51
|
Van Dyck K, Pinto RM, Pully D, Van Dijck P. Microbial Interkingdom Biofilms and the Quest for Novel Therapeutic Strategies. Microorganisms 2021; 9:412. [PMID: 33671126 PMCID: PMC7921918 DOI: 10.3390/microorganisms9020412] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/10/2021] [Accepted: 02/15/2021] [Indexed: 02/06/2023] Open
Abstract
Fungal and bacterial species interact with each other within polymicrobial biofilm communities in various niches of the human body. Interactions between these species can greatly affect human health and disease. Diseases caused by polymicrobial biofilms pose a major challenge in clinical settings because of their enhanced virulence and increased drug tolerance. Therefore, different approaches are being explored to treat fungal-bacterial biofilm infections. This review focuses on the main mechanisms involved in polymicrobial drug tolerance and the implications of the polymicrobial nature for the therapeutic treatment by highlighting clinically relevant fungal-bacterial interactions. Furthermore, innovative treatment strategies which specifically target polymicrobial biofilms are discussed.
Collapse
Affiliation(s)
- Katrien Van Dyck
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, Department of Biology, KU Leuven, 3001 Leuven, Belgium; (K.V.D.); (R.M.P.); (D.P.)
- VIB—KU Leuven Center for Microbiology, 3001 Leuven, Belgium
| | - Rita M. Pinto
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, Department of Biology, KU Leuven, 3001 Leuven, Belgium; (K.V.D.); (R.M.P.); (D.P.)
- VIB—KU Leuven Center for Microbiology, 3001 Leuven, Belgium
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade Do Porto, 4050-313 Porto, Portugal
| | - Durgasruthi Pully
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, Department of Biology, KU Leuven, 3001 Leuven, Belgium; (K.V.D.); (R.M.P.); (D.P.)
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, Department of Biology, KU Leuven, 3001 Leuven, Belgium; (K.V.D.); (R.M.P.); (D.P.)
- VIB—KU Leuven Center for Microbiology, 3001 Leuven, Belgium
| |
Collapse
|
52
|
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: 6] [Impact Index Per Article: 1.5] [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
|
53
|
Ponde NO, Lortal L, Ramage G, Naglik JR, Richardson JP. Candida albicans biofilms and polymicrobial interactions. Crit Rev Microbiol 2021; 47:91-111. [PMID: 33482069 PMCID: PMC7903066 DOI: 10.1080/1040841x.2020.1843400] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/05/2020] [Accepted: 10/25/2020] [Indexed: 12/16/2022]
Abstract
Candida albicans is a common fungus of the human microbiota. While generally a harmless commensal in healthy individuals, several factors can lead to its overgrowth and cause a range of complications within the host, from localized superficial infections to systemic life-threatening disseminated candidiasis. A major virulence factor of C. albicans is its ability to form biofilms, a closely packed community of cells that can grow on both abiotic and biotic substrates, including implanted medical devices and mucosal surfaces. These biofilms are extremely hard to eradicate, are resistant to conventional antifungal treatment and are associated with high morbidity and mortality rates, making biofilm-associated infections a major clinical challenge. Here, we review the current knowledge of the processes involved in C. albicans biofilm formation and development, including the central processes of adhesion, extracellular matrix production and the transcriptional network that regulates biofilm development. We also consider the advantages of the biofilm lifestyle and explore polymicrobial interactions within multispecies biofilms that are formed by C. albicans and selected microbial species.
Collapse
Affiliation(s)
- Nicole O. Ponde
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, SE1 9RT, United Kingdom
| | - Léa Lortal
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, SE1 9RT, United Kingdom
| | - Gordon Ramage
- School of Medicine, Dentistry & Nursing, Glasgow Dental School and Hospital, Faculty of Medicine, University of Glasgow, G2 3JZ, United Kingdom
| | - Julian R. Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, SE1 9RT, United Kingdom
| | - Jonathan P. Richardson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, SE1 9RT, United Kingdom
| |
Collapse
|
54
|
Paul D, Saha S, Singh N, Sengupta J, Mandal SM. Successful Control of a Co-Infection Caused by Candida albicans and Pseudomonas aeruginosa in Keratitis. Infect Disord Drug Targets 2021; 21:284-288. [PMID: 32188397 DOI: 10.2174/1871526520666200318111957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 01/19/2020] [Accepted: 03/08/2020] [Indexed: 06/10/2023]
Abstract
Nowadays, the co-infection of different classes of pathogens is a major concern. The objective of this study was to develop a successful therapy for keratitis caused by the co-infection of Candida sp. with Pseudomonas sp, which is difficult to cure. The study is based on a 47 years old male farmer showing redness and watering in the right eye for 15-days. ; Methods: The microbiological examination was performed to isolate the causative organisms, i.e. Pseudomonas aeruginosa and Candida albicans. They were cultured separately along with their co-culture and treated with ciprofloxacin and amphotericin B during the growing stage to predict a definite cure. ; Results: Scanning electron microscope (SEM) results confirmed the inter-specific interaction between the two different types of microorganisms. Amphotericin-B and Ciprofloxacin showed the least MIC value for both organisms in co-culture. ; Conclusion: Treatment with Amphotericin-B and 5% ciprofloxacin effectively hindered the growth of Pseudomonas aeruginosa and Candida albicans, the co-infection of which caused keratitis. This therapy may be successfully implied for such cases of co-infection in the future.
Collapse
Affiliation(s)
- Debarati Paul
- Amity Institute of Biotechnology, Amity University, Noida, Sector 125, 201313, India
| | - Suman Saha
- Priyamvada Birla Aravind Eye Hospital, Kolkata, 700017, WB, India
| | - Neelam Singh
- Babasaheb Bhimrao ambedkar University, Department of Brain and Cognition Science, 226025Lucknow, India
| | | | - Santi M Mandal
- Central Research Facility, Indian Institute of Technology Kharagpur, Kharagpur- 721302, West Bengal, India
| |
Collapse
|
55
|
Černáková L, Rodrigues CF. Microbial interactions and immunity response in oral Candida species. Future Microbiol 2020; 15:1653-1677. [PMID: 33251818 DOI: 10.2217/fmb-2020-0113] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Oral candidiasis are among the most common noncommunicable diseases, related with serious local and systemic illnesses. Although these infections can occur in all kinds of patients, they are more recurrent in immunosuppressed ones such as patients with HIV, hepatitis, cancer or under long antimicrobial treatments. Candida albicans continues to be the most frequently identified Candida spp. in these disorders, but other non-C. albicans Candida are rising. Understanding the immune responses involved in oral Candida spp. infections is a key feature to a successful treatment and to the design of novel therapies. In this review, we performed a literature search in PubMed and WoS, in order to examine and analyze common oral Candida spp.-bacteria/Candida-Candida interactions and the host immunity response in oral candidiasis.
Collapse
Affiliation(s)
- Lucia Černáková
- Department of Microbiology & Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovičova 6, 842 15 Bratislava, Slovakia
| | - Célia F Rodrigues
- Department of Chemical Engineering, LEPABE - Laboratory for Process Engineering, Environment, Biotechnology & Energy, Faculty of Engineering, University of Porto, Portugal
| |
Collapse
|
56
|
Manzer HS, Nobbs AH, Doran KS. The Multifaceted Nature of Streptococcal Antigen I/II Proteins in Colonization and Disease Pathogenesis. Front Microbiol 2020; 11:602305. [PMID: 33329493 PMCID: PMC7732690 DOI: 10.3389/fmicb.2020.602305] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/29/2020] [Indexed: 12/22/2022] Open
Abstract
Streptococci are Gram-positive bacteria that belong to the natural microbiota of humans and animals. Certain streptococcal species are known as opportunistic pathogens with the potential to cause severe invasive disease. Antigen I/II (AgI/II) family proteins are sortase anchored cell surface adhesins that are nearly ubiquitous across streptococci and contribute to many streptococcal diseases, including dental caries, respiratory tract infections, and meningitis. They appear to be multifunctional adhesins with affinities to various host substrata, acting to mediate attachment to host surfaces and stimulate immune responses from the colonized host. Here we will review the literature including recent work that has demonstrated the multifaceted nature of AgI/II family proteins, focusing on their overlapping and distinct functions and their important contribution to streptococcal colonization and disease.
Collapse
Affiliation(s)
- Haider S. Manzer
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Angela H. Nobbs
- Bristol Dental School, University of Bristol, Bristol, United Kingdom
| | - Kelly S. Doran
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
57
|
The Role of the Microbiome in Oral Squamous Cell Carcinoma with Insight into the Microbiome-Treatment Axis. Int J Mol Sci 2020; 21:ijms21218061. [PMID: 33137960 PMCID: PMC7662318 DOI: 10.3390/ijms21218061] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/06/2020] [Accepted: 10/12/2020] [Indexed: 12/24/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the leading presentations of head and neck cancer (HNC). The first part of this review will describe the highlights of the oral microbiome in health and normal development while demonstrating how both the oral and gut microbiome can map OSCC development, progression, treatment and the potential side effects associated with its management. We then scope the dynamics of the various microorganisms of the oral cavity, including bacteria, mycoplasma, fungi, archaea and viruses, and describe the characteristic roles they may play in OSCC development. We also highlight how the human immunodeficiency viruses (HIV) may impinge on the host microbiome and increase the burden of oral premalignant lesions and OSCC in patients with HIV. Finally, we summarise current insights into the microbiome–treatment axis pertaining to OSCC, and show how the microbiome is affected by radiotherapy, chemotherapy, immunotherapy and also how these therapies are affected by the state of the microbiome, potentially determining the success or failure of some of these treatments.
Collapse
|
58
|
Abstract
Interactions among microbes are key drivers of evolutionary progress and constantly shape ecological niches. Microorganisms rely on chemical communication to interact with each other and surrounding organisms. They synthesize natural products as signaling molecules, antibiotics, or modulators of cellular processes that may be applied in agriculture and medicine. Whereas major insight has been gained into the principles of intraspecies interaction, much less is known about the molecular basis of interspecies interplay. In this review, we summarize recent progress in the understanding of chemically mediated bacterial-fungal interrelations. We discuss pairwise interactions among defined species and systems involving additional organisms as well as complex interactions among microbial communities encountered in the soil or defined as microbiota of higher organisms. Finally, we give examples of how the growing understanding of microbial interactions has contributed to drug discovery and hypothesize what may be future directions in studying and engineering microbiota for agricultural or medicinal purposes.
