, size and cell viability of subgingival biofilms formed on dental implant surfaces. The structure and microbial biomass of biofilms developed after 72 h, treated and untreated with both extracts, were compared by scanning electron microscopy (SEM) and confocal laser microscopy (CLSM). Quantitative polymerase chain reaction (qPCR) was used to quantify the number of viable and total microorganisms of each of the biofilm-forming strains in each condition. A general linear model was used to compare and validate the CLSM and qPCR results. The presence of xanthohumol and thymol during biofilm development inhibited the filamentous growth of C. albicans. The biofilm incubated with xanthohumol had significantly lower bacterial biomass and cell viability than the biofilm not exposed to the extract (p < 0.05). In contrast, these global parameters showed no differences when the biofilm was incubated with thymol. In the presence of xanthohumol, there was a decrease in counts and cell viability of Fusobacterium nucleatum, Porphyromonas gingivalis, and Aggregatibacter actinomycetemcomitans. Thymol treatment reduced the viability of F. nucleatum and P. gingivalis. The presence of these vegetable extracts during the development of a dynamic in vitro multispecies biofilm model inhibited the filamentous growth of C. albicans, partially reversing the effect that the fungus exerted on the structure, size and vitality of periodontopathogenic bacteria.
Collapse
Affiliation(s)
- Enrique Bravo
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group, Faculty of Dentistry, Complutense University, 28040 Madrid, Spain; (E.B.); (D.H.)
| | - Marion Arce
- Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Santiago 8380544, Chile;
| | - David Herrera
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group, Faculty of Dentistry, Complutense University, 28040 Madrid, Spain; (E.B.); (D.H.)
| | - Mariano Sanz
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group, Faculty of Dentistry, Complutense University, 28040 Madrid, Spain; (E.B.); (D.H.)
| |
Collapse
|
3
|
Shang L, Deng D, Krom BP, Gibbs S. Oral host-microbe interactions investigated in 3D organotypic models. Crit Rev Microbiol 2024; 50:397-416. [PMID: 37166371 DOI: 10.1080/1040841x.2023.2211665] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/12/2023]
Abstract
The oral cavity is inhabited by abundant microbes which continuously interact with the host and influence the host's health. Such host-microbe interactions (HMI) are dynamic and complex processes involving e.g. oral tissues, microbial communities and saliva. Due to difficulties in mimicking the in vivo complexity, it is still unclear how exactly HMI influence the transition between healthy status and disease conditions in the oral cavity. As an advanced approach, three-dimensional (3D) organotypic oral tissues (epithelium and mucosa/gingiva) are being increasingly used to study underlying mechanisms. These in vitro models were designed with different complexity depending on the research questions to be answered. In this review, we summarised the existing 3D oral HMI models, comparing designs and readouts, discussing applications as well as future perspectives.
Collapse
Affiliation(s)
- Lin Shang
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bastiaan P Krom
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Susan Gibbs
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Elnagar RM. Cross interaction between bacterial and fungal microbiota and their relevance to human health and disease: mechanistic pathways and prospective therapy. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 43:309-320. [PMID: 39364131 PMCID: PMC11444862 DOI: 10.12938/bmfh.2024-031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/27/2024] [Indexed: 10/05/2024]
Abstract
Diverse bacterial and fungal microbiota communities inhabit the human body, and their presence is essential for maintaining host homeostasis. The oral cavity, lung, gut, and vagina are just a few of the bodily cavities where these microorganisms communicate with one another, either directly or indirectly. The effects of this interaction can be either useful or detrimental to the host. When the healthy microbial diversity is disturbed, for instance, as a result of prolonged treatment with broad spectrum antibiotics, this allows the growth of specific microbes at the expense of others and alters their pathogenicity, causing a switch of commensal germs into pathogenic germs, which could promote tissue invasion and damage, as occurs in immunocompromised patients. Consequently, antimicrobials that specifically target pathogens may help in minimizing secondary issues that result from the disruption of useful bacterial/fungal interactions (BFIs). The interface between Candida albicans and Aspergillus fumigatus with bacteria at various body sites is emphasized in the majority of the medically important BFIs that have been reported thus far. This interface either supports or inhibits growth, or it enhances or blocks the generation of virulence factors. The aim of this review is to draw attention to the link between the bacterial and fungal microbiota and how they contribute to both normal homeostasis and disease development. Additionally, recent research that has studied microbiota as novel antimicrobials is summarized.
Collapse
Affiliation(s)
- Rasha Mokhtar Elnagar
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
- Medical Microbiology and Immunology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
5
|
Bittner-Eddy PD, Fischer LA, Parachuru PV, Costalonga M. MHC-II presentation by oral Langerhans cells impacts intraepithelial Tc17 abundance and Candida albicans oral infection via CD4 T cells. FRONTIERS IN ORAL HEALTH 2024; 5:1408255. [PMID: 38872986 PMCID: PMC11169704 DOI: 10.3389/froh.2024.1408255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/13/2024] [Indexed: 06/15/2024] Open
Abstract
In a murine model (LCΔMHC-II) designed to abolish MHC-II expression in Langerhans cells (LCs), ∼18% of oral LCs retain MHC-II, yet oral mucosal CD4 T cells numbers are unaffected. In LCΔMHC-II mice, we now show that oral intraepithelial conventional CD8αβ T cell numbers expand 30-fold. Antibody-mediated ablation of CD4 T cells in wild-type mice also resulted in CD8αβ T cell expansion in the oral mucosa. Therefore, we hypothesize that MHC class II molecules uniquely expressed on Langerhans cells mediate the suppression of intraepithelial resident-memory CD8 T cell numbers via a CD4 T cell-dependent mechanism. The expanded oral CD8 T cells co-expressed CD69 and CD103 and the majority produced IL-17A [CD8 T cytotoxic (Tc)17 cells] with a minority expressing IFN-γ (Tc1 cells). These oral CD8 T cells showed broad T cell receptor Vβ gene usage indicating responsiveness to diverse oral antigens. Generally supporting Tc17 cells, transforming growth factor-β1 (TGF-β1) increased 4-fold in the oral mucosa. Surprisingly, blocking TGF-β1 signaling with the TGF-R1 kinase inhibitor, LY364947, did not reduce Tc17 or Tc1 numbers. Nonetheless, LY364947 increased γδ T cell numbers and decreased CD49a expression on Tc1 cells. Although IL-17A-expressing γδ T cells were reduced by 30%, LCΔMHC-II mice displayed greater resistance to Candida albicans in early stages of oral infection. These findings suggest that modulating MHC-II expression in oral LC may be an effective strategy against fungal infections at mucosal surfaces counteracted by IL-17A-dependent mechanisms.
Collapse
Affiliation(s)
- Peter D. Bittner-Eddy
- Division of Basic Sciences, Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| | - Lori A. Fischer
- Division of Basic Sciences, Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| | - Praveen Venkata Parachuru
- Division of Periodontology, Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| | - Massimo Costalonga
- Division of Basic Sciences, Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
6
|
Vazquez-Munoz R, Thompson A, Sobue T, Dongari-Bagtzoglou A. Powder diet exacerbates oropharyngeal candidiasis in a mouse model. Appl Environ Microbiol 2024; 90:e0171323. [PMID: 38319097 PMCID: PMC10952443 DOI: 10.1128/aem.01713-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/04/2024] [Indexed: 02/07/2024] Open
Abstract
This study reports on the influence of a powder diet in a mouse model of oropharyngeal candidiasis (OPC), a significant health concern caused primarily by Candida albicans. Despite identical nutritional composition, we found that a powdered diet significantly increased Candida burdens and oral lesions, and aggravated weight loss compared to a standard pelleted diet. High fungal burdens and severe oral lesions were accomplished within 48 hours after infection with only one dose of cortisone. Moreover, mice on a powder diet recovered a week after infection. Using a powder diet, we thus modified the cortisone OPC murine model in a way that simplifies the infection process, enhances reproducibility, and facilitates studies investigating both pathogenesis and recovery processes. Our findings also underscore the pivotal role of the physical form of the diet in the progression and severity of oral Candida infection in this model. Future research should investigate this relationship further to broaden our understanding of the underlying mechanisms, potentially leading to novel prevention strategies and improved disease management.IMPORTANCEOropharyngeal candidiasis (OPC) is a multifactorial disease and a significant health concern. We found that the physical form of the diet plays a critical role in the severity and progression of OPC. We developed a modified cortisone OPC murine model that facilitates studies investigating pathogenesis and recovery processes.
Collapse
Affiliation(s)
- Roberto Vazquez-Munoz
- Department of General Dentistry, The University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Angela Thompson
- Department of General Dentistry, The University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Takanori Sobue
- Department of General Dentistry, The University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Anna Dongari-Bagtzoglou
- Department of General Dentistry, The University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
7
|
Monteiro JS, Kaushik K, de Arruda JAA, Georgakopoulou E, Vieira AT, Silva TA, Devadiga D, Anyanechi CE, Shetty S. Fungal footprints in oral cancer: unveiling the oral mycobiome. FRONTIERS IN ORAL HEALTH 2024; 5:1360340. [PMID: 38550775 PMCID: PMC10973146 DOI: 10.3389/froh.2024.1360340] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/14/2024] [Indexed: 11/12/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common type of head and neck cancer, with a high mortality rate. There is growing evidence supporting a link between oral cancer and the microbiome. The microbiome can impact various aspects of cancer, such as pathogenesis, diagnosis, treatment, and prognosis. While there is existing information on bacteria and its connection to oral cancer, the fungi residing in the oral cavity represent a significant component of the microbiome that remains in its early stages of exploration and understanding. Fungi comprise a minuscule part of the human microbiome called the mycobiome. Mycobiome is ubiquitous in the human body but a weakened immune system offers a leeway space for fungi to showcase its virulence. The role of mycobiome as a colonizer, facilitator, or driver of carcinogenesis is still ambiguous. Reactivating the mycobiome that undergoes collateral damage associated with cancer treatment can be watershed event in cancer research. The coordinated, virulent, non-virulent behavior of the fungi once they reach a critical density must be hacked, considering its diagnostic, prognostic and therapeutic implications in cancer. This review highlights the diversity of the mycobiome and its potential role in oral cancer.
