scholarly journals Candida albicans and Staphylococcus aureus Pathogenicity and Polymicrobial Interactions: Lessons beyond Koch’s Postulates

2019 ◽  
Vol 5 (3) ◽  
pp. 81 ◽  
Author(s):  
Olivia A. Todd ◽  
Brian M Peters

While Koch’s Postulates have established rules for microbial pathogenesis that have been extremely beneficial for monomicrobial infections, new studies regarding polymicrobial pathogenesis defy these standards. The explosion of phylogenetic sequence data has revolutionized concepts of microbial interactions on and within the host. However, there remains a paucity of functional follow-up studies to delineate mechanisms driven by such interactions and how they shape health or disease. That said, one particular microbial pairing, the fungal opportunist Candida albicans and the bacterial pathogen Staphylococcus aureus, has received much attention over the last decade. Therefore, the objective of this review is to discuss the multi-faceted mechanisms employed by these two ubiquitous human pathogens during polymicrobial growth, including how they: establish and persist in inter-Kingdom biofilms, tolerate antimicrobial therapy, co-invade host tissue, exacerbate quorum sensing and staphylococcal toxin production, and elicit infectious synergism. Commentary regarding new challenges and remaining questions related to future discovery of this fascinating fungal–bacterial interaction is also provided.

mSphere ◽  
2019 ◽  
Vol 4 (6) ◽  
Author(s):  
Olivia A. Todd ◽  
Mairi C. Noverr ◽  
Brian M. Peters

ABSTRACT Candida albicans and Staphylococcus aureus are common causes of nosocomial infections with severe morbidity and mortality. Murine polymicrobial intra-abdominal infection (IAI) with C. albicans and S. aureus results in acute mortality dependent on the secreted cytolytic effector alpha-toxin. Here, we confirmed that alpha-toxin is elevated during polymicrobial growth compared to monomicrobial growth in vitro. Therefore, this study sought to unravel the mechanism by which C. albicans drives enhanced staphylococcal alpha-toxin production. Using a combination of functional and genetic approaches, we determined that an intact agr quorum sensing regulon is necessary for enhanced alpha-toxin production during coculture and that a secreted candidal factor likely is not implicated in elevating agr activation. As the agr system is pH sensitive, we observed that C. albicans raises the pH during polymicrobial growth and that this correlates with increased agr activity and alpha-toxin production. Modulation of the pH could predictably attenuate or activate agr activity during coculture. By using a C. albicans mutant deficient in alkalinization (stp2Δ/Δ), we confirmed that modulation of the extracellular pH by C. albicans can drive agr expression and toxin production. Additionally, the use of various Candida species (C. glabrata, C. dubliniensis, C. tropicalis, C. parapsilosis, and C. krusei) demonstrated that those capable of raising the extracellular pH correlated with elevated agr activity and alpha-toxin production during coculture. Overall, we demonstrate that alkalinization of the extracellular pH by the Candida species leads to sustained activation of the staphylococcal agr system. IMPORTANCE Candida albicans and Staphylococcus aureus are commonly coisolated from central venous catheters and deep-seated infections, including intra-abdominal sepsis. Thus, they represent a significant cause of nosocomial morbidity and mortality. Yet how these organisms behave in the context of polymicrobial growth remains poorly understood. In this work, we set out to determine the mechanism by which activation of the staphylococcal agr quorum sensing system and production of its major virulence effector alpha-toxin is enhanced during coculture with C. albicans. Surprisingly, we likely ruled out that a secreted candidal factor drives this process. Instead, we demonstrated that alkalinization of the extracellular milieu by C. albicans and other Candida species correlated with elevated agr activity. Thus, we propose a mechanism where modulation of the extracellular pH by fungal opportunists can indirectly alter virulence of a bacterial pathogen. Uncovering molecular events that drive interkingdom pathogenicity mechanisms may enhance surveillance and treatment for devastating polymicrobial infections.