Collapse
Affiliation(s)
- Kirstin Scherlach
- Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology, 07745 Jena, Germany
| | - Christian Hertweck
- Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology, 07745 Jena, Germany
- Faculty of Biological Sciences, Friedrich Schiller University Jena, 07745 Jena, Germany
| |
Collapse
|
59
|
Wan SX, Tian J, Liu Y, Dhall A, Koo H, Hwang G. Cross-Kingdom Cell-to-Cell Interactions in Cariogenic Biofilm Initiation. J Dent Res 2020; 100:74-81. [PMID: 32853527 DOI: 10.1177/0022034520950286] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Candida albicans is known to form polymicrobial biofilms with various Streptococcus spp., including mitis and mutans group streptococci. Streptococcus gordonii (mitis group) has been shown to bind avidly to C. albicans hyphae via direct cell-to-cell interaction, while the cariogenic pathogen Streptococcus mutans (mutans group) interacts with the fungal cells via extracellular glucans. However, the biophysical properties of these cross-kingdom interactions at the single-cell level during the early stage of biofilm formation remain understudied. Here, we examined the binding forces between S. mutans (or S. gordonii) and C. albicans in the presence and absence of in situ glucans on the fungal surface using single-cell atomic force microscopy and their influence on biofilm initiation and subsequent development under cariogenic conditions. The data show that S. gordonii binding force to the C. albicans surface is significantly higher than that ofS. mutans to the fungal surface (~2-fold). However, S. mutans binding forces are dramatically enhanced when the C. albicans cell surface is locally coated with extracellular glucans (~6-fold vs. uncoated C. albicans), which vastly exceeds the forces between S. gordonii andC. albicans. The enhanced binding affinity of S. mutans to glucan-coated C. albicans resulted in a larger structure during early biofilm initiation compared to S. gordonii-C. albicans biofilms. Ultimately, this resulted in S. mutans dominance composition in the 3-species biofilm model under cariogenic conditions. This study provides a novel biophysical aspect of Candida-streptococcal interaction whereby extracellular glucans may selectively favor S. mutans binding interactions with C. albicans during cariogenic biofilm development.
Collapse
Affiliation(s)
- S X Wan
- Biofilm Research Labs, Levy Center for Oral Health, Department of Orthodontics, Divisions of Pediatric Dentistry and Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - J Tian
- Biofilm Research Labs, Levy Center for Oral Health, Department of Orthodontics, Divisions of Pediatric Dentistry and Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Y Liu
- Biofilm Research Labs, Levy Center for Oral Health, Department of Orthodontics, Divisions of Pediatric Dentistry and Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - A Dhall
- Department of Preventive and Restorative Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - H Koo
- Biofilm Research Labs, Levy Center for Oral Health, Department of Orthodontics, Divisions of Pediatric Dentistry and Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Center for Innovation and Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - G Hwang
- Biofilm Research Labs, Levy Center for Oral Health, Department of Orthodontics, Divisions of Pediatric Dentistry and Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Preventive and Restorative Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Center for Innovation and Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
60
|
Interactions between invasive fungi and symbiotic bacteria. World J Microbiol Biotechnol 2020; 36:137. [PMID: 32794072 DOI: 10.1007/s11274-020-02913-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/08/2020] [Indexed: 12/17/2022]
Abstract
Infection rates and mortality associated with the invasive fungi Candida, Aspergillus, and Cryptococcus are increasing rapidly in prevalence. Meanwhile, screening pressure brought about by traditional antifungal drugs has induced an increase in drug resistance of invasive fungi, which creates a great challenge for the preservation of physical health. Development of new drugs and novel strategies are therefore important to meet these growing challenges. Recent studies have confirmed that the dynamic balance of microorganisms in the body is correlated with the occurrence of infectious diseases. This discovery of interactions between bacteria and fungi provides innovative insight for the treatment of invasive fungal infections. However, different invasive fungi and symbiotic bacteria interact with each other through various ways and targets, leading to different effects on their growth, morphology, and virulence. And the mechanism and implication of these interactions remains largely unknown. The present review aims to summarize the research progress into the interaction between invasive fungi and symbiotic bacteria with a focus on the anti-fungal mechanisms of symbiotic bacteria, providing a new strategy against drug-resistant fungal infections.
Collapse
|
61
|
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: 14] [Impact Index Per Article: 2.8] [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
|
62
|
Bhardwaj RG, Ellepolla A, Drobiova H, Karched M. Biofilm growth and IL-8 & TNF-α-inducing properties of Candida albicans in the presence of oral gram-positive and gram-negative bacteria. BMC Microbiol 2020; 20:156. [PMID: 32527216 PMCID: PMC7291589 DOI: 10.1186/s12866-020-01834-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/28/2020] [Indexed: 12/16/2022] Open
Abstract
Background Interaction of C. albicans with oral bacteria is crucial for its persistence, but also plays a potential role in the infection process. In the oral cavity, it grows as part of dental plaque biofilms. Even though growth and interaction of C. albicans with certain bacterial species has been studied, little is known about its biofilm growth in vitro in the simultaneous presence of Gram-negative and Gram-positive bacteria. The aim was to evaluate the growth of C. albicans in polymicrobial biofilms comprising oral Gram-negative and Gram-positive bacteria. Further, we also aimed to assess the potential of C. albicans in the Candida-bacteria polymicrobial biofilm to elicit cytokine gene expression and cytokine production from human blood cells. Results C. albicans cell counts increased significantly up to 48 h in polymicrobial biofilms (p < 0.05), while the bacterial counts in the same biofilms increased only marginally as revealed by qPCR absolute quantification. However, the presence of bacteria in the biofilm did not seem to affect the growth of C. albicans. Expression of IL-8 gene was significantly (p < 0.05) higher upon stimulation from biofilm-supernatants than from biofilms in polymicrobial setting. On the contrary, TNF-α expression was significantly higher in biofilms than in supernatants but was very low (1–4 folds) in the monospecies biofilm of C. albicans. ELISA cytokine quantification data was in agreement with mRNA expression results. Conclusion Persistence and enhanced growth of C. albicans in polymicrobial biofilms may imply that previously reported antagonistic effect of A. actinomycetemcomitans was negated. Increased cytokine gene expression and cytokine production induced by Candida-bacteria polymicrobial biofilms and biofilm supernatants suggest that together they possibly exert an enhanced stimulatory effect on IL-8 and TNF-α production from the host.
Collapse
Affiliation(s)
- Radhika G Bhardwaj
- Oral Microbiology Research Laboratory, Department of Bioclinical Sciences, Faculty of Dentistry, Kuwait University, PO Box 24923, 13110, Safat, Kuwait
| | - Arjuna Ellepolla
- Oral Microbiology Research Laboratory, Department of Bioclinical Sciences, Faculty of Dentistry, Kuwait University, PO Box 24923, 13110, Safat, Kuwait
| | - Hana Drobiova
- Oral Microbiology Research Laboratory, Department of Bioclinical Sciences, Faculty of Dentistry, Kuwait University, PO Box 24923, 13110, Safat, Kuwait
| | - Maribasappa Karched
- Oral Microbiology Research Laboratory, Department of Bioclinical Sciences, Faculty of Dentistry, Kuwait University, PO Box 24923, 13110, Safat, Kuwait.
| |
Collapse
|
63
|
Lohse MB, Gulati M, Craik CS, Johnson AD, Nobile CJ. Combination of Antifungal Drugs and Protease Inhibitors Prevent Candida albicans Biofilm Formation and Disrupt Mature Biofilms. Front Microbiol 2020; 11:1027. [PMID: 32523574 PMCID: PMC7261846 DOI: 10.3389/fmicb.2020.01027] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/27/2020] [Indexed: 02/04/2023] Open
Abstract
Biofilms formed by the fungal pathogen Candida albicans are resistant to many of the antifungal agents commonly used in the clinic. Previous reports suggest that protease inhibitors, specifically inhibitors of aspartyl proteases, could be effective antibiofilm agents. We screened three protease inhibitor libraries, containing a total of 80 compounds for the abilities to prevent C. albicans biofilm formation and to disrupt mature biofilms. The compounds were screened individually and in the presence of subinhibitory concentrations of the most commonly prescribed antifungal agents for Candida infections: fluconazole, amphotericin B, or caspofungin. Although few of the compounds affected biofilms on their own, seven aspartyl protease inhibitors inhibited biofilm formation when combined with amphotericin B or caspofungin. Furthermore, nine aspartyl protease inhibitors disrupted mature biofilms when combined with caspofungin. These results suggest that the combination of standard antifungal agents together with specific protease inhibitors may be useful in the prevention and treatment of C. albicans biofilm infections.
Collapse
Affiliation(s)
- Matthew B Lohse
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, United States.,Department of Biology, BioSynesis, Inc., San Francisco, CA, United States
| | - Megha Gulati
- Department of Molecular and Cell Biology, University of California, Merced, Merced, CA, United States
| | - Charles S Craik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, United States
| | - Alexander D Johnson
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, United States
| | - Clarissa J Nobile
- Department of Molecular and Cell Biology, University of California, Merced, Merced, CA, United States
| |
Collapse
|
64
|
A Selective Serotonin Reuptake Inhibitor, a Proton Pump Inhibitor, and Two Calcium Channel Blockers Inhibit Candida albicans Biofilms. Microorganisms 2020; 8:microorganisms8050756. [PMID: 32443498 PMCID: PMC7285287 DOI: 10.3390/microorganisms8050756] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/08/2020] [Accepted: 05/17/2020] [Indexed: 01/05/2023] Open
Abstract
Biofilms formed by the human fungal pathogen Candida albicans are naturally resistant to many of the antifungal agents commonly used in the clinic. We screened a library containing 1600 clinically tested drug compounds to identify compounds that inhibit C. albicans biofilm formation. The compounds that emerged from the initial screen were validated in a secondary screen and then tested for (1) their abilities to disrupt mature biofilms and (2) for synergistic interactions with representatives of the three antifungal agents most commonly prescribed to treat Candida infections, fluconazole, amphotericin B, and caspofungin. Twenty compounds had antibiofilm activity in at least one of the secondary assays and several affected biofilms but, at the same concentration, had little or no effect on planktonic (suspension) growth of C. albicans. Two calcium channel blockers, a selective serotonin reuptake inhibitor, and an azole-based proton pump inhibitor were among the hits, suggesting that members of these three classes of drugs or their derivatives may be useful for treating C. albicans biofilm infections.
