Next Article in Journal
Lipid Species in the GI Tract are Increased by the Commensal Fungus Candida albicans and Decrease the Virulence of Clostridioides difficile
Next Article in Special Issue
Susceptibility of the Candida haemulonii Complex to Echinocandins: Focus on Both Planktonic and Biofilm Life Styles and a Literature Review
Previous Article in Journal
Bloodstream Infection by Saccharomyces cerevisiae in Two COVID-19 Patients after Receiving Supplementation of Saccharomyces in the ICU
Previous Article in Special Issue
Biofilm Formed by Candida haemulonii Species Complex: Structural Analysis and Extracellular Matrix Composition
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Fungal Quorum-Sensing Molecules: A Review of Their Antifungal Effect against Candida Biofilms

by
Renátó Kovács
1,2,* and
László Majoros
1
1
Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
2
Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
*
Author to whom correspondence should be addressed.
J. Fungi 2020, 6(3), 99; https://doi.org/10.3390/jof6030099
Submission received: 4 June 2020 / Revised: 28 June 2020 / Accepted: 30 June 2020 / Published: 2 July 2020
(This article belongs to the Special Issue Fungal Biofilms 2020)

Abstract

:
The number of effective therapeutic strategies against biofilms is limited; development of novel therapies is urgently needed to treat a variety of biofilm-associated infections. Quorum sensing is a special form of microbial cell-to-cell communication that is responsible for the release of numerous extracellular molecules, whose concentration is proportional with cell density. Candida-secreted quorum-sensing molecules (i.e., farnesol and tyrosol) have a pivotal role in morphogenesis, biofilm formation, and virulence. Farnesol can mediate the hyphae-to-yeast transition, while tyrosol has the opposite effect of inducing transition from the yeast to hyphal form. A number of questions regarding Candida quorum sensing remain to be addressed; nevertheless, the literature shows that farnesol and tyrosol possess remarkable antifungal and anti-biofilm effect at supraphysiological concentration. Furthermore, previous in vitro and in vivo data suggest that they may have a potent adjuvant effect in combination with certain traditional antifungal agents. This review discusses the most promising farnesol- and tyrosol-based in vitro and in vivo results, which may be a foundation for future development of novel therapeutic strategies to combat Candida biofilms.

1. Introduction

It has been estimated that there are 2.2 to 3.8 million fungal species worldwide; however, approximately 300 species have been described to cause human disease [1]. Candida species are among the most common human fungal pathogens. The annual incidence rate of Candida-associated bloodstream infections ranged from 9.5 to 14.4 per 100,000 in the United States of America [2]. This value ranged from 1.4 to 5.7 per 100,000 in Europe, depending on the country [3]. In the last two decades, the prevalence of resistant fungal infections has been steadily increasing due to the widespread use of antifungals in agriculture and veterinary and human medicine [4,5]. Global warming and anthropogenic effects have resulted in the emergence of previously little-known, potentially multi-resistant fungal pathogens in clinical practice, such as Candida auris, azole-resistant Aspergillus spp., or Lomentospora prolificans. These emerging pathogens have caused further challenges for therapy [6,7].
Several fungal species can switch their morphology from yeast to hyphal or pseudohyphal forms, which is coupled with biofilm formation and plays a pivotal role both in fungal virulence and in resistance to antifungals [8,9,10]. The increased number of biofilm-associated infections is exacerbated by a paucity of antifungal agents or therapeutic strategies in development that have unique mechanisms of action or possess alternative approaches, respectively [11]. Currently, the most promising antifungal agents are already in Phase 3 including ibrexafungerp [12], rezafungin [13], super bioavailable itraconazole [14], and VT-1161 [15]. Recently investigated alternative therapeutic approaches involve high-dose therapy with available antifungal agents [16,17,18], antifungal lock therapy [19], and combination-based therapies [20,21]. Based on in vitro and in vivo data, echinocandins and amphotericin B solutions are the most promising combination-based and/or antifungal lock strategies [19]. Further innovative therapeutic approaches may be the natural products-based treatments [22,23]. One of the more well-studied compounds is carbohydrate-derived fulvic acid as a heat stable colloidal material, which has an inhibitory effect on Candida and bacterial biofilm formation [24]. Moreover, a further alternative approach is the treatments disrupting quorum sensing. The usage of quorum sensing molecules at supraphysiological concentration may adversely influence the cell-to-cell communication in biofilms [25,26,27]. In addition, the quorum-sensing system can be inactivated, which is generally known as quorum quenching. Quorum quenching can be triggered by inhibiting the production of quorum sensing molecules, their detection by receptors or their degradation [28].
In this review, a detailed overview is provided of the recent status of quorum-sensing molecule-based therapeutic approaches and their potential future perspectives against Candida biofilms.