Collapse
Affiliation(s)
- Jessica Sonal Monteiro
- Department of Oral and Maxillofacial Surgery, Manipal College of Dental Sciences Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Kriti Kaushik
- Department of Oral and Maxillofacial Surgery, Manipal College of Dental Sciences, Mangalore, India
| | - José Alcides Almeida de Arruda
- Department of Oral Diagnosis and Pathology, School of Dentistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eleni Georgakopoulou
- Laboratory of Histology-Embryology, Molecular Carcinogenesis Group, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Angelica Thomaz Vieira
- Laboratory of Microbiota and Immunomodulation, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tarcilia A. Silva
- Department of Oral Surgery, Pathology and Clinical Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Darshana Devadiga
- Department of Conservative Dentistry and Endodontics, AB Shetty Memorial Institute of Dental Sciences, NITTE (Deemed to be University), Mangalore, India
| | - Charles E. Anyanechi
- Department of Oral and Maxillofacial Surgery, University of Calabar/University of Calabar Teaching Hospital, Calabar, Nigeria
| | - Sameep Shetty
- Department of Oral and Maxillofacial Surgery, Manipal College of Dental Sciences, Mangalore, India
| |
Collapse
|
8
|
Hernández-Cabanyero C, Vonaesch P. Ectopic colonization by oral bacteria as an emerging theme in health and disease. FEMS Microbiol Rev 2024; 48:fuae012. [PMID: 38650052 PMCID: PMC11065354 DOI: 10.1093/femsre/fuae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/23/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024] Open
Abstract
The number of research papers published on the involvement of the oral microbiota in systemic diseases has grown exponentially over the last 4 years clearly demonstrating the growing interest in this field. Indeed, accumulating evidence highlights the central role of ectopic colonization by oral bacteria in numerous noncommunicable diseases including inflammatory bowel diseases (IBDs), undernutrition, preterm birth, neurological diseases, liver diseases, lung diseases, heart diseases, or colonic cancer. There is thus much interest in understanding the molecular mechanisms that lead to the colonization and maintenance of ectopic oral bacteria. The aim of this review is to summarize and conceptualize the current knowledge about ectopic colonization by oral bacteria, highlight wherever possible the underlying molecular mechanisms and describe its implication in health and disease. The focus lies on the newly discovered molecular mechanisms, showcasing shared pathophysiological mechanisms across different body sites and syndromes and highlighting open questions in the field regarding the pathway from oral microbiota dysbiosis to noncommunicable diseases.
Collapse
Affiliation(s)
- Carla Hernández-Cabanyero
- Department of Fundamental Microbiology, University of Lausanne, Biophore Building, UNIL-Sorge, 1015 Lausanne, Switzerland
| | - Pascale Vonaesch
- Department of Fundamental Microbiology, University of Lausanne, Biophore Building, UNIL-Sorge, 1015 Lausanne, Switzerland
| |
Collapse
|
9
|
Bloch S, Hager-Mair FF, Andrukhov O, Schäffer C. Oral streptococci: modulators of health and disease. Front Cell Infect Microbiol 2024; 14:1357631. [PMID: 38456080 PMCID: PMC10917908 DOI: 10.3389/fcimb.2024.1357631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
Streptococci are primary colonizers of the oral cavity where they are ubiquitously present and an integral part of the commensal oral biofilm microflora. The role oral streptococci play in the interaction with the host is ambivalent. On the one hand, they function as gatekeepers of homeostasis and are a prerequisite for the maintenance of oral health - they shape the oral microbiota, modulate the immune system to enable bacterial survival, and antagonize pathogenic species. On the other hand, also recognized pathogens, such as oral Streptococcus mutans and Streptococcus sobrinus, which trigger the onset of dental caries belong to the genus Streptococcus. In the context of periodontitis, oral streptococci as excellent initial biofilm formers have an accessory function, enabling late biofilm colonizers to inhabit gingival pockets and cause disease. The pathogenic potential of oral streptococci fully unfolds when their dissemination into the bloodstream occurs; streptococcal infection can cause extra-oral diseases, such as infective endocarditis and hemorrhagic stroke. In this review, the taxonomic diversity of oral streptococci, their role and prevalence in the oral cavity and their contribution to oral health and disease will be discussed, focusing on the virulence factors these species employ for interactions at the host interface.
Collapse
Affiliation(s)
- Susanne Bloch
- Competence Center for Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
- Department of Chemistry, Institute of Biochemistry, NanoGlycobiology Research Group, Universität für Bodenkultur Wien, Vienna, Austria
| | - Fiona F. Hager-Mair
- Department of Chemistry, Institute of Biochemistry, NanoGlycobiology Research Group, Universität für Bodenkultur Wien, Vienna, Austria
| | - Oleh Andrukhov
- Competence Center for Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Christina Schäffer
- Department of Chemistry, Institute of Biochemistry, NanoGlycobiology Research Group, Universität für Bodenkultur Wien, Vienna, Austria
| |
Collapse
|
10
|
Smirnov A, Yanushevich O, Krikheli N, Solis Pinargote NW, Peretyagin P, Grigoriev S, Alou L, Sevillano D, López-Piriz R, Guitian F, Bartolomé JF. 3Y-TZP/Ta Biocermet as a Dental Material: An Analysis of the In Vitro Adherence of Streptococcus Oralis Biofilm and an In Vivo Pilot Study in Dogs. Antibiotics (Basel) 2024; 13:175. [PMID: 38391561 PMCID: PMC10886202 DOI: 10.3390/antibiotics13020175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 02/24/2024] Open
Abstract
The surface adhesion of bacterial cells and the in vivo biocompatibility of a new ceramic-metal composite made of zirconium dioxide and tantalum were evaluated. Within the framework of an in vitro study using the crystal violet staining and colony counting methods, a relatively similar adhesion of Streptococcus oralis to the 3Y-TZP/Ta biocermet (roughness Ra = 0.12 ± 0.04 µm) and Ti-Al6-V4 titanium alloy (Ra = 0.04 ± 0.01 µm) was found. In addition, in an in vivo preliminary study focused on the histological analysis of a series of rods implanted in the jaws of beagle dogs for a six-month period, the absence of any fibrous tissue or inflammatory reaction at the interface between the implanted 3Y-TZP/Ta biocermets and the new bone was found. Thus, it can be concluded that the developed ceramic-metal biocomposite may be a promising new material for use in dentistry.
Collapse
Affiliation(s)
- Anton Smirnov
- Spark Plasma Sintering Research Laboratory, Moscow State University of Technology "STANKIN", Vadkovsky per. 1, Moscow 127055, Russia
| | - Oleg Yanushevich
- Scientific Department, A.I. Evdokimov Moscow State University of Medicine and Dentistry, Delegatskaya St., 20, p. 1, Moscow 127473, Russia
| | - Natella Krikheli
- Scientific Department, A.I. Evdokimov Moscow State University of Medicine and Dentistry, Delegatskaya St., 20, p. 1, Moscow 127473, Russia
| | - Nestor Washington Solis Pinargote
- Spark Plasma Sintering Research Laboratory, Moscow State University of Technology "STANKIN", Vadkovsky per. 1, Moscow 127055, Russia
| | - Pavel Peretyagin
- Spark Plasma Sintering Research Laboratory, Moscow State University of Technology "STANKIN", Vadkovsky per. 1, Moscow 127055, Russia
- Scientific Department, A.I. Evdokimov Moscow State University of Medicine and Dentistry, Delegatskaya St., 20, p. 1, Moscow 127473, Russia
| | - Sergey Grigoriev
- Spark Plasma Sintering Research Laboratory, Moscow State University of Technology "STANKIN", Vadkovsky per. 1, Moscow 127055, Russia
| | - Luis Alou
- Microbiology Department, School of Medicine, Universidad Complutense, Avda. Complutense s/n, 28040 Madrid, Spain
| | - David Sevillano
- Microbiology Department, School of Medicine, Universidad Complutense, Avda. Complutense s/n, 28040 Madrid, Spain
| | - Roberto López-Piriz
- Instituto de Cirugía Oral Avanzada-ICOA, Calle de Fray Luis de León, 14, 28012 Madrid, Spain
| | - Francisco Guitian
- Instituto de Materiales, iMATUS-USC, Santiago de Compostela, Avenida do Mestre Mateo 25, 15782 La Coruña, Spain
| | - José Florindo Bartolomé
- Instituto de Ciencia de Materiales de Madrid (ICMM), Consejo Superior de Investigaciones Científicas (CSIC), Campus de Cantoblanco, Calle Sor Juana Inés de la Cruz 3, 28049 Madrid, Spain
| |
Collapse
|
11
|
Khan F, Jeong GJ, Javaid A, Thuy Nguyen Pham D, Tabassum N, Kim YM. Surface adherence and vacuolar internalization of bacterial pathogens to the Candida spp. cells: Mechanism of persistence and propagation. J Adv Res 2023; 53:115-136. [PMID: 36572338 PMCID: PMC10658324 DOI: 10.1016/j.jare.2022.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/17/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The co-existence of Candida albicans with the bacteria in the host tissues and organs displays interactions at competitive, antagonistic, and synergistic levels. Several pathogenic bacteria take advantage of such types of interaction for their survival and proliferation. The chemical interaction involves the signaling molecules produced by the bacteria or Candida spp., whereas the physical attachment occurs by involving the surface proteins of the bacteria and Candida. In addition, bacterial pathogens have emerged to internalize inside the C. albicans vacuole, which is one of the inherent properties of the endosymbiotic relationship between the bacteria and the eukaryotic host. AIM OF REVIEW The interaction occurring by the involvement of surface protein from diverse bacterial species with Candida species has been discussed in detail in this paper. An in silico molecular docking study was performed between the surface proteins of different bacterial species and Als3P of C. albicans to explain the molecular mechanism involved in the Als3P-dependent interaction. Furthermore, in order to understand the specificity of C. albicans interaction with Als3P, the evolutionary relatedness of several bacterial surface proteins has been investigated. Furthermore, the environmental factors that influence bacterial pathogen internalization into the Candida vacuole have been addressed. Moreover, the review presented future perspectives for disrupting the cross-kingdom interaction and eradicating the endosymbiotic bacterial pathogens. KEY SCIENTIFIC CONCEPTS OF REVIEW With the involvement of cross-kingdom interactions and endosymbiotic relationships, the bacterial pathogens escape from the environmental stresses and the antimicrobial activity of the host immune system. Thus, the study of interactions between Candida and bacterial pathogens is of high clinical significance.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea.
| | - Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Aqib Javaid
- Department of Biotechnology and Bioinformatics, University of Hyderabad, India
| | - Dung Thuy Nguyen Pham
- Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
12
|
Lueyar TK, Karygianni L, Attin T, Thurnheer T. Dynamic interactions between Candida albicans and different streptococcal species in a multispecies oral biofilm. Microbiologyopen 2023; 12:e1381. [PMID: 37877656 PMCID: PMC10548025 DOI: 10.1002/mbo3.1381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 10/26/2023] Open
Abstract
The oral cavity is colonized by a plethora of bacteria, fungi, and archaea, including streptococci of the mitis group (MSG) and the yeast Candida albicans. This study aims to investigate the role of streptococcal species in the development of oral biofilm and the cross-kingdom interactions between some of the members of the commensal MSG and the pathogen yeast C. albicans using a multispecies supragingival biofilm model. A total of nine different in vitro biofilms were grown, quantified with culture analyses, and visually examined with confocal laser scanning microscopy (CLSM). A four-species biofilm without any streptococcal species was used as a basic biofilm. In each subsequent inoculum, one species of MSG was added and afterward combined with Streptococcus mutans. The eight-species biofilm contained all eight strains used in this study. Culture analyses showed that the presence of S. mutans in a four-species biofilm with Streptococcus oralis or S. oralis subsp. tigurinus did not differ significantly in C. albicans colony-forming unit (CFU) counts compared to biofilms without S. mutans. However, compared to other mitis species, Streptococcus gordonii combined with S. mutans resulted in the lowest CFUs of C. albicans. Visual observation by CLSM showed that biofilms containing both S. mutans and one species of MSG seemed to induce the formation of filamentous form of C. albicans. However, when several species of MSG were combined with S. mutans, C. albicans was again found in its yeast form.