Collapse
|
65
|
Bernard C, Girardot M, Imbert C. Candida albicans interaction with Gram-positive bacteria within interkingdom biofilms. J Mycol Med 2020; 30:100909. [DOI: 10.1016/j.mycmed.2019.100909] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 10/08/2019] [Accepted: 10/27/2019] [Indexed: 12/19/2022]
|
66
|
Vila T, Sultan AS, Montelongo-Jauregui D, Jabra-Rizk MA. Oral Candidiasis: A Disease of Opportunity. J Fungi (Basel) 2020; 6:jof6010015. [PMID: 31963180 PMCID: PMC7151112 DOI: 10.3390/jof6010015] [Citation(s) in RCA: 214] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/14/2022] Open
Abstract
Oral candidiasis, commonly referred to as “thrush,” is an opportunistic fungal infection that commonly affects the oral mucosa. The main causative agent, Candida albicans, is a highly versatile commensal organism that is well adapted to its human host; however, changes in the host microenvironment can promote the transition from one of commensalism to pathogen. This transition is heavily reliant on an impressive repertoire of virulence factors, most notably cell surface adhesins, proteolytic enzymes, morphologic switching, and the development of drug resistance. In the oral cavity, the co-adhesion of C. albicans with bacteria is crucial for its persistence, and a wide range of synergistic interactions with various oral species were described to enhance colonization in the host. As a frequent colonizer of the oral mucosa, the host immune response in the oral cavity is oriented toward a more tolerogenic state and, therefore, local innate immune defenses play a central role in maintaining Candida in its commensal state. Specifically, in addition to preventing Candida adherence to epithelial cells, saliva is enriched with anti-candidal peptides, considered to be part of the host innate immunity. The T helper 17 (Th17)-type adaptive immune response is mainly involved in mucosal host defenses, controlling initial growth of Candida and inhibiting subsequent tissue invasion. Animal models, most notably the mouse model of oropharyngeal candidiasis and the rat model of denture stomatitis, are instrumental in our understanding of Candida virulence factors and the factors leading to host susceptibility to infections. Given the continuing rise in development of resistance to the limited number of traditional antifungal agents, novel therapeutic strategies are directed toward identifying bioactive compounds that target pathogenic mechanisms to prevent C. albicans transition from harmless commensal to pathogen.
Collapse
Affiliation(s)
- Taissa Vila
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (T.V.); (A.S.S.); (D.M.-J.)
| | - Ahmed S. Sultan
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (T.V.); (A.S.S.); (D.M.-J.)
| | - Daniel Montelongo-Jauregui
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (T.V.); (A.S.S.); (D.M.-J.)
| | - Mary Ann Jabra-Rizk
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (T.V.); (A.S.S.); (D.M.-J.)
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
- Correspondence: ; Tel.: +1-410-706-0508; Fax: +1-410-706-0519
| |
Collapse
|
67
|
Diaz P, Valm A. Microbial Interactions in Oral Communities Mediate Emergent Biofilm Properties. J Dent Res 2020; 99:18-25. [PMID: 31590609 PMCID: PMC6927214 DOI: 10.1177/0022034519880157] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Oral microbial communities are extraordinarily complex in taxonomic composition and comprise interdependent biological systems. The bacteria, archaea, fungi, and viruses that thrive within these communities engage in extensive cell-cell interactions, which are both beneficial and antagonistic. Direct physical interactions among individual cells mediate large-scale architectural biofilm arrangements and provide spatial proximity for chemical communication and metabolic cooperation. In this review, we summarize recent work in identifying specific molecular components that mediate cell-cell interactions and describe metabolic interactions, such as cross-feeding and exchange of electron acceptors and small molecules, that modify the growth and virulence of individual species. We argue, however, that although pairwise interaction models have provided useful information, complex community-like systems are needed to study the properties of oral communities. The networks of multiple synergistic and antagonistic interactions within oral biofilms give rise to the emergent properties of persistence, stability, and long-range spatial structure, with these properties mediating the dysbiotic transitions from health to oral diseases. A better understanding of the fundamental properties of interspecies networks will lead to the development of effective strategies to manipulate oral communities.
Collapse
Affiliation(s)
- P.I. Diaz
- Division of Periodontology, Department of Oral Health and Diagnostic Sciences, School of Dental Medicine, UConn Health, Farmington, CT, USA
| | - A.M. Valm
- Department of Biological Sciences, University at Albany, SUNY, Albany, NY, USA,A.M. Valm, Department of Biological Sciences, University at Albany, SUNY, 1400 Washington Ave., Albany, NY 12222, USA.
| |
Collapse
|
68
|
Deng L, Xue J, Jiang L, Zou L, Li W. [Research progress on interactions between Candida albicans and common oral pathogens]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2019; 37:671-676. [PMID: 31875449 DOI: 10.7518/hxkq.2019.06.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Increasing numbers of microbiome studies have enabled the development of a greater understanding of how antagonistic and synergetic microbial interactions influence disease outcomes. Candida albicans is an opportunistic pathogen that is commonly found in human oral microflora. In a healthy oral environment, Candida albicans may potentially but sig-nificantly influence the balance between the oral bacterial ecosystem and the host, leading tooral diseases. The aim of this study is to review the correlation between Candida albicans and oral pathogens and provide a deeper understanding of the nature of oral infec-tious diseases.
Collapse
Affiliation(s)
- Ling Deng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jing Xue
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Li Jiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of General Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ling Zou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Wei Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
69
|
Lyons KM, Cannon RD, Beumer J, Bakr MM, Love RM. The Role of Biofilms and Material Surface Characteristics in Microbial Adhesion to Maxillary Obturator Materials: A Literature Review. Cleft Palate Craniofac J 2019; 57:487-498. [PMID: 31665902 DOI: 10.1177/1055665619882555] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Maxillofacial prosthetics includes restoration of maxillary defects resulting from resection of palate and nasosinus neoplasms with obturator prostheses which may be colonized by microorganisms and function as a reservoir of infection. Patients with neoplasms commonly also require radiotherapy that can result in changes in saliva quality and quantity and changes in the oral microbial flora. The altered flora, in individuals immunocompromised from cancer therapy, increases their risk of prosthesis-related infections. OBJECTIVES In this review article, we explore microbial biofilms, their main components, mechanisms of microbial adhesion, and stages of biofilm development. We also discuss the different materials that are used for manufacturing maxillary obturators, their characteristic features, and how these can affect microbial adhesion. Furthermore, we shed some light on the factors that affect microbial adhesion to the surface of maxillary obturators including tissue proteins, protein adsorption, and the acquired enamel pellicle. CONCLUSIONS The conclusions drawn from this literature review are that it is imperative to minimize the risk of local and systemic infections in immunocompromised patients with cancer having maxillary defects. It is also important to determine the role of saliva in microbial adhesion to obturator materials as well as develop materials that have a longer life span with surface characteristics that promote less microbial adhesion than current materials.
Collapse
Affiliation(s)
- Karl M Lyons
- Department of Oral Rehabilitation and Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Richard D Cannon
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin, New Zealand
| | - John Beumer
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Mahmoud M Bakr
- School of Dentistry and Oral Health, Griffith University, Queensland, Australia
| | - Robert M Love
- School of Dentistry and Oral Health, Griffith University, Queensland, Australia
| |
Collapse
|
70
|
Nogueira F, Sharghi S, Kuchler K, Lion T. Pathogenetic Impact of Bacterial-Fungal Interactions. Microorganisms 2019; 7:microorganisms7100459. [PMID: 31623187 PMCID: PMC6843596 DOI: 10.3390/microorganisms7100459] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/20/2019] [Accepted: 10/15/2019] [Indexed: 12/17/2022] Open
Abstract
Polymicrobial infections are of paramount importance because of the potential severity of clinical manifestations, often associated with increased resistance to antimicrobial treatment. The intricate interplay with the host and the immune system, and the impact on microbiome imbalance, are of importance in this context. The equilibrium of microbiota in the human host is critical for preventing potential dysbiosis and the ensuing development of disease. Bacteria and fungi can communicate via signaling molecules, and produce metabolites and toxins capable of modulating the immune response or altering the efficacy of treatment. Most of the bacterial–fungal interactions described to date focus on the human fungal pathogen Candida albicans and different bacteria. In this review, we discuss more than twenty different bacterial–fungal interactions involving several clinically important human pathogens. The interactions, which can be synergistic or antagonistic, both in vitro and in vivo, are addressed with a focus on the quorum-sensing molecules produced, the response of the immune system, and the impact on clinical outcome.
Collapse
Affiliation(s)
- Filomena Nogueira
- CCRI-St. Anna Children's Cancer Research Institute, Vienna 1090, Austria.
- Labdia-Labordiagnostik GmbH, Vienna 1090, Austria.
- Center of Medical Biochemistry, Max Perutz Labs, Campus Vienna Biocenter, Medical University of Vienna, Vienna 1030, Austria.
| | - Shirin Sharghi
- CCRI-St. Anna Children's Cancer Research Institute, Vienna 1090, Austria.
- Labdia-Labordiagnostik GmbH, Vienna 1090, Austria.
- Center of Medical Biochemistry, Max Perutz Labs, Campus Vienna Biocenter, Medical University of Vienna, Vienna 1030, Austria.
| | - Karl Kuchler
- Center of Medical Biochemistry, Max Perutz Labs, Campus Vienna Biocenter, Medical University of Vienna, Vienna 1030, Austria.
| | - Thomas Lion
- CCRI-St. Anna Children's Cancer Research Institute, Vienna 1090, Austria.
- Labdia-Labordiagnostik GmbH, Vienna 1090, Austria.