2009 ◽  
Vol 8 (5) ◽  
pp. 732-737 ◽  
Author(s):  
Emmanouil Tampakakis ◽  
Anton Y. Peleg ◽  
Eleftherios Mylonakis

ABSTRACT Candida albicans is an opportunistic human fungal pathogen that normally resides in the gastrointestinal tract and on the skin as a commensal but can cause life-threatening invasive disease. Salmonella enterica serovar Typhimurium is a gram-negative bacterial pathogen that causes a significant amount of gastrointestinal infection in humans. Both of these organisms are also pathogenic to the nematode Caenorhabditis elegans, causing a persistent gut infection leading to worm death. In the present study, we used a previously developed C. elegans polymicrobial infection model to assess the interactions between S. Typhimurium and C. albicans. We observed that when C. elegans is infected with C. albicans and serovar Typhimurium, C. albicans filamentation is inhibited. The inhibition of C. albicans filamentation by S. Typhimurium in C. elegans appeared to be mediated by a secretary molecule, since filter-sterilized bacterial supernatant was able to inhibit C. albicans filamentation. In vitro coculture assays under planktonic conditions showed that S. Typhimurium reduces the viability of C. albicans, with greater effects seen at 37°C than at 30°C. Interestingly, S. Typhimurium reduces the viability of both yeast and filamentous forms of C. albicans, but the killing appeared more rapid for the filamentous cells. The antagonistic interaction was also observed in a C. albicans biofilm environment. This study describes the interaction between two diverse human pathogens that reside within the gastrointestinal tract and shows that the prokaryote, S. Typhimurium, reduces the viability of the eukaryote, C. albicans. Identifying the molecular mechanisms of this interaction may provide important insights into microbial pathogenesis.


2012 ◽  
Vol 57 (1) ◽  
pp. 74-82 ◽  
Author(s):  
Brian M. Peters ◽  
Raven M. Ward ◽  
Hallie S. Rane ◽  
Samuel A. Lee ◽  
Mairi C. Noverr

ABSTRACTCandida albicans, an opportunistic fungus, andStaphylococcus aureus, a bacterial pathogen, are two clinically relevant biofilm-forming microbes responsible for a majority of catheter-related infections, with such infections often resulting in catheter loss and removal. Not only do these pathogens cause a substantial number of nosocomial infections independently, but also they are frequently found coexisting as polymicrobial biofilms on host and environmental surfaces. Antimicrobial lock therapy is a current strategy to sterilize infected catheters. However, the robustness of this technique against polymicrobial biofilms has remained largely untested. Due to its antimicrobial activity, safety, stability, and affordability, we tested the hypothesis that ethanol (EtOH) could serve as a potentially efficacious catheter lock solution againstC. albicansandS. aureusbiofilms. Therefore, we optimized the dose and time necessary to achieve killing of both monomicrobial and polymicrobial biofilms formed on polystyrene and silicone surfaces in a static microplate lock therapy model. Treatment with 30% EtOH for a minimum of 4 h was inhibitory for monomicrobial and polymicrobial biofilms, as evidenced by XTT {sodium 2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-5-[(phenylamino)carbonyl]-2H-tetrazolium hydroxide inner salt} metabolic activity assays and confocal microscopy. Experiments to determine the regrowth of microorganisms on silicone after EtOH treatment were also performed. Importantly, incubation with 30% EtOH for 4 h was sufficient to kill and inhibit the growth ofC. albicans, while 50% EtOH was needed to completely inhibit the regrowth ofS. aureus. In summary, we have systematically defined the dose and duration of EtOH treatment that are effective against and prevent regrowth ofC. albicansandS. aureusmonomicrobial and polymicrobial biofilms in anin vitrolock therapy model.


Pathogens ◽  
2021 ◽  
Vol 10 (8) ◽  
pp. 1036
Author(s):  
Yao Hu ◽  
Yulong Niu ◽  
Xingchen Ye ◽  
Chengguang Zhu ◽  
Ting Tong ◽  
...  

The mixed species of Staphylococcus aureus and Candida albicans can cause infections on skin, mucosa or bloodstream; however, mechanisms of their cross-kingdom interactions related to pathogenesis and drug resistance are still not clear. Here an increase of S. aureus proliferation and biofilm formation was observed in S. aureus and C. albicans dual-species culture, and the synergistic pathogenic effect was then confirmed in both local (cutaneous abscess) and systemic infection (peritonitis) murine models. According to the transcriptome analysis of the dual-species culture, virulence factors of S. aureus were significantly upregulated. Surprisingly, the beta-lactams and vancomycin-resistant genes in S. aureus as well as azole-resistant genes in C. albicans were also significantly increased. The synergistic effects on drug resistance to both antibacterial and antifungal agents were further proved both in vitro and in cutaneous abscess and peritonitis murine models treated by methicillin, vancomycin and fluconazole. The synergistic interactions between S. aureus and C. albicans on pathogenesis and drug resistance highlight the importance of targeting the microbial interactions in polyspecies-associated infections.


mBio ◽  
2018 ◽  
Vol 9 (1) ◽  
Author(s):  
Lamia Harper ◽  
Divya Balasubramanian ◽  
Elizabeth A. Ohneck ◽  
William E. Sause ◽  
Jessica Chapman ◽  
...  