Collapse
Affiliation(s)
- Tenzin Kunchok Lueyar
- Division of Clinical Oral Microbiology and Immunology, Clinic of Conservative and Preventive DentistryCenter of Dental Medicine, University of ZurichZurichSwitzerland
| | - Lamprini Karygianni
- Division of Clinical Oral Microbiology and Immunology, Clinic of Conservative and Preventive DentistryCenter of Dental Medicine, University of ZurichZurichSwitzerland
| | - Thomas Attin
- Division of Clinical Oral Microbiology and Immunology, Clinic of Conservative and Preventive DentistryCenter of Dental Medicine, University of ZurichZurichSwitzerland
| | - Thomas Thurnheer
- Division of Clinical Oral Microbiology and Immunology, Clinic of Conservative and Preventive DentistryCenter of Dental Medicine, University of ZurichZurichSwitzerland
| |
Collapse
|
13
|
Martorano-Fernandes L, Ricomini-Filho AP, Del Bel Cury AA. Does Streptococcus oralis supernatant influence on the proliferation and virulence of Candida albicans? Arch Oral Biol 2023; 154:105763. [PMID: 37437424 DOI: 10.1016/j.archoralbio.2023.105763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023]
Abstract
OBJECTIVE To evaluate the influence of Streptococcus oralis supernatant on the proliferation and virulence of Candida albicans. DESIGN S. oralis supernatant was obtained by filtration of overnight cultures. Single or dual-species cultures of C. albicans and S. oralis were cultivated in both planktonic and biofilm-based models. Planktonic culture growth was measured, and mature biofilms formed on resin disks were collected to measure biofilm metabolic activity, total biomass, and cell counts. Hyphae formation (virulence factor) and biofilm thickness were analyzed by confocal laser scanning microscopy. Data were analyzed by a one-way ANOVA test followed by the Tukey posthoc test (α = 0.05). RESULTS We found that S. oralis supernatant did not influence C. albicans proliferation in planktonic cultures. However, biofilms containing S. oralis supernatant showed higher cell metabolism than C. albicans monoculture biofilms and C. albicans-S. oralis dual-culture biofilms (p < 0.05). Though S. oralis supernatants did increase biofilm metabolic activity, they did not affect the total biomass and cell counts of C. albicans (p > 0.05). However, biofilm imaging revealed enhanced C. albicans hyphae formation in biofilms containing S. oralis supernatant compared to C. albicans monoculture biofilms. CONCLUSIONS Secreted metabolites in S. oralis supernatant may contribute to C. albicans metabolism and virulence.
Collapse
Affiliation(s)
- Loyse Martorano-Fernandes
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - Antônio Pedro Ricomini-Filho
- Department of Physiological Science, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - Altair Antoninha Del Bel Cury
- Department of Physiological Science, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil.
| |
Collapse
|
14
|
Lamont RJ, Hajishengallis G, Koo H. Social networking at the microbiome-host interface. Infect Immun 2023; 91:e0012423. [PMID: 37594277 PMCID: PMC10501221 DOI: 10.1128/iai.00124-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023] Open
Abstract
Microbial species colonizing host ecosystems in health or disease rarely do so alone. Organisms conglomerate into dynamic heterotypic communities or biofilms in which interspecies and interkingdom interactions drive functional specialization of constituent species and shape community properties, including nososymbiocity or pathogenic potential. Cell-to-cell binding, exchange of signaling molecules, and nutritional codependencies can all contribute to the emergent properties of these communities. Spatial constraints defined by community architecture also determine overall community function. Multilayered interactions thus occur between individual pairs of organisms, and the relative impact can be determined by contextual cues. Host responses to heterotypic communities and impact on host surfaces are also driven by the collective action of the community. Additionally, the range of interspecies interactions can be extended by bacteria utilizing host cells or host diet to indirectly or directly influence the properties of other organisms and the community microenvironment. In contexts where communities transition to a dysbiotic state, their quasi-organismal nature imparts adaptability to nutritional availability and facilitates resistance to immune effectors and, moreover, exploits inflammatory and acidic microenvironments for their persistence.
Collapse
Affiliation(s)
- Richard J. Lamont
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, Kentucky, USA
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hyun Koo
- Department of Orthodontics and Divisions of Pediatric Dentistry and Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
15
|
Vazquez-Munoz R, Thompson A, Sobue T, Dongari-Bagtzoglou A. A prebiotic diet modulates the oral microbiome composition and results in the attenuation of oropharyngeal candidiasis in mice. Microbiol Spectr 2023; 11:e0173423. [PMID: 37671879 PMCID: PMC10580959 DOI: 10.1128/spectrum.01734-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/06/2023] [Indexed: 09/07/2023] Open
Abstract
Oral bacteria can influence the ability of Candida albicans to cause oropharyngeal candidiasis (OPC). We recently reported that a Lactobacillus johnsonii-enriched oral microbiota reduced C. albicans virulence in an immunosuppressed OPC mouse model. As a follow-up, in this work, we aimed to enrich the resident oral Lactobacillus communities with a prebiotic diet to further assess their effect on the severity of OPC. We tested the effect of a prebiotic xylo-oligosaccharides (XOS)-enriched diet in the oral global bacterial composition and severity of OPC. We assessed changes in the oral microbiome composition via 16S-rRNA gene high-throughput sequencing, validated by qPCR. The impact of the prebiotic diet on Candida infection was assessed by quantifying changes in oral fungal and bacterial biomass and scoring tongue lesions. Contrary to expectations, oral Lactobacillus communities were not enriched by the XOS-supplemented diet. Yet, XOS modulated the oral microbiome composition, increasing Bifidobacterium abundance and reducing enterococci and staphylococci. In the OPC model, the XOS diet attenuated Candida virulence and bacterial dysbiosis, increasing lactobacilli and reducing enterococci on the oral mucosa. We conclude that XOS attenuates Candida virulence by promoting a bacterial microbiome structure more resilient to Candida infection. IMPORTANCE This is the first study on the effects of a prebiotic diet on the oral mucosal bacterial microbiome and an oropharyngeal candidiasis (OPC) mouse model. We found that xylo-oligosaccharides change the oral bacterial community composition and attenuate OPC. Our results contribute to the understanding of the impact of the oral bacterial communities on Candida virulence.
Collapse
Affiliation(s)
- Roberto Vazquez-Munoz
- Department of General Dentistry, The University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Angela Thompson
- Department of General Dentistry, The University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Takanori Sobue
- Department of General Dentistry, The University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Anna Dongari-Bagtzoglou
- Department of General Dentistry, The University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
16
|
Chen X, Wang J, Chen J, Wang G, Zhang R, Qiu J. Vaginal homeostasis features of Vulvovaginal Candidiasis through vaginal metabolic profiling. Med Mycol 2023; 61:myad085. [PMID: 37573133 DOI: 10.1093/mmy/myad085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 07/25/2023] [Accepted: 08/11/2023] [Indexed: 08/14/2023] Open
Abstract
Vulvovaginal candidiasis (VVC) is an inflammatory disease primarily infected by Candida albicans. The condition has good short-term treatment effects, high recurrence, and seriously affects the quality of life of women. Metabolomics has been applied to research a variety of inflammatory diseases. In the present study, the vaginal metabolic profiles of VVC patients and healthy populations (Cnotrol (CTL)) were explored by a non-targeted metabolomics approach. In total, 211 differential metabolites were identified, with the VVC group having 128 over-expressed and 83 under-expressed metabolites compared with healthy individuals. Functional analysis showed that these metabolites were mainly involved in amino acid metabolism and lipid metabolism. In addition, network software analysis indicated that the differential metabolites were associated with mitogen-activated protein kinase (MAPK) signaling and NF-κB signaling. Further molecular docking suggested that linoleic acid can bind to the acyl-CoA synthetase 1 (ACSL1) protein, which has been shown to be associated with multiple inflammatory diseases and is an upstream regulator of the MAPK and NF-κB signaling pathways that mediate inflammation. Therefore, our preliminary analysis results suggest that VVC has a unique metabolic profile. Linoleic acid, a significantly elevated unsaturated fatty acid in the VVC group, may promote VVC development through the ACSL1/MAPK and ACSL1/NF-κB signaling pathways. This study's findings contribute to further exploring the mechanism of VVC infection and providing new perspectives for the treatment of Candida albicans vaginal infection.