- Department of Pediatrics, Medical University of Vienna, Vienna 1090, Austria.
| |
Collapse
|
71
|
Chinnici J, Yerke L, Tsou C, Busarajan S, Mancuso R, Sadhak ND, Kim J, Maddi A. Candida albicans cell wall integrity transcription factors regulate polymicrobial biofilm formation with Streptococcus gordonii. PeerJ 2019; 7:e7870. [PMID: 31616604 PMCID: PMC6791342 DOI: 10.7717/peerj.7870] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/11/2019] [Indexed: 12/13/2022] Open
Abstract
Polymicrobial biofilms play important roles in oral and systemic infections. The oral plaque bacterium Streptococcus gordonii is known to attach to the hyphal cell wall of the fungus Candida albicans to form corn-cob like structures in biofilms. However, the role of C. albicans in formation of polymicrobial biofilms is not completely understood. The objective of this study was to determine the role of C. albicans transcription factors in regulation of polymicrobial biofilms and antibiotic tolerance of S. gordonii. The proteins secreted by C. albicans and S. gordonii in mixed planktonic cultures were determined using mass spectrometry. Antibiotic tolerance of S. gordonii to ampicillin and erythromycin was determined in mixed cultures and mixed biofilms with C. albicans. Additionally, biofilm formation of S. gordonii with C. albicans knock-out mutants of 45 transcription factors that affect cell wall integrity, filamentous growth and biofilm formation was determined. Furthermore, these mutants were also screened for antibiotic tolerance in mixed biofilms with S. gordonii. Analysis of secreted proteomes resulted in the identification of proteins being secreted exclusively in mixed cultures. Antibiotic testing showed that S. gordonii had significantly increased survival in mixed planktonic cultures with antibiotics as compared to single cultures. C. albicans mutants of transcription factors Sfl2, Brg1, Leu3, Cas5, Cta4, Tec1, Tup1, Rim101 and Efg1 were significantly affected in mixed biofilm formation. Also mixed biofilms of S. gordonii with mutants of C. albicans transcription factors, Tec1 and Sfl2, had significantly reduced antibiotic tolerance as compared to control cultures. Our data indicates that C. albicans may have an important role in mixed biofilm formation as well as antibiotic tolerance of S. gordonii in polymicrobial biofilms. C. albicans may play a facilitating role than being just an innocent bystander in oral biofilms and infections.
Collapse
Affiliation(s)
- Jennifer Chinnici
- Departments of Periodontics & Endodontics and Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States of America
| | - Lisa Yerke
- Departments of Periodontics & Endodontics and Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States of America
| | - Charlene Tsou
- Departments of Periodontics & Endodontics and Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States of America
| | - Sujay Busarajan
- Departments of Periodontics & Endodontics and Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States of America
| | - Ryan Mancuso
- Departments of Periodontics & Endodontics and Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States of America
| | - Nishanth D Sadhak
- Departments of Periodontics & Endodontics and Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States of America
| | - Jaewon Kim
- Departments of Periodontics & Endodontics and Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States of America
| | - Abhiram Maddi
- Departments of Periodontics & Endodontics and Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States of America
| |
Collapse
|
72
|
Veerapandian R, Vediyappan G. Gymnemic Acids Inhibit Adhesive Nanofibrillar Mediated Streptococcus gordonii-Candida albicans Mono-Species and Dual-Species Biofilms. Front Microbiol 2019; 10:2328. [PMID: 31681200 PMCID: PMC6797559 DOI: 10.3389/fmicb.2019.02328] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/24/2019] [Indexed: 12/17/2022] Open
Abstract
Dental caries and periodontitis are the most common oral disease of all age groups, affecting billions of people worldwide. These oral diseases are mostly associated with microbial biofilms in the oral cavity. Streptococcus gordonii, an early tooth colonizing bacterium and Candida albicans, an opportunistic pathogenic fungus, are the two abundant oral microbes that form mixed biofilms with augmented virulence, affecting oral health negatively. Understanding the molecular mechanisms of the pathogen interactions and identifying non-toxic compounds that block the growth of biofilms are important steps in the development of effective therapeutic approaches. In this in vitro study we report the inhibition of mono-species or dual-species biofilms of S. gordonii and C. albicans, and decreased levels of biofilm extracellular DNA (eDNA), when biofilms were grown in the presence of gymnemic acids (GAs), a non-toxic small molecule inhibitor of fungal hyphae. Scanning electron microscopic images of biofilms on saliva-coated hydroxyapatite (sHA) surfaces revealed attachment of S. gordonii cells to C. albicans hyphae and to sHA surfaces via nanofibrils only in the untreated control, but not in the GAs-treated biofilms. Interestingly, C. albicans produced fibrillar adhesive structures from hyphae when grown with S. gordonii as a mixed biofilm; addition of GAs abrogated the nanofibrils and reduced the growth of both hyphae and the biofilm. To our knowledge, this is the first report that C. albicans produces adhesive fibrils from hyphae in response to S. gordonii mixed biofilm growth. Semi-quantitative PCR of selected genes related to biofilms from both microbes showed differential expression in control vs. treated biofilms. Further, GAs inhibited the activity of recombinant S. gordonii glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Taken together, our results suggest that S. gordonii stimulates the expression of adhesive materials in C. albicans by direct interaction and/or signaling, and the adhesive material expression can be inhibited by GAs.
Collapse
Affiliation(s)
- Raja Veerapandian
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | | |
Collapse
|
73
|
Negative interaction of Staphylococcus aureus on Fusarium falciforme growth ocular isolates in an in vitro mixed biofilm. Microb Pathog 2019; 135:103644. [DOI: 10.1016/j.micpath.2019.103644] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 07/19/2019] [Accepted: 07/22/2019] [Indexed: 01/27/2023]
|
74
|
Graf K, Last A, Gratz R, Allert S, Linde S, Westermann M, Gröger M, Mosig AS, Gresnigt MS, Hube B. Keeping Candida commensal: how lactobacilli antagonize pathogenicity of Candida albicans in an in vitro gut model. Dis Model Mech 2019; 12:dmm.039719. [PMID: 31413153 PMCID: PMC6765188 DOI: 10.1242/dmm.039719] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 08/02/2019] [Indexed: 12/20/2022] Open
Abstract
The intestine is the primary reservoir of Candida albicans that can cause systemic infections in immunocompromised patients. In this reservoir, the fungus exists as a harmless commensal. However, antibiotic treatment can disturb the bacterial microbiota, facilitating fungal overgrowth and favoring pathogenicity. The current in vitro gut models that are used to study the pathogenesis of C. albicans investigate the state in which C. albicans behaves as a pathogen rather than as a commensal. We present a novel in vitro gut model in which the fungal pathogenicity is reduced to a minimum by increasing the biological complexity. In this model, enterocytes represent the epithelial barrier and goblet cells limit C. albicans adhesion and invasion. Significant protection against C. albicans-induced necrotic damage was achieved by the introduction of a microbiota of antagonistic lactobacilli. We demonstrated a time-, dose- and species-dependent protective effect against C. albicans-induced cytotoxicity. This required bacterial growth, which relied on the presence of host cells, but was not dependent on the competition for adhesion sites. Lactobacillus rhamnosus reduced hyphal elongation, a key virulence attribute. Furthermore, bacterial-driven shedding of hyphae from the epithelial surface, associated with apoptotic epithelial cells, was identified as a main and novel mechanism of damage protection. However, host cell apoptosis was not the driving mechanism behind shedding. Collectively, we established an in vitro gut model that can be used to experimentally dissect commensal-like interactions of C. albicans with a bacterial microbiota and the host epithelial barrier. We also discovered fungal shedding as a novel mechanism by which bacteria contribute to the protection of epithelial surfaces.This article has an associated First Person interview with the joint first authors of the paper.
Collapse
Affiliation(s)
- Katja Graf
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Antonia Last
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Rena Gratz
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Stefanie Allert
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Susanne Linde
- Center for Electron Microscopy Jena University Hospital, Ziegelmühlenweg 1, 07743 Jena, Germany
| | - Martin Westermann
- Center for Electron Microscopy Jena University Hospital, Ziegelmühlenweg 1, 07743 Jena, Germany
| | - Marko Gröger
- Center for Sepsis Control and Care (CSCC), University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Alexander S Mosig
- Center for Sepsis Control and Care (CSCC), University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany.,Institute of Biochemistry II, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Mark S Gresnigt
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany .,Friedrich Schiller University, Fürstengraben 1, 07743 Jena, Germany
| |
Collapse
|
75
|
Miller DP, Fitzsimonds ZR, Lamont RJ. Metabolic Signaling and Spatial Interactions in the Oral Polymicrobial Community. J Dent Res 2019; 98:1308-1314. [PMID: 31356756 DOI: 10.1177/0022034519866440] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Oral supra- and subgingival biofilms are complex communities in which hundreds of bacteria, viruses, and fungi reside and interact. In these social environments, microbes compete and cooperate for resources, such as living space and nutrients. The metabolic activities of bacteria can transform their microenvironment and dynamically influence the fitness and growth of cohabitating organisms. Biofilm communities are temporally and spatially organized largely due to cell-to-cell communication, which promotes synergistic interactions. Metabolic interactions maintain biofilm homeostasis through mutualistic cross-feeding, metabolic syntrophy, and cross-respiration. These interactions include reciprocal metabolite exchanges that promote the growth of physiologically compatible bacteria, processive catabolism of complex substrates, and unidirectional interactions that are globally important for the polymicrobial community. Additionally, oral bacterial interactions can lead to detoxification of oxidative compounds, which will provide protection to the community at large. It has also been established that specific organisms provide terminal electron acceptors to partner species that result in a shift from fermentation to respiration, thus increasing ATP yields and improving fitness. Indeed, many interspecies relationships are multidimensional, and the net outcome can be spatially and temporally dependent. Cross-kingdom interactions also occur as oral yeast are antagonistic to some oral bacteria, while numerous mutualistic interactions contribute to yeast-bacterial colonization, fitness in the oral community, and the pathogenesis of caries. Consideration of this social environment reveals behaviors and phenotypes that are not apparent through the study of microbes in isolation. Here, we provide a comprehensive overview of the metabolic interactions that shape the oral microbial community.
Collapse
Affiliation(s)
- D P Miller
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, USA
| | - Z R Fitzsimonds
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, USA
| | - R J Lamont
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, USA
| |
Collapse
|
76
|
Valentine M, Benadé E, Mouton M, Khan W, Botha A. Binary interactions between the yeast Candida albicans and two gut-associated Bacteroides species. Microb Pathog 2019; 135:103619. [PMID: 31291601 DOI: 10.1016/j.micpath.2019.103619] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/11/2019] [Accepted: 07/06/2019] [Indexed: 01/15/2023]
Abstract
The yeast Candida albicans forms part of the natural gut microbiota of healthy human individuals and its interactions with other microbial symbionts can impact host well-being. We therefore studied binary interactions between potentially pathogenic representatives of the gut-associated bacterial genus Bacteroides and C. albicans using anaerobic bacteria/yeast co-cultures prepared with a quarter-strength brain heart infusion (¼ BHI; 9.25 g/l) broth. We found that, except for minor changes observed in the cell numbers of one out of four C. albicans strains tested, yeast growth was largely unaffected by the presence of the bacteria. In contrast, growth of Bacteroides fragilis NCTC 9343 and Bacteroides vulgatus ATCC 8482 was significantly enhanced in the presence of C. albicans. Supplementation of Bacteroides monocultures with dead Candida albicans CAB 392 cells, containing intact outer cell wall mannan layers, resulted in increased bacterial concentrations. Subsequent culturing of the Bacteroides strains in a liquid minimal medium supplemented with candidal mannan demonstrated that B. vulgatus ATCC 8482, unlike B. fragilis NCTC 9343, utilized the mannan. Furthermore, by reducing the initial oxygen levels in monocultures prepared with ¼ BHI broth, bacterial numbers were significantly enhanced compared to in monocultures prepared with ¼ BHI broth not supplemented with the reducing agent l-cysteine hydrochloride. This suggests that C. albicans can stimulate Bacteroides growth via aerobic respiration and/or antioxidant production. The cell-free supernatant of 24-h-old C. albicans CAB 392 monocultures was also found to increase Bacteroides growth and chloramphenicol sensitivity.