2. The Medical Importance of Candida Biofilms

Despite their importance, Candida biofilms remain a relatively underappreciated and understudied area. Therefore, effective therapeutic strategies against these sessile communities remain scarce. Biofilms are usually found in medical devices such as joint prostheses, pacemakers, urinary and central venous catheters, dentures, and mechanical heart valves, hindering the eradication of Candida infections [10]. In addition, several chronic Candida-related diseases are also associated with biofilm development [29]. Biofilm formation on the vaginal mucosa has been observed in in vivo models of vulvovaginal candidiasis [30]. Oral- and oesophageal mucosae-associated biofilms are a very important contributor to oral diseases caused by Candida species; gastrointestinal and urogenital tracts are also common sites of Candida-associated opportunistic infections [31]. Candida is one of the most commonly identified fungal genera in wounds whose environment can also promote the formation of biofilms [32]. A series of recent studies has indicated that strains defective in hyphal formation display significantly milder symptoms, highlighting the role of biofilm formation in pathogenesis of these chronic or recurrent infections [30,33].
These sessile communities exhibit five- to eightfold higher resistance to all licenced antifungal drugs when compared to their planktonic counterparts [10]. This high rate of resistance can be explained by the increased metabolic activity of cells in the early development phase of biofilm formation [10]. On the other hand, dormant, non-proliferating persister cells have also been observed, especially in mature biofilms, that have demonstrated high tolerance to antifungals [34]. Furthermore, the various Candida species can produce dense extracellular polymeric substances which serve as a solid barrier to prevent the diffusion of antifungal drugs and account for almost 90% of the biofilm dry mass [10]. As has been previously reported in the literature, sessile Candida communities exhibit an altered gene expression profile, including the upregulation of CDR and MDR genes which encode azole resistance transporter proteins, and pose further challenges for treatment [35].
To date, there is no definitive therapy against Candida biofilms; nevertheless, there are several promising in vitro, in vivo and clinical results. The increasing number of resistant Candida species and isolates highlight the need for new molecules with new targets. Alternative therapeutic approaches against multidrug-resistant fungal biofilms may be the result of a combination of traditional antifungal agents with quorum-sensing molecules [36].

3. Fungal Quorum Sensing

A major mechanism of microbial communication is a population density-dependent stimulus-response system called quorum sensing. This process occurs by the continuous release and monitoring of low molecular weight hormone-like secreted molecules (quorum-sensing molecules), which are not elementary in the central metabolism but have a variety of biological activities. The concentration of these quorum-sensing molecules is proportional with the size of population; after reaching a critical threshold, a response is triggered leading to the coordinated expression or repression of quorum sensing-related target genes [37].
In the fungal kingdom, quorum sensing was a relatively unknown phenomenon until Hornby et al. (2001) described the effect of the isoprenoid farnesol on Candida albicans morphogenesis; this opened a new branch of science focusing on fungal quorum sensing [38]. At the same time, quorum sensing has been already reported in Aspergillus spp. [39] and Penicillium spp. [40]. To date, four main quorum-sensing molecules were described including farnesol, tyrosol, phenylethanol, and tryptophol, which have a remarkable effect on the regulation of morphogenesis (yeast to hyphae transition and vice versa), initiation of fungal apoptosis, and virulence [41].
Recently, several authors reported that certain quorum-sensing molecules may generate oxidative stress, especially at supraphysiological concentrations, which may have an antifungal effect [42,43,44,45]. The majority of data concerning fungal quorum sensing molecule-related therapeutic potential derived from C. albicans experiments, and these results cannot be always directly extrapolated to non-albicans species. Recently, the number of studies dealing with the effect of quorum-sensing molecules on non-albicans species has steadily increased, supporting the comprehensive understanding of the in vitro and in vivo antifungal effects exerted by these molecules.