Collapse
Affiliation(s)
- Xinyi Chen
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinbo Wang
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Chen
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanghua Wang
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runjie Zhang
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Qiu
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Yang L, Cheng T, Shao J. Perspective on receptor-associated immune response to Candida albicans single and mixed infections: Implications for therapeutics in oropharyngeal candidiasis. Med Mycol 2023; 61:myad077. [PMID: 37533203 DOI: 10.1093/mmy/myad077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/11/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023] Open
Abstract
Oropharyngeal candidiasis (OPC), commonly known as 'thrush', is an oral infection that usually dismantles oral mucosal integrity and malfunctions local innate and adaptive immunities in compromised individuals. The major pathogen responsible for the occurrence and progression of OPC is the dimorphic opportunistic commensal Candida albicans. However, the incidence induced by non-albicans Candida species including C. glabrata, C. tropicalis, C. dubliniensis, C. parapsilosis, and C. krusei are increasing in company with several oral bacteria, such as Streptococcus mutans, S. gordonii, S. epidermidis, and S. aureus. In this review, the microbiological and infection features of C. albicans and its co-contributors in the pathogenesis of OPC are outlined. Since the invasion and concomitant immune response lie firstly on the recognition of oral pathogens through diverse cellular surface receptors, we subsequently emphasize the roles of epidermal growth factor receptor, ephrin-type receptor 2, human epidermal growth factor receptor 2, and aryl hydrocarbon receptor located on oral epithelial cells to delineate the underlying mechanism by which host immune recognition to oral pathogens is mediated. Based on these observations, the therapeutic approaches to OPC comprising conventional and non-conventional antifungal agents, fungal vaccines, cytokine and antibody therapies, and antimicrobial peptide therapy are finally overviewed. In the face of newly emerging life-threatening microbes (C. auris and SARS-CoV-2), risks (biofilm formation and interconnected translocation among diverse organs), and complicated clinical settings (HIV and oropharyngeal cancer), the research on OPC is still a challenging task.
Collapse
Affiliation(s)
- Liu Yang
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, P. R. China
| | - Ting Cheng
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, P. R. China
| | - Jing Shao
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, P. R. China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Zhijing Building, 350 Longzihu Road, Xinzhan District, Hefei 230012, P. R. China
| |
Collapse
|
18
|
Kulshrestha A, Gupta P. Combating polymicrobial biofilm: recent approaches. Folia Microbiol (Praha) 2023:10.1007/s12223-023-01070-y. [PMID: 37310652 DOI: 10.1007/s12223-023-01070-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/04/2023] [Indexed: 06/14/2023]
Abstract
The polymicrobial biofilm (PMBF) is formed when microbes from multiple species co-aggregate into an envelope made of extra polymeric substances (EPS) that keep the microbes safe from external stresses. The formation of PMBF has been linked to a variety of human infections, including cystic fibrosis, dental caries, urinary tract infections, etc. Multiple microbial species co-aggregation during an infection results in a recalcitrant biofilm formation, which is a seriously threatening phenomenon. It is challenging to treat polymicrobial biofilms since they contain multiple microbes which show drug resistance to various antibiotics/antifungals. The present study discusses various approaches by which an antibiofilm compound works. Depending on their mode of action, antibiofilm compounds can block the adhesion of cells to one another, modify membranes/walls, or disrupt quorum-sensing systems.
Collapse
Affiliation(s)
- Anmol Kulshrestha
- Department of Biotechnology, National Institute of Technology, Raipur, India
| | - Pratima Gupta
- Department of Biotechnology, National Institute of Technology, Raipur, India.
| |
Collapse
|
19
|
Chow EWL, Mei Pang L, Wang Y. Impact of the host microbiota on fungal infections: new possibilities for intervention? Adv Drug Deliv Rev 2023; 198:114896. [PMID: 37211280 DOI: 10.1016/j.addr.2023.114896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/23/2023]
Abstract
Many human fungal pathogens are opportunistic. They are primarily benign residents of the human body and only become infectious when the host's immunity and microbiome are compromised. Bacteria dominate the human microbiome, playing an essential role in keeping fungi harmless and acting as the first line of defense against fungal infection. The Human Microbiome Project, launched by NIH in 2007, has stimulated extensive investigation and significantly advanced our understanding of the molecular mechanisms governing the interaction between bacteria and fungi, providing valuable insights for developing future antifungal strategies by exploiting the interaction. This review summarizes recent progress in this field and discusses new possibilities and challenges. We must seize the opportunities presented by researching bacterial-fungal interplay in the human microbiome to address the global spread of drug-resistant fungal pathogens and the drying pipelines of effective antifungal drugs.
Collapse
Affiliation(s)
- Eve W L Chow
- A*STAR Infectious Diseases Laboratories (ID Labs), Agency for Science and Technology Research (A*STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648
| | - Li Mei Pang
- A*STAR Infectious Diseases Laboratories (ID Labs), Agency for Science and Technology Research (A*STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648
| | - Yue Wang
- A*STAR Infectious Diseases Laboratories (ID Labs), Agency for Science and Technology Research (A*STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore.
| |
Collapse
|
20
|
Wijesinghe GK, Nobbs AH, Bandara HMHN. Cross-kingdom Microbial Interactions Within the Oral Cavity and Their Implications for Oral Disease. CURRENT CLINICAL MICROBIOLOGY REPORTS 2023. [DOI: 10.1007/s40588-023-00191-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Abstract
Purpose of Review
This review serves to highlight the cross-kingdom interactions that can occur within the human oral cavity between fungus Candida albicans and oral bacteria, and their impact on the delicate balance between oral health and disease.
Recent Findings
A growing number of physical, chemical, and metabolic networks have been identified that underpin these cross-kingdom interactions. Moreover, these partnerships are often synergistic and can modulate microbial burden or virulence. This, in turn, can drive the onset or progression of oral diseases such as dental caries, periodontitis, denture-associated stomatitis, and oral cancer.
Summary
The impact of cross-kingdom interactions on the cellular, biochemical, and communal composition of oral microbial biofilms is increasingly clear. With growing insight into these processes at the molecular level, so this knowledge can be used to better inform the development of novel strategies to manipulate the oral microbiota to promote oral health and combat oral disease.
Collapse
|
21
|
Kingsley C, Kourtidis A. Critical roles of adherens junctions in diseases of the oral mucosa. Tissue Barriers 2023; 11:2084320. [PMID: 35659464 PMCID: PMC10161952 DOI: 10.1080/21688370.2022.2084320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/18/2022] [Accepted: 05/26/2022] [Indexed: 10/18/2022] Open
Abstract
The oral cavity is directly exposed to a variety of environmental stimuli and contains a diverse microbiome that continuously interacts with the oral epithelium. Therefore, establishment and maintenance of the barrier function of the oral mucosa is of paramount importance for its function and for the body's overall health. The adherens junction is a cell-cell adhesion complex that is essential for epithelial barrier function. Although a considerable body of work has associated barrier disruption with oral diseases, the molecular underpinnings of these associations have not been equally investigated. This is critical, since adherens junction components also possess significant signaling roles in the cell, in addition to their architectural ones. Here, we summarize current knowledge involving adherens junction components in oral pathologies, such as cancer and oral pathogen-related diseases, while we also discuss gaps in the knowledge and opportunities for future investigation of the relationship between adherens junctions and oral diseases.
Collapse
Affiliation(s)
- Christina Kingsley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Antonis Kourtidis
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
22
|
Archambault L, Koshy-Chenthittayil S, Thompson A, Dongari-Bagtzoglou A, Laubenbacher R, Mendes P. Corrected and Republished from: "Understanding Lactobacillus paracasei and Streptococcus oralis Biofilm Interactions through Agent-Based Modeling". mSphere 2023; 8:e0065622. [PMID: 36942961 DOI: 10.1128/msphere.00656-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
As common commensals residing on mucosal tissues, Lactobacillus species are known to promote health, while some Streptococcus species act to enhance the pathogenicity of other organisms in those environments. In this study we used a combination of in vitro imaging of live biofilms and computational modeling to explore biofilm interactions between Streptococcus oralis, an accessory pathogen in oral candidiasis, and Lactobacillus paracasei, an organism with known probiotic properties. A computational agent-based model was created where the two species interact only by competing for space, oxygen, and glucose. Quantification of bacterial growth in live biofilms indicated that S. oralis biomass and cell numbers were much lower than predicted by the model. Two subsequent models were then created to examine more complex interactions between these species, one where L. paracasei secretes a surfactant and another where L. paracasei secretes an inhibitor of S. oralis growth. We observed that the growth of S. oralis could be affected by both mechanisms. Further biofilm experiments support the hypothesis that L. paracasei may secrete an inhibitor of S. oralis growth, although they do not exclude that a surfactant could also be involved. This contribution shows how agent-based modeling and experiments can be used in synergy to address multiple-species biofilm interactions, with important roles in mucosal health and disease. IMPORTANCE We previously discovered a role of the oral commensal Streptococcus oralis as an accessory pathogen. S. oralis increases the virulence of Candida albicans infections in murine oral candidiasis and epithelial cell models through mechanisms which promote the formation of tissue-damaging biofilms. Lactobacillus species have known inhibitory effects on biofilm formation of many microbes, including Streptococcus species. Agent-based modeling has great advantages as a means of exploring multifaceted relationships between organisms in complex environments such as biofilms. Here, we used an iterative collaborative process between experimentation and modeling to reveal aspects of the mostly unexplored relationship between S. oralis and L. paracasei in biofilm growth. The inhibitory nature of L. paracasei on S. oralis in biofilms may be exploited as a means of preventing or alleviating mucosal fungal infections.