Collapse
Affiliation(s)
- Marisa Valentine
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| | - Eliska Benadé
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| | - Marnel Mouton
- Department of Botany and Zoology, Stellenbosch University, Stellenbosch, South Africa
| | - Wesaal Khan
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| | - Alfred Botha
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
77
|
Brown AO, Graham CE, Cruz MR, Singh KV, Murray BE, Lorenz MC, Garsin DA. Antifungal Activity of the Enterococcus faecalis Peptide EntV Requires Protease Cleavage and Disulfide Bond Formation. mBio 2019; 10:e01334-19. [PMID: 31266876 PMCID: PMC6606811 DOI: 10.1128/mbio.01334-19] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 05/29/2019] [Indexed: 02/04/2023] Open
Abstract
Enterococcus faecalis, a Gram-positive bacterium, and Candida albicans, a polymorphic fungus, are common constituents of the microbiome as well as increasingly problematic causes of infections. Interestingly, we previously showed that these two species antagonize each other's virulence and that E. faecalis inhibition of C. albicans was specifically mediated by EntV. EntV is a bacteriocin encoded by the entV (ef1097) locus that reduces C. albicans virulence and biofilm formation by inhibiting hyphal morphogenesis. In this report, we studied the posttranslational modifications necessary for EntV antifungal activity. First, we show that the E. faecalis secreted enzyme gelatinase (GelE) is responsible for cleaving EntV into its 68-amino-acid, active form and that this process does not require the serine protease SprE. Furthermore, we demonstrate that a disulfide bond that forms within EntV is necessary for antifungal activity. Abrogating this bond by chemical treatment or genetic modification rendered EntV inactive against C. albicans Moreover, we identified the likely catalyst of this disulfide bond, a previously uncharacterized thioredoxin within the E. faecalis genome called DsbA. Loss of DsbA, or disruption of its redox-active cysteines, resulted in loss of EntV antifungal activity. Finally, we show that disulfide bond formation is not a prerequisite for cleavage; EntV cleavage proceeded normally in the absence of DsbA. In conclusion, we present a model in which following secretion, EntV undergoes disulfide bond formation by DsbA and cleavage by GelE in order to generate a peptide capable of inhibiting C. albicansIMPORTANCEEnterococcus faecalis and Candida albicans are among the most important and problematic pathobionts, organisms that normally are harmless commensals but can cause dangerous infections in immunocompromised hosts. In fact, both organisms are listed by the Centers for Disease Control and Prevention as serious global public health threats stemming from the increased prevalence of antimicrobial resistance. The rise in antifungal resistance is of particular concern considering the small arsenal of currently available therapeutics. EntV is a peptide with antifungal properties, and it, or a similar compound, could be developed into a therapeutic alternative, either alone or in combination with existing agents. However, to do so requires understanding what properties of EntV are necessary for its antifungal activity. In this work, we studied the posttranslational processing of EntV and what modifications are necessary for inhibition of C. albicans in order to fill this gap in knowledge.
Collapse
Affiliation(s)
- Armand O Brown
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Carrie E Graham
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Melissa R Cruz
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Kavindra V Singh
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Barbara E Murray
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Michael C Lorenz
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Danielle A Garsin
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
78
|
Bunetel L, Tamanai-Shacoori Z, Martin B, Autier B, Guiller A, Bonnaure-Mallet M. Interactions between oral commensal Candida and oral bacterial communities in immunocompromised and healthy children. J Mycol Med 2019; 29:223-232. [PMID: 31235209 DOI: 10.1016/j.mycmed.2019.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 06/07/2019] [Accepted: 06/07/2019] [Indexed: 01/09/2023]
Abstract
Candida species are usually found as commensal microorganisms in the oral cavity of healthy people. During chemotherapy, cytostatic drugs lead to depletion of the oral flora with the emergence of a dominant bacterial species. The transition from commensal to pathogenic state, further associated with yeast colonization and oral mucositis implies a replacement of the dominant microorganism by Candida albicans. This process goes plausibly through cooperation between C. albicans and bacteria. This study focused on the first step of cooperation between microorganisms isolated from the same oral flora either of leukemic or healthy children. C. albicans isolated from 8/20 children were cultured to display their noninvasive blastosporic yeast form and mixed with their dominant bacteria to study the capacity of planktonic aggregation and the early state of biofilm formation. None of the dominant bacteria opposed the presence of yeast, on the contrary, an interesting cooperation was observed. This behavior is apparently different from that observed when mixing the type strains. In fact, three mutated C. albicans strains display, by their spontaneous ability to form filament, enhanced risks of virulence for leukemic ill carriers. Despite such risks, neither oral nor systemic pathology were observed in ill patients probably because the study was conducted during the first course of chemotherapy and Candida colonization is related to the number of chemotherapeutic cycles. The presence of C. albicans during the initial cycle represents, by its ability to interact with oral bacteria, an actual threat for further cures.
Collapse
Affiliation(s)
- L Bunetel
- CNRS, ISCR UMR 6226, université Rennes, 35000 Rennes, France.
| | | | - B Martin
- Inserm U 1241, Inra, université Rennes, 35043 Rennes, France
| | - B Autier
- Centre hospitalier universitaire Rennes, 35033 Rennes, France
| | - A Guiller
- CNRS - UPJV Edysan FRE 3498, université Amiens, 80000 Amiens, France
| | - M Bonnaure-Mallet
- Inserm U 1241, Inra, université Rennes, 35043 Rennes, France; Centre hospitalier universitaire Rennes, 35033 Rennes, France
| |
Collapse
|
79
|
Almeida-Paes R, Brito-Santos F, Oliveira MME, Bailão AM, Borges CL, Araújo GRDS, Frases S, Soares CMDA, Zancopé-Oliveira RM. Interaction with Pantoea agglomerans Modulates Growth and Melanization of Sporothrix brasiliensis and Sporothrix schenckii. Mycopathologia 2019; 184:367-381. [PMID: 31214857 DOI: 10.1007/s11046-019-00350-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/06/2019] [Indexed: 01/22/2023]
Abstract
Sporothrix brasiliensis and Sporothrix schenckii stand as the most virulent agents of sporotrichosis, a worldwide-distributed subcutaneous mycosis. The origin of Sporothrix virulence seems to be associated with fungal interactions with organisms living in the same environment. To assess this hypothesis, the growth of these two species in association with Pantoea agglomerans, a bacterium with a habitat similar to Sporothrix spp., was evaluated. Growth, melanization, and gene expression of the fungus were compared in the presence or absence of the bacterium in the same culture medium. Both S. brasiliensis and S. schenckii grew in contact with P. agglomerans yielding heavily melanized conidia after 5 days of incubation at 30 °C in Sabouraud agar. This increased melanin production occurred around bacterial colonies, suggesting that fungal melanization is triggered by a diffusible bacterial product, which is also supported by a similar pattern of melanin production during Sporothrix spp. growth in contact with heat-killed P. agglomerans. Growth of P. agglomerans was similar in the presence or absence of the fungus. However, the growth of S. brasiliensis and S. schenckii was initially inhibited, but further enhanced when these species were co-cultured with P. agglomerans. Moreover, fungi were able to use killed bacteria as both carbon and nitrogen sources for growth. Representational difference analysis identified overexpressed genes related to membrane transport when S. brasiliensis was co-cultured with the bacteria. The down-regulation of metabolism-related genes appears to be related to nutrient availability during bacterial exploitation. These findings can lead to a better knowledge on Sporothrix ecology and virulence.
Collapse
Affiliation(s)
- Rodrigo Almeida-Paes
- Laboratório de Micologia, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Fábio Brito-Santos
- Laboratório de Micologia, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Manoel Marques Evangelista Oliveira
- Laboratório de Pesquisa Clínica em Dermatozoonoses, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Alexandre Melo Bailão
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Clayton Luiz Borges
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Glauber Ribeiro de Souza Araújo
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Susana Frases
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Célia Maria de Almeida Soares
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Rosely Maria Zancopé-Oliveira
- Laboratório de Micologia, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
80
|
Obata J, Fujishima K, Nagata E, Oho T. Pathogenic mechanisms of cariogenic Propionibacterium acidifaciens. Arch Oral Biol 2019; 105:46-51. [PMID: 31254840 DOI: 10.1016/j.archoralbio.2019.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/06/2019] [Accepted: 06/17/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Dental caries is one of the most common infectious diseases in humans. Older adults retain more teeth than did earlier generations and thus are at high risk of root caries. The root surface is covered by cementum, which facilitates the spread of caries lesions into dentinal tissues. Propionibacterium acidifaciens has been detected in dentinal caries lesions; however, the pathogenetic mechanisms are not known. The purpose of this study was to investigate the pathogenic mechanisms of cariogenic P. acidifaciens. METHODS Saliva-induced aggregation of P. acidifaciens cells and adherence of the organism to saliva-coated hydroxyapatite were examined. Coaggregation of P. acidifaciens with other bacterial cells and binding of the organism to collagen were examined. Effect of Streptococcus mutans on the biofilm formation by P. acidifaciens was also examined. In addition, the effects of acids on the growth of P. acidifaciens were evaluated. RESULTS P. acidifaciens exhibited strong binding to collagen but weak or moderate interaction with salivary proteins. P. acidifaciens showed weak coaggregation with streptococcal strains and Fusobacerium nucleatum. Biofilm formation by P. acidifaciens was inhibited by S. mutans. Moreover, P. acidifaciens tolerated to self-produced acids up to threshold concentrations. CONCLUSIONS The results suggest that P. acidifaciens can bind to and survive inside dentinal tissue, and its acid production at low pH condition is involved in the development of dentinal caries.