ABSTRACTStaphylococcus aureusis a versatile bacterial pathogen that can cause significant disease burden and mortality. Like other pathogens,S. aureusmust adapt to its environment to produce virulence factors to survive the immune responses evoked by infection. Despite the importance of environmental signals forS. aureuspathogenicity, only a limited number of these signals have been investigated in detail for their ability to modulate virulence. Here we show that pyruvate, a central metabolite, causes alterations in the overall metabolic flux ofS. aureusand enhances its pathogenicity. We demonstrate that pyruvate induces the production of virulence factors such as the pore-forming leucocidins and that this induction results in increased virulence of community-acquired methicillin-resistantS. aureus(CA-MRSA) clone USA300. Specifically, we show that an efficient “pyruvate response” requires the activation ofS. aureusmaster regulators AgrAC and SaeRS as well as the ArlRS two-component system. Altogether, our report further establishes a strong relationship between metabolism and virulence and identifies pyruvate as a novel regulatory signal for the coordination of theS. aureusvirulon through intricate regulatory networks.IMPORTANCEDelineation of the influence of host-derived small molecules on the makeup of human pathogens is a growing field in understanding host-pathogen interactions.S. aureusis a prominent pathogen that colonizes up to one-third of the human population and can cause serious infections that result in mortality in ~15% of cases. Here, we show that pyruvate, a key nutrient and central metabolite, causes global changes to the metabolic flux ofS. aureusand activates regulatory networks that allow significant increases in the production of leucocidins. These and other virulence factors are critical forS. aureusto infect diverse host niches, initiate infections, and effectively subvert host immune responses. Understanding how environmental signals, particularly ones that are essential to and prominent in the human host, affect virulence will allow us to better understand pathogenicity and consider more-targeted approaches to tackling the currentS. aureusepidemic.


Microbial interactions in Staphylococcus aureus–Candida albicans dual-species biofilms is a relevant research topic given the significant contribution of these microorganisms to hospital-acquired infections. Therefore, the purpose of our investigation was to study the interaction of opportunistic C. albicans and S. aureus in vivo and in vitro, both with the participation of normal microflora and in mice with antibacterial dysbiosis. The study of mentioned interactions was carried out on 100 white male mice weighing approximately 18 grams in vivo and using smears prepared from the grown mixed cultures of C. albicans and S. aureus and the Japan JEM 1400 transmission electron microscope for the purpose of electron microscopic study of microorganisms in vitro. Healthy mice forming control groups and mice with antibiotic-induced dysbiosis (after introduction of vancomycin, gentamicin, ampicillin) were divided into groups to create a mono- and associative infection: Ι group was given 1×107 CFU of C. albicans, II group – 1×108 CFU of S. aureus, and III group – a mixture of specified concentrations of C. albicans and S. aureus in the same proportion. Microorganisms causing monoinfection were being isolated from the body of animals treated with antibiotics till the end of the experiments in large quantities unlike in case of the healthy mice. Co-inoculation of these microbes in the same dose to animals (co-infection), which were injected with antibiotics, turned out to be fatal for them, whereas an adhesive bond was seen between the cells of C. albicans vs. S. aureus in vitro. As can be seen, such bacterial-fungal co-infection reduce substantially the effectiveness of antibiotic therapy and the likelihood of successful treatment and can not be ignored when choosing the appropriate treatment.


Plant Disease ◽  
2011 ◽  
Vol 95 (3) ◽  
pp. 354-354 ◽  
Author(s):  
A. S. Corrêa ◽  
A. B. Rocha ◽  
S. A. Willani ◽  
J. M. Dariva ◽  
M. V. Souza ◽  
...  