Collapse
Affiliation(s)
- Linda Archambault
- Center for Quantitative Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, Farmington, Connecticut, USA
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Sherli Koshy-Chenthittayil
- Center for Quantitative Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Angela Thompson
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, Farmington, Connecticut, USA
| | - Anna Dongari-Bagtzoglou
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, Farmington, Connecticut, USA
| | | | - Pedro Mendes
- Center for Quantitative Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
23
|
Bagirova NS, Petukhova IN, Grigorievskaya ZV, Sytov AV, Slukin PV, Goremykina EA, Khokhlova OE, Fursova NK, Kazimov AE. Oral microbiota in patients with oropharyngeal cancer with an emphasis on <i>Candida</i> spp. HEAD AND NECK TUMORS (HNT) 2022. [DOI: 10.17650/2222-1468-2022-12-3-71-85] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction. Interactions between the 2 microbiota components – bacteria and fungi – are of interest as diagnostic and prognostic markers in selection of treatment tactics for oncological patients.Aim. To study microbiota of the oral cavity in patients with primary squamous cell carcinoma of the oropharyngeal area before and after surgical intervention to find biomarkers for rational selection of antifungal drugs.Materials and methods. At the Surgical Department of Head and Neck Tumors of the N. N. Blokhin National Research Center of Oncology, three-component study was performed: investigations of spectrum of Candida spp. isolates, Candida spp. strains’ resistance to antifungals, and oral washes in primary patients before and after surgery. mALDI-Tof microflex LT (Biotyper, Bruker Daltonics, germany) was used for strain identification; Sensititre Yeast ONE, YO10 (Trek Diagnostic System, united kingdom) plates were used for determination of minimal inhibiting concentrations of anti fungals. values of minimal inhibiting concentrations were evaluated based on the European Committee on Antimicrobial Susceptibility Testing (EuCAST) criteria (version 10.0).Results. four-year observation of patients at the surgical department of head and neck tumors of the N. N. Blokhin National Research Center of Oncology showed that the most common species of Candida is C. albicans (73.5 % of cases). Candida spp. resistance to antifungals was detected only for fluconazole (9.3 % of cases) and micafungin (8.0 % of cases), mostly among C. albicans strains. In 31.8 % of primary patients, oral washes prior to surgery showed growth of Candida spp. (probably, tissue colonization). After surgical intervention, Candida spp. growth was detected in 36.4 % of cases, only 1 of which was diagnosed as invasive mycosis. In 54.5 % of cases before and in 72.7 % of cases after surgery, gram-negative rods were detected. After surgical intervention, percentage of enterobacteria and non-fermenters significantly increased: 59.1 % versus 27.3 % (p <0.05) and 63.6 % versus 27.3 % (p <0.02), respectively. prior to surgery, non-fermenting gram-negative bacteria were represented only by P. aeruginosa; after surgery, the spectrum of non-fermenting gram-negative bacteria became wider but percentage of P. aeruginosa remained high: 71.4 %. ERG11 gene was identified only in 1 strain: C. albicans. FKS1 gene also was identified only in 1 strain: C. inconspicua. virulence factor genes were detected in 57.1 % of strains.Conclusion. Surgical intervention is associated with changes in bacterial microbiota but not fugal microbiota. presence of virulence factor genes and resistance genes in Candida spp. strains should be considered a biomarker allowing to differentiate between colonization and candida infection and can be used for rational selection of antifungal drugs in prevention and treatment of invasive candidiasis, especially in the absence of criteria for interpretation of measured minimal inhibiting concentrations of antifungals.
Collapse
Affiliation(s)
- N. S. Bagirova
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia
| | - I. N. Petukhova
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia
| | - Z. V. Grigorievskaya
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia
| | - A. V. Sytov
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia
| | - P. V. Slukin
- State Scientific Center of Applied Microbiology and Biotechnology of Rospotrebnadzor
| | - E. A. Goremykina
- State Scientific Center of Applied Microbiology and Biotechnology of Rospotrebnadzor; Pushchinsky State Natural Science Institute
| | - O. E. Khokhlova
- State Scientific Center of Applied Microbiology and Biotechnology of Rospotrebnadzor; Pushchinsky State Natural Science Institute
| | - N. K. Fursova
- State Scientific Center of Applied Microbiology and Biotechnology of Rospotrebnadzor; Pushchinsky State Natural Science Institute
| | - A. E. Kazimov
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia
| |
Collapse
|
24
|
Bertolini M, Costa RC, Barão VAR, Cunha Villar C, Retamal-Valdes B, Feres M, Silva Souza JG. Oral Microorganisms and Biofilms: New Insights to Defeat the Main Etiologic Factor of Oral Diseases. Microorganisms 2022; 10:microorganisms10122413. [PMID: 36557666 PMCID: PMC9781395 DOI: 10.3390/microorganisms10122413] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 07/19/2022] [Indexed: 12/12/2022] Open
Abstract
The oral cavity presents a highly diverse community of microorganisms due to the unique environmental conditions for microbial adhesion and growth [...].
Collapse
Affiliation(s)
- Martinna Bertolini
- Department of Periodontics and Preventive Dentistry, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA 15106, USA
- Correspondence:
| | - Raphael Cavalcante Costa
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba 13083-970, SP, Brazil
| | - Valentim Adelino Ricardo Barão
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba 13083-970, SP, Brazil
| | - Cristina Cunha Villar
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-010, SP, Brazil
| | | | - Magda Feres
- Dental Research Division, Guarulhos University, Guarulhos 05508-010, SP, Brazil
- Center for Clinical and Translational Research, Forsyth Institute, Boston, MA 02142, USA
| | - João Gabriel Silva Souza
- Dental Research Division, Guarulhos University, Guarulhos 05508-010, SP, Brazil
- Dental Science School (Faculdade de Ciências Odontológicas—FCO), Montes Claros 39401-303, MG, Brazil
- Oncovida Cancer Research Center, Montes Claros 39400-111, MG, Brazil
| |
Collapse
|
25
|
Du Q, Ren B, Zhou X, Zhang L, Xu X. Cross-kingdom interaction between Candida albicans and oral bacteria. Front Microbiol 2022; 13:911623. [PMID: 36406433 PMCID: PMC9668886 DOI: 10.3389/fmicb.2022.911623] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 09/28/2022] [Indexed: 08/27/2023] Open
Abstract
Candida albicans is a symbiotic fungus that commonly colonizes on oral mucosal surfaces and mainly affects immuno-compromised individuals. Polymicrobial interactions between C. albicans and oral microbes influence the cellular and biochemical composition of the biofilm, contributing to change clinically relevant outcomes of biofilm-related oral diseases, such as pathogenesis, virulence, and drug-resistance. Notably, the symbiotic relationships between C. albicans and oral bacteria have been well-documented in dental caries, oral mucositis, endodontic and periodontal diseases, implant-related infections, and oral cancer. C. albicans interacts with co-existing oral bacteria through physical attachment, extracellular signals, and metabolic cross-feeding. This review discusses the bacterial-fungal interactions between C. albicans and different oral bacteria, with a particular focus on the underlying mechanism and its relevance to the development and clinical management of oral diseases.
Collapse
Affiliation(s)
- Qian Du
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Biao Ren
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Zhang
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
26
|
Higa B, Cintra BS, Álvarez CM, Ribeiro AB, Ferreira JC, Tavares DC, Enriquez V, Martinez LR, Pires RH. Ozonated oil is effective at killing Candida species and Streptococcus mutans biofilm-derived cells under aerobic and microaerobic conditions. Med Mycol 2022; 60:myac055. [PMID: 35869980 PMCID: PMC9359064 DOI: 10.1093/mmy/myac055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/12/2022] [Accepted: 07/20/2022] [Indexed: 07/25/2023] Open
Abstract
This study explores the growth of bacterial, fungal, and interkingdom biofilms under aerobiosis or microaerobic conditions and the effect of ozonated sunflower oil on these biofilms. Candida species and Streptococcus mutans were used to study this interaction due to their importance in oral health and disease as these microorganisms display a synergistic relationship that manifests in the onset of caries and tooth decay. Biofilms were developed in a 96-well microtiter plate at 37ºC for 24 h, under aerobiosis or microaerobic conditions, and treated with ozonated oil for 5 to 120 min. All the microorganisms formed biofilms in both oxygenation conditions. Scanning electron microscopy was used to visualize biofilm morphology. Rodent experiments were performed to verify the oil-related toxicity and its efficacy in oral candidiasis. The growth of all Candida species was increased when co-cultured with S. mutans, whilst the growth of bacterium was greater only when co-cultured with C. krusei and C. orthopsilosis under aerobiosis and microaerobic conditions, respectively. Regardless of the oxygenation condition, ozonated oil significantly reduced the viability of all the tested biofilms and infected mice, showing remarkable microbicidal activity as corroborated with confocal microscopy and minimal toxicity. Thus, ozonated oil therapy can be explored as a strategy to control diseases associated with these biofilms especially in the oral cavity. LAY SUMMARY We demonstrated that ozonated sunflower oil is effective at killing the biofilms formed by Candida species, by the bacterium Streptococcus mutans, or by both micoorganisms that can interact in the oral cavity, making it a potential therapeutic option for the treatment of these infections.
Collapse
Affiliation(s)
- Barbara Higa
- Laboratory of Mycology and Environmental Diagnosis, Universidade de Franca, Franca, São Paulo 14.404-600, Brazil
| | - Bianca Souza Cintra
- Laboratory of Mycology and Environmental Diagnosis, Universidade de Franca, Franca, São Paulo 14.404-600, Brazil
- Postgraduate Program in Animal Science, Universidade de Franca, Franca, São Paulo 14.404-600, Brazil
| | - Carmen Magaly Álvarez
- Laboratory of Mycology and Environmental Diagnosis, Universidade de Franca, Franca, São Paulo 14.404-600, Brazil
- Faculty of Veterinary Medicine and Zootechnics, Universidad Agraria del Ecuador, Guayaquil 090101, Ecuador
| | | | - Jair Camargo Ferreira
- Postgraduate Program in Animal Science, Universidade de Franca, Franca, São Paulo 14.404-600, Brazil
| | | | - Vanessa Enriquez
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida 32610, USA
| | - Luis R Martinez
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida 32610, USA
- Emerging Pathogens Institute, Center for Immunology and Transplantation, and Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida 32610, USA
| | - Regina Helena Pires
- To whom correspondence should be addressed. Dr. Regina Helena Pires, Laboratory of Mycology and Environmental Diagnosis, Universidade de Franca, 201 Dr. Armando Salles Oliveira Ave, Franca, SP, 14.404-600, Brazil. Tel.: +55-16-3711-8945; E-mail:
| |
Collapse
|
27
|
Le Bars P, Kouadio AA, Bandiaky ON, Le Guéhennec L, de La Cochetière MF. Host's Immunity and Candida Species Associated with Denture Stomatitis: A Narrative Review. Microorganisms 2022; 10:microorganisms10071437. [PMID: 35889156 PMCID: PMC9323190 DOI: 10.3390/microorganisms10071437] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 02/04/2023] Open
Abstract
Denture-related Candida stomatitis, which has been described clinically in the literature, is either localized or generalized inflammation of the oral mucosa in connection with a removable prosthesis. During this inflammatory process, the mycobacterial biofilm and the host’s immune response play an essential role. Among microorganisms of this mixed biofilm, the Candida species proliferates easily and changes from a commensal to an opportunistic pathogen. In this situation, the relationship between the Candida spp. and the host is influenced by the presence of the denture and conditioned both by the immune response and the oral microbiota. Specifically, this fungus is able to hijack the innate immune system of its host to cause infection. Additionally, older edentulous wearers of dentures may experience an imbalanced and decreased oral microbiome diversity. Under these conditions, the immune deficiency of these aging patients often promotes the spread of commensals and pathogens. The present narrative review aimed to analyze the innate and adaptive immune responses of patients with denture stomatitis and more particularly the involvement of Candida albicans sp. associated with this pathology.
Collapse
Affiliation(s)
- Pierre Le Bars
- Department of Prosthetic Dentistry, Faculty of Dentistry, Nantes University, 1 Place Alexis Ricordeau, 44042 Nantes, France; (A.A.K.); (O.N.B.); (L.L.G.)