Collapse
Affiliation(s)
- Junko Obata
- Department of Preventive Dentistry, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kei Fujishima
- Division of Preventive Dentistry, Kagoshima University Hospital, Kagoshima, Japan
| | - Emi Nagata
- Division of Preventive Dentistry, Kagoshima University Hospital, Kagoshima, Japan
| | - Takahiko Oho
- Department of Preventive Dentistry, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan.
| |
Collapse
|
81
|
Contributions of Candida albicans Dimorphism, Adhesive Interactions, and Extracellular Matrix to the Formation of Dual-Species Biofilms with Streptococcus gordonii. mBio 2019; 10:mBio.01179-19. [PMID: 31213561 PMCID: PMC6581863 DOI: 10.1128/mbio.01179-19] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Microbial communities have a great impact in health and disease. C. albicans interacts with multiple microorganisms in the oral cavity, frequently forming polymicrobial biofilms. We report on the synergistic interactions between C. albicans and the Gram-positive bacterium S. gordonii, for which we have examined the different contributions of adhesive interactions, filamentation, and the extracellular matrix to the formation of dual-species biofilms. Our results demonstrate that growth in the presence of the bacterium can restore the biofilm-forming ability of different C. albicans mutant strains with defects in adhesion and filamentation. The mixed-species biofilms also show high levels of resistance to antibacterial and antifungal antibiotics, and our results indicate that the fungal biofilm matrix protects bacterial cells within these mixed-species biofilms. Our observations add to a growing body of evidence indicating a high level of complexity in the reciprocal interactions and consortial behavior of fungal/bacterial biofilms. Fungal and bacterial populations coexist in the oral cavity, frequently forming mixed-species biofilms that complicate treatment against polymicrobial infections. However, despite relevance to oral health, the bidirectional interactions between these microbial populations are poorly understood. In this study, we aimed to elucidate the mechanisms underlying the interactions between the fungal species Candida albicans and the bacterial species Streptococcus gordonii as they coexist in mixed-species biofilms. Specifically, the interactions of different C. albicans mutant strains deficient in filamentation (efg1Δ/Δ and brg1Δ/Δ), adhesive interactions (als3Δ/Δ and bcr1Δ/Δ), and production of matrix exopolymeric substances (EPS) (kre5Δ/Δ, mnn9Δ/Δ, rlm1Δ/Δ, and zap1Δ/Δ) were evaluated with S. gordonii under different conditions mimicking the environment in the oral cavity. Interestingly, our results revealed that growth of the biofilm-deficient C. albicansals3Δ/Δ and bcr1Δ/Δ mutant strains in synthetic saliva or with S. gordonii restored their biofilm-forming ability. Moreover, challenging previous observations indicating an important role of morphogenetic conversions in the interactions between these two species, our results indicated a highly synergistic interaction between S. gordonii and the C. albicans filamentation-deficient efg1Δ/Δ and brg1Δ/Δ deletion mutants, which was particularly noticeable when the mixed biofilms were grown in synthetic saliva. Importantly, dual-species biofilms were found to exhibit increase in antimicrobial resistance, indicating that components of the fungal exopolymeric material confer protection to streptococcal cells against antibacterial treatment. Collectively, these findings unravel a high degree of complexity in the interactions between C. albicans and S. gordonii in mixed-species biofilms, which may impact homeostasis in the oral cavity.
Collapse
|
82
|
Candida albicans Augments Staphylococcus aureus Virulence by Engaging the Staphylococcal agr Quorum Sensing System. mBio 2019; 10:mBio.00910-19. [PMID: 31164467 PMCID: PMC6550526 DOI: 10.1128/mbio.00910-19] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Relatively little is known about the complex interactions and signaling events that occur between microbes and even less so about how microbial “cross talk” shapes human health and disease. Candida albicans (a fungus) and Staphylococcus aureus (a bacterium) are formidable human nosocomial pathogens, causing severe morbidity and mortality. Moreover, they are frequently coisolated from central venous catheters and deep-seated infections, including intra-abdominal sepsis. In this work, we have shown that coinfection with C. albicans and S. aureus is highly lethal, leading to >80% mortality by day 1 postinfection, whereas monoinfection with C. albicans or S. aureus does not cause mortality. This infectious synergism is dependent on the expression of staphylococcal alpha-toxin, and secretion of this potent virulence factor is actually augmented by C. albicans via an agr-dependent mechanism. Moreover, prophylactic neutralization of alpha-toxin with a monoclonal antibody is sufficient to elicit protection during coinfection. Therefore, we have demonstrated that a pathogenic fungus can enhance virulence determinants of a bacterium in vivo with devastating consequences to the host. These results have important implications in the surveillance and treatment of polymicrobial disease and highlight the dynamic intersection of environment, pathogens, and host. Candida albicans and Staphylococcus aureus are among the most prevalent nosocomial pathogens that are responsible for severe morbidity and mortality, even with appropriate treatment. Using a murine model of polymicrobial intra-abdominal infection (IAI), we have previously shown that coinfection with these pathogens results in synergistic lethality that is partially dependent on exacerbated prostaglandin signaling, while monomicrobial infection is nonlethal. Therefore, the objective of this study was to identify staphylococcal virulence determinants that drive lethal synergism during polymicrobial IAI. Using the toxigenic S. aureus strain JE2, we observed that coinfection with C. albicans led to a striking 80 to 100% mortality rate within 20 h postinoculation (p.i.) while monomicrobial infections were nonlethal. Use of a green fluorescent protein (GFP)-P3 promoter S. aureus reporter strain revealed enhanced activation of the staphylococcal agr quorum sensing system during in vitro polymicrobial versus monomicrobial growth. Analyses by quantitative real-time PCR (qPCR), Western blot, and toxin functional assays confirmed enhanced agr-associated gene transcription and increases in secreted alpha- and delta-toxins. C. albicans-mediated elevated toxin production and hemolytic activity were determined to be agrA dependent, and genetic knockout and complementation of hla identified alpha-toxin as the key staphylococcal virulence factor driving lethal synergism. Analysis of mono- and polymicrobial infections 8 h p.i. demonstrated equivalent bacterial burdens in the peritoneal cavity but significantly elevated levels of alpha-toxin (3-fold) and the eicosanoid prostaglandin E2 (PGE2) (4-fold) during coinfection. Importantly, prophylactic passive immunization using the monoclonal anti-alpha-toxin antibody MEDI4893* led to significantly improved survival rates compared to those following treatment with isotype control antibody. Collectively, these results define alpha-toxin as an essential virulence determinant during C. albicans-S. aureus IAI and describe a novel mechanism by which a human-pathogenic fungus can augment the virulence of a highly pathogenic bacterium in vivo.
Collapse
|
83
|
The gut mycobiota: insights into analysis, environmental interactions and role in gastrointestinal diseases. Nat Rev Gastroenterol Hepatol 2019; 16:331-345. [PMID: 30824884 DOI: 10.1038/s41575-019-0121-2] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The gut microbiota is a dense and diverse ecosystem that is involved in many physiological functions as well as in disease pathogenesis. It is dominated by bacteria, which have been extensively studied in the past 15 years; however, other microorganisms, such as fungi, phages, archaea and protists, are also present in the gut microbiota. Exploration of the fungal component, namely, the mycobiota, is at an early stage, and several specific technical challenges are associated with mycobiota analysis. The number of fungi in the lower gastrointestinal tract is far lower than that of bacteria, but fungal cells are much larger and much more complex than bacterial cells. In addition, a role of the mycobiota in disease, notably in IBD, is indicated by both descriptive data in humans and mechanistic data in mice. Interactions between bacteria and fungi within the gut, their functional roles and their interplay with the host and its immune system are fascinating areas that researchers are just beginning to investigate. In this Review, we discuss the newest data on the gut mycobiota and explore both the technical aspects of its study and its role in health and gastrointestinal diseases.
Collapse
|
84
|
Pathirana RU, McCall AD, Norris HL, Edgerton M. Filamentous Non- albicans Candida Species Adhere to Candida albicans and Benefit From Dual Biofilm Growth. Front Microbiol 2019; 10:1188. [PMID: 31231324 PMCID: PMC6558389 DOI: 10.3389/fmicb.2019.01188] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/10/2019] [Indexed: 12/18/2022] Open
Abstract
Non-albicans Candida species (NACS) are often isolated along with Candida albicans in cases of oropharyngeal candidiasis. C. albicans readily forms biofilms in conjunction with other oral microbiota including both bacteria and yeast. Adhesion between species is important to the establishment of these mixed biofilms, but interactions between C. albicans and many NACS are not well-characterized. We adapted a real-time flow biofilm model to study adhesion interactions and biofilm establishment in C. albicans and NACS in mono- and co-culture. Out of five NACS studied, only the filamenting species C. tropicalis and C. dubliniensis were capable of adhesion with C. albicans, while C. parapsilosis, C. lusitaniae, and C. krusei were not. Over the early phase (0-4 h) of biofilm development, both mono- and co-culture followed similar kinetics of attachment and detachment events, indicating that initial biofilm formation is not influenced by inter-species interactions. However, the NACS showed a preference for inter-species cell-cell interactions with C. albicans, and at later time points (5-11 h) we found that dual-species interactions impacted biofilm surface coverage. Dual-species biofilms of C. tropicalis and C. albicans grew more slowly than C. albicans alone, but achieved higher surface coverage than C. tropicalis alone. Biofilms of C. dubliniensis with C. albicans increased surface coverage more rapidly than either species alone. We conclude that dual culture biofilm of C. albicans with C. tropicalis or C. dubliniensis offers a growth advantage for both NACS. Furthermore, the growth and maintenance, but not initial establishment, of dual-species biofilms is likely facilitated by interspecies cell-cell adherence.