Yellow stunt, an emerging disease of tobacco (Nicotiana tabacum), has become increasingly prevalent in tobacco-growing regions in southern Brazil. Major symptoms are moderate to severe stunting, yellowing of leaves, severe wilting, darkened roots, necrosis of stem tissue directly above the soil line, and plant death. Phytophthora glovera was first proposed in 1999 as the primary causal agent of yellow stunt (1), but since then, there has been no data or completion of Koch's postulates to support this. Fifty-six isolates of fungi and fungus-like organisms were obtained from stem and root samples of tobacco plants with typical symptoms of yellow stunt in the Brazilian States of Rio Grande do Sul, Santa Catarina, and Paraná during the growing seasons of 2004/05 to 2006/07. They were identified to species level by analysis of the morphological characteristics (2) and sequence of rDNA internal transcribed spacer regions 1 and 2 (ITS1 and ITS2) (3). The isolates were identified as Pythium dissotocum (29), Fusarium oxysporum (10), P. graminicola (5), Rhizoctonia solani (4), F. solani (3), P. ultimum (3), P. deliense (1), and P. inflatum (1). The ITS sequences of the 29 isolates identified as P. dissotocum were identical. The nucleotide sequence of one isolate, LFM27/2005, has been deposited in GenBank (GQ495982). Analysis of ITS sequences alone was not sufficient to differentiate this isolate from other species in the Pythium subclade B2, such as P. coloratum, P. lutarium, P. marinum, or P. diclinum. However, the combination of morphological and cultural characteristics (2) and sequence data support our identification of LFM27/2005 and similar isolates as P. dissotocum. Colonies of LFM27/2005 on cornmeal agar had filamentous sporangia and formed slightly inflated, dendroid structures. Zoospores formed at 5°C. Daily growth rate on potato carrot agar was 13 mm at 25°C. The oogonia (22 μm in diameter) were nonornamented and either intercalary or terminal. Antheridia, commonly 1 to 2 per oogonium, were sessile, born on unbranched stalks, and either monoclinous or diclinous. Aplerotic or nearly plerotic oospores measured 20 μm in diameter with a smooth wall 2.5 μm thick. Pathogenicity tests for each pathogen were performed in a greenhouse at ~24°C in pots filled with pine bark substrate infested with inoculum at the time Burley tobacco plants showed five expanded leaves. Each test consisted of five plants and was repeated three times. Inoculum for one to three isolates representative of each pathogen was prepared by growing 2-month-old cultures at 28°C in the dark for 7 days on potato dextrose agar medium overlaid with three sterile oat kernels. Noninfested oat kernels were used for control plants. Forty days after inoculation, only plants inoculated with isolates of P. dissotocum exhibited all symptoms associated with yellow stunt. P. inflatum and R. solani did not induce yellow stunt symptoms and the others induced only wilting and root rot. P. dissotocum was recovered from an inoculated, symptomatic plant, fulfilling Koch's postulates. Its morphology was identical to isolates obtained from original field samples. The results demonstrate the association of isolates of P. dissotocum with tobacco yellow stunt in Brazil. References: (1) H. D. Shew et al. Phytopathology (Abstr.) 89(suppl):S72, 1999. (2) A. J. van der Plaats-Niterink. Stud. Mycol. 21:1, 1981. (3) T. J. White et al. Page 315 in: PCR Protocols: A Guide to Methods and Applications. Academic Press, San Diego, CA, 1990.


2021 ◽  
Vol 12 ◽  
Author(s):  
Kara R. Eichelberger ◽  
James E. Cassat

Successful pathogens require metabolic flexibility to adapt to diverse host niches. The presence of co-infecting or commensal microorganisms at a given infection site can further influence the metabolic processes required for a pathogen to cause disease. The Gram-positive bacterium Staphylococcus aureus and the polymorphic fungus Candida albicans are microorganisms that asymptomatically colonize healthy individuals but can also cause superficial infections or severe invasive disease. Due to many shared host niches, S. aureus and C. albicans are frequently co-isolated from mixed fungal-bacterial infections. S. aureus and C. albicans co-infection alters microbial metabolism relative to infection with either organism alone. Metabolic changes during co-infection regulate virulence, such as enhancing toxin production in S. aureus or contributing to morphogenesis and cell wall remodeling in C. albicans. C. albicans and S. aureus also form polymicrobial biofilms, which have greater biomass and reduced susceptibility to antimicrobials relative to mono-microbial biofilms. The S. aureus and C. albicans metabolic programs induced during co-infection impact interactions with host immune cells, resulting in greater microbial survival and immune evasion. Conversely, innate immune cell sensing of S. aureus and C. albicans triggers metabolic changes in the host cells that result in an altered immune response to secondary infections. In this review article, we discuss the metabolic programs that govern host-pathogen interactions during S. aureus and C. albicans co-infection. Understanding C. albicans-S. aureus interactions may highlight more general principles of how polymicrobial interactions, particularly fungal-bacterial interactions, shape the outcome of infectious disease. We focus on how co-infection alters microbial metabolism to enhance virulence and how infection-induced changes to host cell metabolism can impact a secondary infection.