- Correspondence: authors:
| | - Alain Ayepa Kouadio
- Department of Prosthetic Dentistry, Faculty of Dentistry, Nantes University, 1 Place Alexis Ricordeau, 44042 Nantes, France; (A.A.K.); (O.N.B.); (L.L.G.)
- Department of Prosthetic Dentistry, Faculty of Dentistry, CHU, Abidjan P.O. Box 612, Côte d’Ivoire
| | - Octave Nadile Bandiaky
- Department of Prosthetic Dentistry, Faculty of Dentistry, Nantes University, 1 Place Alexis Ricordeau, 44042 Nantes, France; (A.A.K.); (O.N.B.); (L.L.G.)
| | - Laurent Le Guéhennec
- Department of Prosthetic Dentistry, Faculty of Dentistry, Nantes University, 1 Place Alexis Ricordeau, 44042 Nantes, France; (A.A.K.); (O.N.B.); (L.L.G.)
| | - Marie-France de La Cochetière
- EA 3826 Thérapeutiques Cliniques Et expérimentales des Infections, Faculté de Médecine, CHU Hôtel-Dieu, Université de Nantes, 1, rue G. Veil, 44000 Nantes, France;
| |
Collapse
|
28
|
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
|
29
|
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: 23] [Impact Index Per Article: 7.7] [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
|
30
|
Fultz R, Ticer T, Glover J, Stripe L, Engevik MA. Select Streptococci Can Degrade Candida Mannan To Facilitate Growth. Appl Environ Microbiol 2022; 88:e0223721. [PMID: 34936835 PMCID: PMC8863070 DOI: 10.1128/aem.02237-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/19/2021] [Indexed: 11/20/2022] Open
Abstract
Multiple studies have found that streptococci have a synergistic relationship with Candida species, but the details of these interactions are still being discovered. Candida species are covered by mannan, a polymer of mannose, which could serve as a carbon source for certain microbes. We hypothesized that streptococci that possess mannan-degrading glycosyl hydrolases would be able to enzymatically cleave mannose residues, which could serve as a primary carbohydrate source to support growth. We analyzed 90 streptococcus genomes to predict the capability of streptococci to transport and utilize mannose and to degrade diverse mannose linkages found on mannan. The genome analysis revealed mannose transporters and downstream pathways in most streptococci, but only <50% of streptococci harbored the glycosyl hydrolases required for mannan degradation. To confirm the ability of streptococci to use mannose or mannan, we grew 6 representative streptococci in a chemically defined medium lacking glucose supplemented with mannose, yeast extract, or purified mannan isolated from Candida and Saccharomyces strains. Although all tested Streptococcus strains could use mannose, Streptococcus salivarius and Streptococcus agalactiae, which did not possess mannan-degrading glycosyl hydrolases, could not use yeast extract or mannan to enhance their growth. In contrast, we found that Streptococcus mitis, Streptococcus parasanguinis, Streptococcus sanguinis, and Streptococcus pyogenes possessed the necessary glycosyl hydrolases to use yeast extract and isolated mannan, which promoted robust growth. Our data indicate that several streptococci are capable of degrading fungal mannans and harvesting mannose for energy. IMPORTANCE This work highlights a previously undescribed aspect of streptococcal Candida interactions. Our work identifies that certain streptococci possess the enzymes required to degrade mannan, and through this mechanism, they can release mannose residues from the cell wall of fungal species and use them as a nutrient source. We speculate that streptococci that can degrade fungal mannan may have a competitive advantage for colonization. This finding has broad implications for human health, as streptococci and Candida are found at multiple body sites.
Collapse
Affiliation(s)
- Robert Fultz
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, Texas, USA
| | - Taylor Ticer
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Janiece Glover
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Leah Stripe
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Melinda A. Engevik
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
31
|
Understanding Lactobacillus paracasei and Streptococcus oralis Biofilm Interactions through Agent-Based Modeling. mSphere 2021; 6:e0087521. [PMID: 34908459 PMCID: PMC8673396 DOI: 10.1128/msphere.00875-21] [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] [Indexed: 11/20/2022] Open
Abstract
As common commensals residing on mucosal tissues, Lactobacillus species are known to promote health, while some Streptococcus species act to enhance the pathogenicity of other organisms in those environments. In this study, we used a combination of in vitro imaging of live biofilms and computational modeling to explore biofilm interactions between Streptococcus oralis, an accessory pathogen in oral candidiasis, and Lactobacillus paracasei, an organism with known probiotic properties. A computational agent-based model was created where the two species interact only by competing for space, oxygen and glucose. Quantification of bacterial growth in live biofilms indicated that S. oralis biomass and cell numbers were much lower than predicted by the model. Two subsequent models were then created to examine more complex interactions between these species, one where L. paracasei secretes a surfactant, and another where L. paracasei secretes an inhibitor of S. oralis growth. We observed that the growth of S. oralis could be affected by both mechanisms. Further biofilm experiments support the hypothesis that L. paracasei may secrete an inhibitor of S. oralis growth, although they do not exclude that a surfactant could also be involved. This contribution shows how agent-based modeling and experiments can be used in synergy to address multiple species biofilm interactions, with important roles in mucosal health and disease. IMPORTANCE We previously discovered a role of the oral commensal Streptococcus oralis as an accessory pathogen. S. oralis increases the virulence of Candida albicans infections in murine oral candidiasis and epithelial cell models through mechanisms which promote the formation of tissue-damaging biofilms. Lactobacillus species have known inhibitory effects on biofilm formation of many microbes, including Streptococcus species. Agent-based modeling has great advantages as a means of exploring multifaceted relationships between organisms in complex environments such as biofilms. Here, we used an iterative collaborative process between experimentation and modeling to reveal aspects of the mostly unexplored relationship between S. oralis and L. paracasei in biofilm growth. The inhibitory nature of L. paracasei on S. oralis in biofilms may be exploited as a means of preventing or alleviating mucosal fungal infections.
Collapse
|
32
|
Mishra K, Bukavina L, Ghannoum M. Symbiosis and Dysbiosis of the Human Mycobiome. Front Microbiol 2021; 12:636131. [PMID: 34630340 PMCID: PMC8493257 DOI: 10.3389/fmicb.2021.636131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 08/04/2021] [Indexed: 12/13/2022] Open
Abstract
The influence of microbiological species has gained increased visibility and traction in the medical domain with major revelations about the role of bacteria on symbiosis and dysbiosis. A large reason for these revelations can be attributed to advances in deep-sequencing technologies. However, the research on the role of fungi has lagged. With the continued utilization of sequencing technologies in conjunction with traditional culture assays, we have the opportunity to shed light on the complex interplay between the bacteriome and the mycobiome as they relate to human health. In this review, we aim to offer a comprehensive overview of the human mycobiome in healthy and diseased states in a systematic way. The authors hope that the reader will utilize this review as a scaffolding to formulate their understanding of the mycobiome and pursue further research.
Collapse
Affiliation(s)
- Kirtishri Mishra
- University Hospitals Cleveland Medical Center, Urology Institute, Cleveland, OH, United States.,Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Laura Bukavina
- University Hospitals Cleveland Medical Center, Urology Institute, Cleveland, OH, United States.,Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Mahmoud Ghannoum
- Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, United States.,Center for Medical Mycology, and Integrated Microbiome Core, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, United States.,Department of Dermatology, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
33
|
Complete Genome Sequence of Streptococcus oralis 34. Microbiol Resour Announc 2021; 10:e0076021. [PMID: 34472981 PMCID: PMC8411913 DOI: 10.1128/mra.00760-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Streptococcus oralis is an early colonizer and one of the most abundant species found in the human oral cavity. We report the complete genome sequence of S. oralis 34 (1,920,884 bp; GC content, 41.3%), commonly used in many oral microbiology studies exploring bacterial attachment and interaction(s) within mixed-species model systems.
Collapse
|
34
|
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
|
35
|
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: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
36
|
Ardizzoni A, Wheeler RT, Pericolini E. It Takes Two to Tango: How a Dysregulation of the Innate Immunity, Coupled With Candida Virulence, Triggers VVC Onset. Front Microbiol 2021; 12:692491. [PMID: 34163460 PMCID: PMC8215348 DOI: 10.3389/fmicb.2021.692491] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/12/2021] [Indexed: 12/11/2022] Open
Abstract
Vulvovaginal candidiasis (VVC) is a symptomatic inflammation of the vagina mainly caused by C. albicans. Other species, such as C. parapsilosis, C. glabrata, C. tropicalis, and C. krusei, are mainly associated to the recurrent form of the disease (RVVC), although with a lower frequency. In its yeast form, C. albicans is tolerated by the vaginal epithelium, but switching to the invasive hyphal form, co-regulated with the expression of genes encoding virulence factors such as secreted aspartyl proteases (Sap) and candidalysin, allows for tissue damage. Vaginal epithelial cells play an important role by impairing C. albicans tissue invasion through several mechanisms such as epithelial shedding, secretion of mucin and strong interepithelial cell connections. However, morphotype switching coupled to increasing of the fungal burden can overcome the tolerance threshold and trigger an intense inflammatory response. Pathological inflammation is believed to be facilitated by an altered vaginal microbiome, i.e., Lactobacillus dysbiosis. Notwithstanding the damage caused by the fungus itself, the host response to the fungus plays an important role in the onset of VVC, exacerbating fungal-mediated damage. This response can be triggered by host PRR-fungal PAMP interaction and other more complex mechanisms (i.e., Sap-mediated NLRP3 activation and candidalysin), ultimately leading to strong neutrophil recruitment. However, recruited neutrophils appear to be ineffective at reducing fungal burden and invasion; therefore, they seem to contribute more to the symptoms associated with vaginitis than to protection against the disease. Recently, two aspects of the vulvovaginal environment have been found to associate with VVC and induce neutrophil anergy in vitro: perinuclear anti-neutrophil cytoplasmic antibodies (pANCA) and heparan sulfate. Interestingly, CAGTA antibodies have also been found with higher frequency in VVC as compared to asymptomatic colonized women. This review highlights and discusses recent advances on understanding the VVC pathogenesis mechanisms as well as the role of host defenses during the disease.