Collapse
Affiliation(s)
- Ruvini U Pathirana
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, United States
| | - Andrew D McCall
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, United States
| | - Hannah L Norris
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, United States
| | - Mira Edgerton
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
85
|
Heersema LA, Smyth HDC. A Multispecies Biofilm In Vitro Screening Model of Dental Caries for High-Throughput Susceptibility Testing. High Throughput 2019; 8:E14. [PMID: 31151195 PMCID: PMC6631723 DOI: 10.3390/ht8020014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/27/2019] [Accepted: 05/22/2019] [Indexed: 02/08/2023] Open
Abstract
There is a current need to develop and optimize new therapeutics for the treatment of dental caries, but these efforts are limited by the relatively low throughput of relevant in vitro models. The aim of this work was to bridge the 96-well microtiter plate system with a relevant multispecies dental caries model that could be reproducibly grown to allow for the high-throughput screening of anti-biofilm therapies. Various media and inoculum concentrations were assessed using metabolic activity, biomass, viability, and acidity assays to determine the optimal laboratory-controlled conditions for a multispecies biofilm composed of Streptococcus gordonii, Streptococcus mutans, and Candida albicans. The selected model encompasses several of the known fundamental characteristics of dental caries-associated biofilms. The 1:1 RPMI:TSBYE 0.6% media supported the viability and biomass production of mono- and multispecies biofilms best. Kinetic studies over 48 h in 1:1 RPMI:TSBYE 0.6% demonstrated a stable biofilm phase between 10 and 48 h for all mono- and multispecies biofilms. The 1:1:0.1 S. gordonii: S. mutans: C. albicans multispecies biofilm in 1:1 RPMI:TSBYE 0.6% is an excellent choice for a high-throughput multispecies model of dental caries. This high-throughput multispecies model can be used for screening novel therapies and for better understanding the treatment effects on biofilm interactions and stability.
Collapse
Affiliation(s)
- Lara A Heersema
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX 787812, USA.
| | - Hugh D C Smyth
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA.
- The LaMontagne Center for Infectious Disease, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
86
|
Krüger W, Vielreicher S, Kapitan M, Jacobsen ID, Niemiec MJ. Fungal-Bacterial Interactions in Health and Disease. Pathogens 2019; 8:E70. [PMID: 31117285 PMCID: PMC6630686 DOI: 10.3390/pathogens8020070] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/02/2019] [Accepted: 05/16/2019] [Indexed: 12/28/2022] Open
Abstract
Fungi and bacteria encounter each other in various niches of the human body. There, they interact directly with one another or indirectly via the host response. In both cases, interactions can affect host health and disease. In the present review, we summarized current knowledge on fungal-bacterial interactions during their commensal and pathogenic lifestyle. We focus on distinct mucosal niches: the oral cavity, lung, gut, and vagina. In addition, we describe interactions during bloodstream and wound infections and the possible consequences for the human host.
Collapse
Affiliation(s)
- Wibke Krüger
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena 07745, Germany.
| | - Sarah Vielreicher
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena 07745, Germany.
| | - Mario Kapitan
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena 07745, Germany.
- Center for Sepsis Control and Care, Jena 07747, Germany.
| | - Ilse D Jacobsen
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena 07745, Germany.
- Center for Sepsis Control and Care, Jena 07747, Germany.
- Institute of Microbiology, Friedrich Schiller University, Jena 07743, Germany.
| | - Maria Joanna Niemiec
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena 07745, Germany.
- Center for Sepsis Control and Care, Jena 07747, Germany.
| |
Collapse
|
87
|
Bartnicka D, Karkowska-Kuleta J, Zawrotniak M, Satała D, Michalik K, Zielinska G, Bochenska O, Kozik A, Ciaston I, Koziel J, Dutton LC, Nobbs AH, Potempa B, Baster Z, Rajfur Z, Potempa J, Rapala-Kozik M. Adhesive protein-mediated cross-talk between Candida albicans and Porphyromonas gingivalis in dual species biofilm protects the anaerobic bacterium in unfavorable oxic environment. Sci Rep 2019; 9:4376. [PMID: 30867500 PMCID: PMC6416349 DOI: 10.1038/s41598-019-40771-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/22/2019] [Indexed: 12/13/2022] Open
Abstract
The oral cavity contains different types of microbial species that colonize human host via extensive cell-to-cell interactions and biofilm formation. Candida albicans-a yeast-like fungus that inhabits mucosal surfaces-is also a significant colonizer of subgingival sites in patients with chronic periodontitis. It is notable however that one of the main infectious agents that causes periodontal disease is an anaerobic bacterium-Porphyromonas gingivalis. In our study, we evaluated the different strategies of both pathogens in the mutual colonization of an artificial surface and confirmed that a protective environment existed for P. gingivalis within developed fungal biofilm formed under oxic conditions where fungal cells grow mainly in their filamentous form i.e. hyphae. A direct physical contact between fungi and P. gingivalis was initiated via a modulation of gene expression for the major fungal cell surface adhesin Als3 and the aspartic proteases Sap6 and Sap9. Proteomic identification of the fungal surfaceome suggested also an involvement of the Mp65 adhesin and a "moonlighting" protein, enolase, as partners for the interaction with P. gingivalis. Using mutant strains of these bacteria that are defective in the production of the gingipains-the proteolytic enzymes that also harbor hemagglutinin domains-significant roles of these proteins in the formation of bacteria-protecting biofilm were clearly demonstrated.
Collapse
Affiliation(s)
- Dominika Bartnicka
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Justyna Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Marcin Zawrotniak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Dorota Satała
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Kinga Michalik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Gabriela Zielinska
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Oliwia Bochenska
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Andrzej Kozik
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Izabela Ciaston
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Joanna Koziel
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Lindsay C Dutton
- Bristol Dental School, University of Bristol, Bristol, United Kingdom
| | - Angela H Nobbs
- Bristol Dental School, University of Bristol, Bristol, United Kingdom
| | - Barbara Potempa
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Zbigniew Baster
- Institute of Physics; Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - Zenon Rajfur
- Institute of Physics; Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - Jan Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland.
| |
Collapse
|
88
|
Rowan-Nash AD, Korry BJ, Mylonakis E, Belenky P. Cross-Domain and Viral Interactions in the Microbiome. Microbiol Mol Biol Rev 2019; 83:e00044-18. [PMID: 30626617 PMCID: PMC6383444 DOI: 10.1128/mmbr.00044-18] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The importance of the microbiome to human health is increasingly recognized and has become a major focus of recent research. However, much of the work has focused on a few aspects, particularly the bacterial component of the microbiome, most frequently in the gastrointestinal tract. Yet humans and other animals can be colonized by a wide array of organisms spanning all domains of life, including bacteria and archaea, unicellular eukaryotes such as fungi, multicellular eukaryotes such as helminths, and viruses. As they share the same host niches, they can compete with, synergize with, and antagonize each other, with potential impacts on their host. Here, we discuss these major groups making up the human microbiome, with a focus on how they interact with each other and their multicellular host.
Collapse
Affiliation(s)
- Aislinn D Rowan-Nash
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Benjamin J Korry
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Eleftherios Mylonakis
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
89
|
Liu H, Chen H, Sun Y, Zhang X, Lu H, Li J, Cao J, Zhou T. Characterization of the mechanism and impact of staphylokinase on the formation of Candida albicans and Staphylococcus aureus polymicrobial biofilms. J Med Microbiol 2019; 68:355-367. [PMID: 30628885 DOI: 10.1099/jmm.0.000914] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Haiyang Liu
- 1Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
| | - Huale Chen
- 2Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
| | - Yao Sun
- 1Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
| | - Xiaoxiao Zhang
- 1Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
| | - Hong Lu
- 1Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
| | - Jiahui Li
- 1Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
| | - Jianming Cao
- 3School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
| | - Tieli Zhou
- 1Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
| |
Collapse
|
90
|
Deng L, Zou L, Wu J, Liu H, Luo T, Zhou X, Li W, Ren B. Voriconazole inhibits cross-kingdom interactions between Candida albicans and Actinomyces viscosus through the ergosterol pathway. Int J Antimicrob Agents 2019; 53:805-813. [PMID: 30818001 DOI: 10.1016/j.ijantimicag.2019.02.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/11/2019] [Accepted: 02/16/2019] [Indexed: 02/05/2023]
Abstract
Candida albicans and Actinomyces viscosus are prominent microbes associated with dental root caries. The aim of this study was to investigate the effect of C. albicans on A. viscosus biofilms and to identify the mechanisms associated with this interaction. A. viscosus and C. albicans strains (wide-type and mutants) were used to form biofilms in vitro and in vivo, which were subsequently analysed by crystal violet assay and scanning electron microscopy (SEM) to investigate the effect of C. albicans on A. viscosus growth. A viable plate count and survival curve for C. albicans mutants and A. viscosus combinations were used to identify which C. albicans pathway was crucial for cross-kingdom interactions. Voriconazole was used to block their interactions both in vitro and in vivo. SEM, fluorescence in situ hybridisation (FISH), quantitative PCR and survival curve analyses were performed to evaluate the activity of voriconazole on C. albicans and A. viscosus interactions. The biomass and virulence of mixed-species biofilms were significantly enhanced compared with the A. viscosus biofilm alone. However, this was not observed in the mixed-species biofilms with the C. albicans mutant erg11Δ/Δ in vitro and in vivo, indicating that azoles may work on the mixed-species biofilms. As expected, voriconazole can effectively reduce the biomass of mixed-species biofilms. A high concentration of voriconazole (1 µg/mL) reduced the abundance of C. albicans, whilst a low voriconazole concentration (0.25 µg/mL) blocked their interactions similar to the effect of the erg11Δ/Δ mutant. Voriconazole may be a candidate strategy to combat root caries pathogens.
Collapse
Affiliation(s)
- Ling Deng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Ling Zou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Juan Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Haixia Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Tao Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Wei Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China.
| | - Biao Ren
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China.
| |
Collapse
|
91
|
Negrini TDC, Koo H, Arthur RA. Candida–Bacterial Biofilms and Host–Microbe Interactions in Oral Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1197:119-141. [DOI: 10.1007/978-3-030-28524-1_10] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
92
|
Nobbs A, Kreth J. Genetics of sanguinis-Group Streptococci in Health and Disease. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0052-2018. [PMID: 30681069 PMCID: PMC11590441 DOI: 10.1128/microbiolspec.gpp3-0052-2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Indexed: 12/30/2022] Open
Abstract
With the application of increasingly advanced "omics" technologies to the study of our resident oral microbiota, the presence of a defined, health-associated microbial community has been recognized. Within this community, sanguinis-group streptococci, comprising the closely related Streptococcus sanguinis and Streptococcus gordonii, together with Streptococcus parasanguinis, often predominate. Their ubiquitous and abundant nature reflects the evolution of these bacteria as highly effective colonizers of the oral cavity. Through interactions with host tissues and other microbes, and the capacity to readily adapt to prevailing environmental conditions, sanguinis-group streptococci are able to shape accretion of the oral plaque biofilm and promote development of a microbial community that exists in harmony with its host. Nonetheless, upon gaining access to the blood stream, those very same colonization capabilities can confer upon sanguinis-group streptococci the ability to promote systemic disease. This article focuses on the role of sanguinis-group streptococci as the commensurate commensals, highlighting those aspects of their biology that enable the coordination of health-associated biofilm development. This includes the molecular mechanisms, both synergistic and antagonistic, that underpin adhesion to substrata, intercellular communication, and polymicrobial community formation. As our knowledge of these processes advances, so will the opportunities to exploit this understanding for future development of novel strategies to control oral and extraoral disease.