2021 ◽  
Author(s):  
◽  
Olivia Todd ◽  

Candida albicans, an opportunistic fungal pathogen, and Staphylococcus aureus, a ubiquitous pathogenic bacterium, are among the most prevalent causes of nosocomial infections and cause severe morbidity and mortality. Moreover, they are frequently coisolated from central venous catheters and deep-seated infections, including intra-abdominal sepsis. Relatively little is known about the complex interactions and signaling events that occur between microbes and even less so how microbial “cross-talk” shapes human health and disease. Using a murine model of polymicrobial intra-abdominal infection (IAI), we have previously shown that coinfection with C. albicans and S. aureus leads to synergistic lethality whereas monomicrobial infection is nonlethal. Therefore, we aimed to identify staphylococcal virulence determinants that drive lethal synergism in polymicrobial IAI. Using the toxigenic S. aureus strain JE2, we observed that co-infection with C. albicans led to a striking 80-100% mortality rate within 20 h p.i while monomicrobial infections were non-lethal. Use of a 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 qPCR, Western blot, and toxin functional assays confirmed enhanced agr-associated gene transcription and increases in secreted α- and δ-toxins. C. albicans-mediated elevated toxin production and hemolytic activity was determined to be agrA-dependent and genetic knockout and complementation of hla identified ⍺-toxin as the key staphylococcal virulence factor driving lethal synergism. Analysis of mono- and polymicrobial infection 8 h p.i. demonstrated equivalent bacterial burden in the peritoneal cavity, but significantly elevated levels of α-toxin (3-fold) and the eicosanoid PGE2 (4-fold) during co-infection. Importantly, prophylactic passive vaccination using the monoclonal anti-⍺-toxin antibody MEDI4893* led to significantly improved survival rates as compared to treatment with isotype control antibody. Collectively, these results define α-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. We next sought to unravel the mechanism by which C. albicans drives enhanced staphylococcal ⍺-toxin production. Using a combination of functional and genetic approaches, we determined that an intact agr quorum sensing regulon is necessary for enhanced ⍺-toxin production during coculture and that a secreted candidal factor likely is not implicated in elevating agr activation. As the agr system is pH sensitive, we observed that C. albicans raises the pH during polymicrobial growth and that this correlates with increased agr activity and ⍺-toxin production. By using a C. albicans mutant deficient in alkalinization (stp2Δ/Δ), we confirmed that modulation of the extracellular pH by C. albicans can drive agr expression and toxin production. Additionally, the use of various Candida species (C. glabrata, C. dubliniensis, C. tropicalis, C. parapsilosis, and C. krusei) demonstrated that those capable of raising the extracellular pH correlated with elevated agr activity and ⍺-toxin production during coculture. Overall, we demonstrated that alkalinization of the extracellular pH by the Candida species leads to sustained activation of the staphylococcal agr system. Finally, we correlated ⍺-toxin production with significant increases in biomarkers of liver and kidney damage during coinfection and determined that functional toxin was required for morbidity and mortality. We next sought to determine the candidal effector(s) mediating this enhanced virulence by employing an unbiased screening approach. C. albicans transcription factor mutants were evaluated for their ability to induce S. aureus agr activation in polymicrobial culture. Incredibly, we identified several mutants that displayed defects in augmenting S. aureus agr activity in vitro. Two of the mutants failed to completely synergize with S. aureus in vivo and further analysis revealed the necessity of the uncharacterized C. albicans transcription factor, ZCF13, in driving enhanced toxin production both in vitro and in vivo. Collectively, we identified a novel effector by which C. albicans augments S. aureus virulence and identified a potential mechanism of fungal-bacterial lethal synergism.


2016 ◽  
Vol 1 (01) ◽  
Author(s):  
Vemavarapu Bhaskara Rao ◽  
Kandlagunta Guru Prasad ◽  
Krishna Naragani ◽  
Vijayalakshmi Muvva

The air dried rhizosphere soil samples pretreated with calcium carbonate was employed for the isolation of actinomycete strains. Serial dilution plate technique was used for the isolation of actinomycetes. A total of 20 actinomycete strains designated as BS1-BS20 were isolated from the rhizosphere of medicinal plant Clitoria ternatea. All the 20 strains were subjected to primary screening for antimicrobial activity. Among the 20 strains screened, 10 strains exhibited high antimicrobial spectrum against Staphylococcus aureus, Escherichia coli and Candida albicans.


Sign in / Sign up

Export Citation Format

Share Document