Collapse
Affiliation(s)
- Andrea Ardizzoni
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Robert T Wheeler
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, United States.,Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
| | - Eva Pericolini
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy.,Graduate School of Microbiology and Virology, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
37
|
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: 157] [Impact Index Per Article: 39.3] [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
|
38
|
Tsutsumi-Arai C, Arai Y, Terada-Ito C, Imamura T, Tatehara S, Ide S, Wakabayashi N, Satomura K. Microbicidal effect of 405-nm blue LED light on Candida albicans and Streptococcus mutans dual-species biofilms on denture base resin. Lasers Med Sci 2021; 37:857-866. [PMID: 33931832 DOI: 10.1007/s10103-021-03323-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 04/20/2021] [Indexed: 12/01/2022]
Abstract
This study investigated: (1) the microbicidal effect of 405-nm blue LED light irradiation on biofilm formed by Candida albicans hyphae and Streptococcus mutans under dual-species condition on denture base resin, (2) the generation of intracellular reactive oxygen species (ROS) induced by irradiation, and (3) the existence of intracellular porphyrins, which act as a photosensitizer. Denture base resin specimens were prepared and C. albicans and S. mutans dual-species biofilms were allowed to form on the specimens. The biofilms were irradiated with 405-nm blue LED light and analyzed using the colony-forming unit assay, fluorescence microscopy, and scanning electron microscopy (SEM). Single-species biofilms of C. albicans and S. mutans formed on the specimens were irradiated with 405-nm blue LED light. After the irradiation, the intracellular ROS levels in C. albicans and S. mutans cells were measured. In addition, the level of intracellular porphyrins in C. albicans and S. mutans were measured. Irradiation for more than 30 min significantly inhibited the colony formation ability of C. albicans and S. mutans. Fluorescence microscopy revealed that almost all C. albicans and S. mutans cells were killed by irradiation. SEM images showed various cell damage patterns. Irradiation led to the generation of intracellular ROS and porphyrins were present in both C. albicans and S. mutans cells. In conclusion, irradiation with 405-nm blue light-emitting diode light for 40 min effectively disinfect C. albicans hyphae and S. mutans dual-species biofilms and possibly react with intracellular porphyrins resulting in generation of ROS in each microorganism.
Collapse
Affiliation(s)
- Chiaki Tsutsumi-Arai
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, 2-1-3, Tsurumi, Tsurumi-ku, Yokohama, Kanagawa, 230-8501, Japan.
| | - Yuki Arai
- Department of Removable Partial Prosthodontics, Graduate School, Tokyo Medical and Dental University (TMDU), 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Chika Terada-Ito
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, 2-1-3, Tsurumi, Tsurumi-ku, Yokohama, Kanagawa, 230-8501, Japan
| | - Takahiro Imamura
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, 2-1-3, Tsurumi, Tsurumi-ku, Yokohama, Kanagawa, 230-8501, Japan
| | - Seiko Tatehara
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, 2-1-3, Tsurumi, Tsurumi-ku, Yokohama, Kanagawa, 230-8501, Japan
| | - Shinji Ide
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, 2-1-3, Tsurumi, Tsurumi-ku, Yokohama, Kanagawa, 230-8501, Japan
| | - Noriyuki Wakabayashi
- Department of Removable Partial Prosthodontics, Graduate School, Tokyo Medical and Dental University (TMDU), 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Kazuhito Satomura
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, 2-1-3, Tsurumi, Tsurumi-ku, Yokohama, Kanagawa, 230-8501, Japan
| |
Collapse
|
39
|
Crossing Kingdoms: How the Mycobiota and Fungal-Bacterial Interactions Impact Host Health and Disease. Infect Immun 2021; 89:IAI.00648-20. [PMID: 33526565 PMCID: PMC8090948 DOI: 10.1128/iai.00648-20] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The term “microbiota” invokes images of mucosal surfaces densely populated with bacteria. These surfaces and the luminal compartments they form indeed predominantly harbor bacteria. The term “microbiota” invokes images of mucosal surfaces densely populated with bacteria. These surfaces and the luminal compartments they form indeed predominantly harbor bacteria. However, research from this past decade has started to complete the picture by focusing on important but largely neglected constituents of the microbiota: fungi, viruses, and archaea. The community of commensal fungi, also called the mycobiota, interacts with commensal bacteria and the host. It is thus not surprising that changes in the mycobiota have significant impact on host health and are associated with pathological conditions such as inflammatory bowel disease (IBD). In this review we will give an overview of why the mycobiota is an important research area and different mycobiota research tools. We will specifically focus on distinguishing transient and actively colonizing fungi of the oral and gut mycobiota and their roles in health and disease. In addition to correlative and observational studies, we will discuss mechanistic studies on specific cross-kingdom interactions of fungi, bacteria, and the host.
Collapse
|
40
|
Candida albicans promotes tooth decay by inducing oral microbial dysbiosis. THE ISME JOURNAL 2021; 15:894-908. [PMID: 33149208 PMCID: PMC8026629 DOI: 10.1038/s41396-020-00823-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 10/14/2020] [Accepted: 10/22/2020] [Indexed: 02/05/2023]
Abstract
Candida albicans has been detected in root carious lesions. The current study aimed to explore the action of this fungal species on the microbial ecology and the pathogenesis of root caries. Here, by analyzing C. albicans in supragingival dental plaque collected from root carious lesions and sound root surfaces of root-caries subjects as well as caries-free individuals, we observed significantly increased colonization of C. albicans in root carious lesions. Further in vitro and animal studies showed that C. albicans colonization increased the cariogenicity of oral biofilm by altering its microbial ecology, leading to a polymicrobial biofilm with enhanced acidogenicity, and consequently exacerbated tooth demineralization and carious lesion severity. More importantly, we demonstrated that the cariogenicity-promoting activity of C. albicans was dependent on PHR2. Deletion of PHR2 restored microbial equilibrium and led to a less cariogenic biofilm as demonstrated by in vitro artificial caries model or in vivo root-caries rat model. Our data indicate the critical role of C. albicans infection in the occurrence of root caries. PHR2 is the major factor that determines the ecological impact and caries-promoting activity of C. albicans in a mixed microbial consortium.
Collapse
|
41
|
Dash S, Duraivelan K, Samanta D. Cadherin-mediated host-pathogen interactions. Cell Microbiol 2021; 23:e13316. [PMID: 33543826 DOI: 10.1111/cmi.13316] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/04/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022]
Abstract
Cell adhesion molecules mediate cell-to-cell and cell-to-matrix adhesions and play an immense role in a myriad of physiological processes during the growth and development of a multicellular organism. Cadherins belong to a major group of membrane-bound cell surface proteins that, in coordination with nectins, drive the formation and maintenance of adherens junctions for mediating cell to cell adhesion, cellular communication and signalling. Alongside adhesive function, the involvement of cadherins in mediating host-pathogen interactions has been extensively explored in recent years. In this review, we provide an in-depth understanding of microbial pathogens and their virulence factors that exploit cadherins for their strategical invasion into the host cell. Furthermore, macromolecular interactions involving cadherins and various microbial factors such as secretory toxins and adhesins lead to the disintegration of host cell junctions followed by the entry of the pathogen or triggering downstream signalling pathways responsible for successful invasion of the pathogenic microbes are discussed. Besides providing a comprehensive insight into some of the structural complexes involving cadherins and microbial factors to offer the mechanistic details of host-pathogen interactions, the current review also highlights novel constituents of various cell signalling events such as endocytosis machinery elicited upon microbial infections.
Collapse
Affiliation(s)
- Sagarika Dash
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur, India
| | | | - Dibyendu Samanta
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
42
|
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
|
43
|
Ho J, Camilli G, Griffiths JS, Richardson JP, Kichik N, Naglik JR. Candida albicans and candidalysin in inflammatory disorders and cancer. Immunology 2021; 162:11-16. [PMID: 32880925 PMCID: PMC7730014 DOI: 10.1111/imm.13255] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/05/2020] [Accepted: 08/14/2020] [Indexed: 12/30/2022] Open
Abstract
As our understanding of mycology progresses, the impact of fungal microbes on human health has become increasingly evident. Candida albicans is a common commensal fungus that gives rise to local and systemic infections, particularly in immunocompromised patients where it can result in mortality. However, C. albicans has also been quietly linked with a variety of inflammatory disorders, to which it has traditionally been considered incidental; recent studies may now provide new aspects of these relationships for further consideration. This review provides a novel perspective on the impact of C. albicans and its peptide toxin, candidalysin, on human health, exploring their contributions to pathology within a variety of diseases.
Collapse
Affiliation(s)
- Jemima Ho
- Centre for Host‐Microbiome InteractionsFaculty of Dentistry, Oral & Craniofacial SciencesKing's College LondonLondonUK
| | - Giorgio Camilli
- Centre for Host‐Microbiome InteractionsFaculty of Dentistry, Oral & Craniofacial SciencesKing's College LondonLondonUK
| | - James S. Griffiths
- Centre for Host‐Microbiome InteractionsFaculty of Dentistry, Oral & Craniofacial SciencesKing's College LondonLondonUK
| | - Jonathan P. Richardson
- Centre for Host‐Microbiome InteractionsFaculty of Dentistry, Oral & Craniofacial SciencesKing's College LondonLondonUK
| | - Nessim Kichik
- Centre for Host‐Microbiome InteractionsFaculty of Dentistry, Oral & Craniofacial SciencesKing's College LondonLondonUK
| | - Julian R. Naglik
- Centre for Host‐Microbiome InteractionsFaculty of Dentistry, Oral & Craniofacial SciencesKing's College LondonLondonUK
| |
Collapse
|
44
|
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
|
45
|
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: 36] [Impact Index Per Article: 7.2] [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
|
46
|
Chen L, Qu S, Yang K, Liu M, Li YX, Keller NP, Zeng X, Tian J. Perillaldehyde: A promising antifungal agent to treat oropharyngeal candidiasis. Biochem Pharmacol 2020; 180:114201. [PMID: 32822688 DOI: 10.1016/j.bcp.2020.114201] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 12/13/2022]
Abstract
Perillaldehyde (PAE), a natural monoterpenoid agent extracted from Perilla frutescence, PAE has been reported to present various physiological capabilities, such as anti-inflammation, anti-oxidative and anti-fungal. In this study, we show that PAE exhibits strong antifungal activity against Candida albicans (C. albicans). C. albicans, a fungal pathogen with high incidence of antifungal resistance in clinical settings, is the major cause of oropharyngeal candidiasis (OPC). OPC is characterized by inflammatory immunological responses to fungal infections. Our in vitro results show PAE inhibited several virulence attributes of C. albicans including biofilm formation, yeast-to-hyphal transition and secreted aspartic proteinases (SAPs) gene expression. Using an experimental murine model of OPC, we found that PAE inhibited NLRP3 inflammasome assembly, reduced the excessive accumulation of ROS and prevented the p65 transfer in nuclear; processes all leading to reduced inflammation burden in the host. Together, this supports use PAE as a promising new agent to improve OPC.