Collapse
Affiliation(s)
- Angela Nobbs
- Bristol Dental School, University of Bristol, Bristol, United Kingdom
| | - Jens Kreth
- Department of Restorative Dentistry, Oregon Health and Science University, Portland, OR 97239
| |
Collapse
|
93
|
Montelongo-Jauregui D, Lopez-Ribot JL. Candida Interactions with the Oral Bacterial Microbiota. J Fungi (Basel) 2018; 4:jof4040122. [PMID: 30400279 PMCID: PMC6308928 DOI: 10.3390/jof4040122] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 10/31/2018] [Accepted: 11/02/2018] [Indexed: 02/06/2023] Open
Abstract
The human oral cavity is normally colonized by a wide range of microorganisms, including bacteria, fungi, Archaea, viruses, and protozoa. Within the different oral microenvironments these organisms are often found as part of highly organized microbial communities termed biofilms, which display consortial behavior. Formation and maintenance of these biofilms are highly dependent on the direct interactions between the different members of the microbiota, as well as on the released factors that influence the surrounding microbial populations. These complex biofilm dynamics influence oral health and disease. In the latest years there has been an increased recognition of the important role that interkingdom interactions, in particular those between fungi and bacteria, play within the oral cavity. Candida spp., and in particular C. albicans, are among the most important fungi colonizing the oral cavity of humans and have been found to participate in these complex microbial oral biofilms. C. albicans has been reported to interact with individual members of the oral bacterial microbiota, leading to either synergistic or antagonistic relationships. In this review we describe some of the better characterized interactions between Candida spp. and oral bacteria.
Collapse
Affiliation(s)
- Daniel Montelongo-Jauregui
- Department of Biology, South Texas Center for Emerging Infections Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA.
| | - Jose L Lopez-Ribot
- Department of Biology, South Texas Center for Emerging Infections Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA.
| |
Collapse
|
94
|
Influence of Streptococcus mitis and Streptococcus sanguinis on virulence of Candida albicans: in vitro and in vivo studies. Folia Microbiol (Praha) 2018; 64:215-222. [PMID: 30232727 DOI: 10.1007/s12223-018-0645-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 09/04/2018] [Indexed: 12/20/2022]
Abstract
The aim was to evaluate in vitro possible interactions, gene expression, and biofilm formation in species of Candida albicans, Streptococcus mitis, and Streptococcus sanguinis and their in vivo pathogenicity. The in vitro analysis evaluated the effects of S. mitis and S. sanguinis on C. albicans's biofilm formation by CFU count, filamentation capacity, and adhesion (ALS1, ALS3, HWP1) and transcriptional regulatory gene (BCR1, CPH1, EFG1) expression. In vivo studies evaluated the pathogenicity of the interaction of the microorganisms on Galleria mellonella, with analyses of the CFU per milliliter count and filamentation. In vitro results indicated that there was an observed decrease in CFU (79.4-71.5%) in multi-species biofilms. The interaction with S. mitis inhibited filamentation, which seems to increase its virulence factor with over-expression of genes ALS1, ALS3, and HWP1 as well the interaction with S. sanguinis as ALS3 and HWP1. S. mitis upregulated BRC1, CPH1, and EFG1. The histological images of in vivo study indicate an increase in the filamentation of C. albicans when in interaction with the other species. It was concluded that S. mitis interaction suggests increased virulence factors of C. albicans, with periods of lower virulence and proto-cooperation in the interaction with S. sanguinis.
Collapse
|
95
|
Tipping the Balance: C. albicans Adaptation in Polymicrobial Environments. J Fungi (Basel) 2018; 4:jof4030112. [PMID: 30231476 PMCID: PMC6162738 DOI: 10.3390/jof4030112] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 09/14/2018] [Accepted: 09/15/2018] [Indexed: 11/18/2022] Open
Abstract
Candida albicans is a pleiomorphic fungus which co-exists with commensal bacteria in mucosal and skin sites of mammalian hosts. It is also a major co-isolated organism from polymicrobial systemic infections, with high potential for morbidity or mortality in immunocompromised patients. Traditionally, resident mucosal bacteria have been thought to antagonize C. albicans in its ability to colonize or cause infection. However, recent investigations have revealed synergistic relationships with certain bacterial species that colonize the same mucosal sites with C. albicans. Such relationships broaden the research landscape in pathogenesis but also contribute to clinical challenges in the prevention or treatment of mucosal candidiasis. This review sheds light on interactions of C. albicans and mucosal bacteria, with special emphasis on the effects of the resident bacterial microbiota on C. albicans physiology as they relate to its adaptation in mucosal sites as a commensal colonizer or as a pathogenic organism.
Collapse
|
96
|
Fungi at the Scene of the Crime: Innocent Bystanders or Accomplices in Oral Infections? CURRENT CLINICAL MICROBIOLOGY REPORTS 2018. [DOI: 10.1007/s40588-018-0100-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
97
|
An In Vitro Model for Candida albicans⁻Streptococcus gordonii Biofilms on Titanium Surfaces. J Fungi (Basel) 2018; 4:jof4020066. [PMID: 29866990 PMCID: PMC6023327 DOI: 10.3390/jof4020066] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 05/31/2018] [Accepted: 06/03/2018] [Indexed: 11/22/2022] Open
Abstract
The oral cavity serves as a nutrient-rich haven for over 600 species of microorganisms. Although many are essential to maintaining the oral microbiota, some can cause oral infections such as caries, periodontitis, mucositis, and endodontic infections, and this is further exacerbated with dental implants. Most of these infections are mixed species in nature and associated with a biofilm mode of growth. Here, after optimization of different parameters including cell density, growth media, and incubation conditions, we have developed an in vitro model of C. albicans–S. gordonii mixed-species biofilms on titanium discs that is relevant to infections of peri-implant diseases. Our results indicate a synergistic effect for the development of biofilms when both microorganisms were seeded together, confirming the existence of beneficial, mutualistic cross-kingdom interactions for biofilm formation. The morphological and architectural features of these dual-species biofilms formed on titanium were determined using scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM). Mixed biofilms formed on titanium discs showed a high level of resistance to combination therapy with antifungal and antibacterial drugs. This model can serve as a platform for further analyses of complex fungal/bacterial biofilms and can also be applied to screening of new drug candidates against mixed-species biofilms.
Collapse
|
98
|
Tshikantwa TS, Ullah MW, He F, Yang G. Current Trends and Potential Applications of Microbial Interactions for Human Welfare. Front Microbiol 2018; 9:1156. [PMID: 29910788 PMCID: PMC5992746 DOI: 10.3389/fmicb.2018.01156] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/14/2018] [Indexed: 01/06/2023] Open
Abstract
For a long time, it was considered that interactions between microbes are only inhibitory in nature. However, latest developments in research have demonstrated that within our environment, several classes of microbes exist which produce different products upon interaction and thus embrace a wider scope of useful and potentially valuable aspects beyond simple antibiosis. Therefore, the current review explores different types of microbial interactions and describes the role of various physical, chemical, biological, and genetic factors regulating such interactions. It further explains the mechanism of action of biofilm formation and role of secondary metabolites regulating bacteria-fungi interaction. Special emphasis and focus is placed on microbial interactions which are important in medicine, food industry, agriculture, and environment. In short, this review reveals the recent contributions of microbial interaction for the benefit of mankind.
Collapse
Affiliation(s)
| | - Muhammad Wajid Ullah
- Department of Biomedical Engineering Huazhong University of Science and Technology, Wuhan, China
| | - Feng He
- College of Life Sciences Huanggang Normal University, Huanggang, China
| | - Guang Yang
- Department of Biomedical Engineering Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
99
|
Abstract
The three main oral diseases of humans, that is, caries, periodontal diseases, and oral candidiasis, are associated with microbiome shifts initiated by changes in the oral environment and/or decreased effectiveness of mucosal immune surveillance. In this review, we discuss the role that microbial-based therapies may have in the control of these conditions. Most investigations on the use of microorganisms for management of oral disease have been conducted with probiotic strains with some positive but very discrete clinical outcomes. Other strategies such as whole oral microbiome transplantation or modification of community function by enrichment with health-promoting indigenous oral strains may offer more promise, but research in this field is still in its infancy. Any microbial-based therapeutics for oral conditions, however, are likely to be only one component within a holistic preventive strategy that should also aim at modification of the environmental influences responsible for the initiation and perpetuation of microbiome shifts associated with oral dysbiosis.
Collapse
|
100
|
Coassociation between Group B Streptococcus and Candida albicans Promotes Interactions with Vaginal Epithelium. Infect Immun 2018; 86:IAI.00669-17. [PMID: 29339458 DOI: 10.1128/iai.00669-17] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/26/2017] [Indexed: 12/13/2022] Open
Abstract
Group B Streptococcus (GBS) is a leading cause of neonatal sepsis, pneumonia, and meningitis worldwide. In the majority of cases, GBS is transmitted vertically from mother to neonate, making maternal vaginal colonization a key risk factor for neonatal disease. The fungus Candida albicans is an opportunistic pathogen of the female genitourinary tract and the causative agent of vaginal thrush. Carriage of C. albicans has been shown to be an independent risk factor for vaginal colonization by GBS. However, the nature of interactions between these two microbes is poorly understood. This study provides evidence of a reciprocal, synergistic interplay between GBS and C. albicans that may serve to promote their cocolonization of the vaginal mucosa. GBS strains NEM316 (serotype III) and 515 (serotype Ia) are shown to physically interact with C. albicans, with the bacteria exhibiting tropism for candidal hyphal filaments. This interaction enhances association levels of both microbes with the vaginal epithelial cell line VK2/E6E7. The ability of GBS to coassociate with C. albicans is dependent upon expression of the hypha-specific adhesin Als3. In turn, expression of GBS antigen I/II family adhesins (Bsp polypeptides) facilitates this coassociation and confers upon surrogate Lactococcus lactis the capacity to exhibit enhanced interactions with C. albicans on vaginal epithelium. As genitourinary tract colonization is an essential first step in the pathogenesis of GBS and C. albicans, the coassociation mechanism reported here may have important implications for the risk of disease involving both of these pathogens.
Collapse
|