Collapse
Affiliation(s)
- Lei Chen
- College of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu Province, PR China
| | - Su Qu
- College of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu Province, PR China
| | - Kunlong Yang
- College of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu Province, PR China; Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United States
| | - Man Liu
- College of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu Province, PR China
| | - Yong-Xin Li
- College of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu Province, PR China
| | - Nancy P Keller
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United States.
| | - Xiaobin Zeng
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital, 2nd Clinical Medical College of Jinan University, and Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Medicine School of Shenzhen University, Shenzhen 518037, Guangdong Province, PR China.
| | - Jun Tian
- College of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu Province, PR China.
| |
Collapse
|
47
|
Bacteria Modify Candida albicans Hypha Formation, Microcolony Properties, and Survival within Macrophages. mSphere 2020; 5:5/4/e00689-20. [PMID: 32759336 PMCID: PMC7407070 DOI: 10.1128/msphere.00689-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Candida albicans is the predominant fungus colonizing the oral cavity that can have both synergistic and antagonistic interactions with other bacteria. Interkingdom polymicrobial associations modify fungal pathogenicity and are believed to increase microbial resistance to innate immunity. However, it is not known how these interactions alter fungal survival during phagocytic killing. We demonstrated that secreted molecules of S. gordonii and P. aeruginosa alter C. albicans survival within the phagosome of macrophages and alter fungal pathogenic phenotypes, including filamentation and microcolony formation. Moreover, we provide evidence for a dual interaction between S. gordonii and C. albicans such that S. gordonii signaling peptides can promote C. albicans commensalism by decreasing microcolony attachment while increasing invasion in epithelial cells. Our results identify bacterial diffusible factors as an attractive target to modify virulence of C. albicans in polymicrobial infections. Phagocytic cells are crucial components of the innate immune system preventing Candida albicans mucosal infections. Streptococcus gordonii and Pseudomonas aeruginosa often colonize mucosal sites, along with C. albicans, and yet interkingdom interactions that might alter the survival and escape of fungi from macrophages are not understood. Murine macrophages were coinfected with S. gordonii or P. aeruginosa, along with C. albicans to evaluate changes in fungal survival. S. gordonii increased C. albicans survival and filamentation within macrophage phagosomes, while P. aeruginosa reduced fungal survival and filamentation. Coinfection with S. gordonii resulted in greater escape of C. albicans from macrophages and increased size of fungal microcolonies formed on macrophage monolayers, while coinfection with P. aeruginosa reduced macrophage escape and produced smaller microcolonies. Microcolonies formed in the presence of P. aeruginosa cells outside macrophages also had significantly reduced size that was not found with P. aeruginosa phenazine deletion mutants. S. gordonii cells, as well as S. gordonii heat-fixed culture supernatants, increased C. albicans microcolony biomass but also resulted in microcolony detachment. A heat-resistant, trypsin-sensitive pheromone processed by S. gordonii Eep was needed for these effects. The majority of fungal microcolonies formed on human epithelial monolayers with S. gordonii supernatants developed as large floating structures with no detectable invasion of epithelium, along with reduced gene expression of C. albicansHYR1, EAP1, and HWP2 adhesins. However, a subset of C. albicans microcolonies was smaller and had greater epithelial invasiveness compared to microcolonies grown without S. gordonii. Thus, bacteria can alter the killing and escape of C. albicans from macrophages and contribute to changes in C. albicans pathogenicity. IMPORTANCECandida albicans is the predominant fungus colonizing the oral cavity that can have both synergistic and antagonistic interactions with other bacteria. Interkingdom polymicrobial associations modify fungal pathogenicity and are believed to increase microbial resistance to innate immunity. However, it is not known how these interactions alter fungal survival during phagocytic killing. We demonstrated that secreted molecules of S. gordonii and P. aeruginosa alter C. albicans survival within the phagosome of macrophages and alter fungal pathogenic phenotypes, including filamentation and microcolony formation. Moreover, we provide evidence for a dual interaction between S. gordonii and C. albicans such that S. gordonii signaling peptides can promote C. albicans commensalism by decreasing microcolony attachment while increasing invasion in epithelial cells. Our results identify bacterial diffusible factors as an attractive target to modify virulence of C. albicans in polymicrobial infections.
Collapse
|
48
|
In Vitro Effects of Streptococcus oralis Biofilm on Peri-Implant Soft Tissue Cells. Cells 2020; 9:cells9051226. [PMID: 32429151 PMCID: PMC7290395 DOI: 10.3390/cells9051226] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Human gingival epithelial cells (HGEps) and fibroblasts (HGFs) are the main cell types in peri-implant soft tissue. HGEps are constantly exposed to bacteria, but HGFs are protected by connective tissue as long as the mucosa-implant seal is intact. Streptococcus oralis is one of the commensal bacteria, is highly abundant at healthy implant sites, and might modulate soft tissue cells-as has been described for other streptococci. We have therefore investigated the effects of the S. oralis biofilm on HGEps and HGFs. HGEps or HGFs were grown separately on titanium disks and responded to challenge with S. oralis biofilm. HGFs were severely damaged after 4 h, exhibiting transcriptional inflammatory and stress responses. In contrast, challenge with S. oralis only induced a mild transcriptional inflammatory response in HGEps, without cellular damage. HGFs were more susceptible to the S. oralis biofilm than HGEps. The pro-inflammatory interleukin 6 (IL-6) was attenuated in HGFs, as was interleukin 8 (CXCL8) in HGEps. This indicates that S. oralis can actively protect tissue. In conclusion, commensal biofilms can promote homeostatic tissue protection, but only if the implant-mucosa interface is intact and HGFs are not directly exposed.
Collapse
|
49
|
Souza JGS, Bertolini M, Thompson A, Mansfield JM, Grassmann AA, Maas K, Caimano MJ, Barao VAR, Vickerman MM, Dongari-Bagtzoglou A. Role of glucosyltransferase R in biofilm interactions between Streptococcus oralis and Candida albicans. THE ISME JOURNAL 2020; 14:1207-1222. [PMID: 32042100 PMCID: PMC7174356 DOI: 10.1038/s41396-020-0608-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/08/2020] [Accepted: 01/30/2020] [Indexed: 02/08/2023]
Abstract
Streptococcal glucosyltransferases (Gtf) synthesize α-glucan exopolymers which contribute to biofilm matrix. Streptococcus oralis interacts with the opportunistic pathogen Candida albicans to form hypervirulent biofilms. S. oralis 34 has a single gtf gene (gtfR). However, the role of gtfR in single and mixed species biofilms with C. albicans has never been examined. A gtfR deletion mutant, purified GtfR, and recombinant GtfR glucan-binding domain were tested in single and mixed biofilms on different substrata in vitro. A mouse oral infection model was also used. We found that in single species biofilms growing with sucrose on abiotic surfaces S. oralis gtfR increased biofilm matrix, but not bacterial biomass. In biofilms with C. albicans, S. oralis encoding gtfR showed increased bacterial biomass on all surfaces. C. albicans had a positive effect on α-glucan synthesis, and α-glucans increased C. albicans accretion on abiotic surfaces. In single and mixed infection of mice receiving sucrose S. oralis gtfR enhanced mucosal burdens. However, sucrose had a negative impact on C. albicans burdens and reduced S. oralis burdens in co-infected mice. Our data provide new insights on the GtfR-mediated interactions between the two organisms and the influence of biofilm substratum and the mucosal environment on these interactions.
Collapse
Affiliation(s)
- João Gabriel Silva Souza
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, Farmington, CT, USA
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - Martinna Bertolini
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, Farmington, CT, USA
| | - Angela Thompson
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, Farmington, CT, USA
| | - Jillian M Mansfield
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA
| | - André Alex Grassmann
- Departments of Medicine, Pediatrics and Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, USA
| | - Kendra Maas
- Microbial Analysis, Resources, and Services Core, University of Connecticut, Storrs, CT, USA
| | - Melissa J Caimano
- Departments of Medicine, Pediatrics and Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, USA
| | - Valentim Adelino Ricardo Barao
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - M Margaret Vickerman
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA.
| | - Anna Dongari-Bagtzoglou
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, Farmington, CT, USA.
| |
Collapse
|
50
|
Dornelas Figueira LM, Ricomini Filho AP, da Silva WJ, Del BeL Cury AA, Ruiz KGS. Glucose effect on Candida albicans biofilm during tissue invasion. Arch Oral Biol 2020; 117:104728. [PMID: 32585445 DOI: 10.1016/j.archoralbio.2020.104728] [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: 10/15/2019] [Revised: 03/18/2020] [Accepted: 04/07/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To evaluate, in vitro, the effect of two glucose concentrations (0.1 mM and 1.0 mM, simulating glucose concentration in saliva of healthy and diabetic individuals) on Candida albicans biofilm grown on epithelial monolayer. MATERIAL AND METHODS C. albicans was inoculated on epithelial monolayers supplemented with 0.1 mM, 1.0 mM or no glucose. Control groups without C. albicans were also evaluated. Tissue response was assessed through the production of Interleukin-1α, Interleukin-8, Interleukin-6, Interleukin-10 and tumor necrosis factor-α. The complex of monolayer and biofilms were evaluated by quantitative reverse transcription polymerase chain reaction for expression of E-cadherin (CDH1), Caspase-3 (CASP3), β-defensin-1 (DEFB-1) and β-defensin-3 (DEFB-3). The biofilm architecture was visualized by confocal laser scanning microscopy. RESULTS The production of Interleukin-1α and Interleukin-8 were increased in the presence of C. albicans (p < 0.05). Glucose did not interfere in the release of any cytokine evaluated. C. albicans downregulated transcripts for CDH1 (p < 0.05). Glucose did not induce a significant change in CDH1, CASP3, DEFB-1 and DEFB-3 messenger RNA expression. The biofilms were more structured in the presence of glucose, but no difference in the diffusion of hyphae through the epithelial cells were observed. CONCLUSIONS The data suggest that glucose concentration does not affect the behavior of C. albicans during tissue invasion and other mechanisms must be related to the greater susceptibility of diabetic individuals to candidiasis.
Collapse
Affiliation(s)
- Louise Morais Dornelas Figueira
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| | | | - Wander José da Silva
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Altair Antoninha Del BeL Cury
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Karina Gonzales Silvério Ruiz
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| |
Collapse
|