4. Farnesol

4.1. Physiological Effect of Farnesol in Candida Species

Farnesol (3,7,11-trimethyl-2,6,10-dodecatriene-1-ol) was the first described Candida-derived quorum sensing molecule; it is released in C. albicans as a side product of the sterol synthetic pathway by dephosphorylation of farnesol pyrophosphate [38,46]. It is an acyclic sesquiterpene heat-stable molecule, which is produced primarily under aerobic conditions and it is unaffected by extreme pH and the type of carbon or nitrogen source [38,47]. Generally, the farnesol concentration is proportional to the colony-forming unit number [38]. Under physiological conditions, C. albicans isolates secrete a farnesol concentration with a mean of 35.6 μM (range: 13.7 to 58.5 μM) [48]. This concentration was 35 times higher than that secreted by non-albicans species, with the exception of Candida dubliniensis, which has demonstrated a concentration of 8.3 μM (range: 6.0 to 17.5 μM). All other non-albicans species excreted significantly lower farnesol concentrations, ranging from 0.4 to 1 μM [48]. These differences in excretion may be explained by the species-specific characteristics in sterol synthesis [49].
Based on a cDNA microarray analysis, a total of 274 genes were identified as responsive in C. albicans, with 104 genes up-regulated and 170 genes down-regulated [50]. Farnesol has an ability to influence Candida morphology, biofilm formation, drug efflux pump expression, apoptosis regulation, phagocytic response, surface hydrophobicity, iron metabolism, and heat-shock-related pathways [50,51,52,53,54]. One of the most prominent farnesol-associated effects is the induction of hypha-to-yeast transition and the inhibition of biofilm formation in various Candida species. It should be emphasized that 150-fold more farnesol is needed to block germ-tube formation in the presence of 10% serum, showing that it can bind to serum proteins at a high rate [55,56].
In view of this diverse role, it is not surprising that this compound influences several central signalling pathways in different Candida species. One of the best-studied farnesol-related pathways is the Ras1-cAMP-PKA cascade, where farnesol binds to the cyclase domain of the adenylyl cyclase Cyr1, influencing the level of intracellular cAMP [57]. Moreover, farnesol induces the cleavage of the small GTPase Ras1, resulting in a soluble Ras1; soluble Ras1 is a weak activator of Cyr1 and supports the formation of yeast cells [58]. Furthermore, farnesol can directly inhibit the cAMP signalling pathway, supporting the hypha-to-yeast transition [59]. It is noteworthy that farnesol exposure stabilizes the Nrg1 protein, which is the negative regulator of filamentation [60]. While farnesol was described first in C. albicans, it can inhibit filamentation and growth in other fungal species [27,61], including Saccharomyces cerevisiae [62], Aspergillus niger [63], Aspergillus flavus [64], Aspergillus nidulans [65], Penicillium expansum [66], Fusarium graminearum [67], and Paracoccidioides brasiliensis [68].
Regarding reactive oxygen species production, the supraphysiological farnesol concentrations (200–300 μM) are stressful for most fungi, while the physiological concentrations (30–40 μM) protect them from stress [57]. In addition to the farnesol-related effect on growth in the case of different microbes, the molecule also has a relevant immunomodulator effect [57,69]. Farnesol can stimulate both macrophage chemokine synthesis or macrophage recruitment, and trigger activation of neutrophil granulocytes and monocytes. Farnesol exposure also influences the differentiation of monocytes into dendritic cells [57,69].
Farnesol has been reported to induce cell growth inhibition and/or apoptosis in tumor cells where the observed IC50 values varied widely for different tumor types and different cell lines [70]. Farnesol caused 100% cell death at >120 μM in A549 and H460 lung cancer cells [71]. Scheper et al. (2008) observed an IC50 value of 30 to 60 μM for farnesol on the primary human tongue squamous cell carcinoma cell lines (OSCC9, OSCC 25) [70]. Nagy et al. (2020) evaluated 10 μM, 50 μM, 150 μM, and 300 μM farnesol concentrations in terms of toxicity to the Caco-2 cell line, where no toxicity was observed with any concentration tested [45].

4.2. Antimicrobial Activity of Farnesol

At physiological concentrations, farnesol has no significant effect on Candida cells that have already begun hyphae development or biofilm formation [25,38]. However, prior results suggest that farnesol can cause biofilm degradation at supraphysiological concentrations, suggesting the potential use of this compound in biofilm-associated infections [36]. In addition, several authors have published studies demonstrating contribution of farnesol to reduced azole resistance of Candida cells, including in biofilms [72]. This phenomenon can be explained by the modulation of Cdr1 efflux pumps, reactive oxygen species production, or changes in glutathione homeostasis [38,61,72]. Furthermore, farnesol has an effect on genes connected to ergosterol synthesis [46]. Dižová et al. (2018) observed that the presence of 200 μM farnesol down-regulated the ERG20, ERG11 and ERG9 genes. However, this farnesol concentration supplemented with 0.5 mg/L fluconazole restored the original expression level of ERG20 and ERG11. Interestingly, the physiological farnesol concentration (~30 μM) only slightly influences the expression of these genes in 48 h-old biofilms [73]. Chen et al. (2018) reported that CYR1 and PDE2 regulate resistance mechanisms against various antifungals in C. albicans biofilms. However, farnesol can diminish the resistance of C. albicans biofilms by regulating the expression of the gene CYR1 and PDE2 [74]. Yu et al. (2012) observed that the sterol biosynthetic pathway may contribute to the inhibitory effects of farnesol, as the transcription levels of the ERG11, ERG25, ERG6, ERG3, and ERG1 genes decreased following farnesol exposure [75]. Jabra-Rizk et al. (2006) showed that farnesol concentrations of 30–50 mM decrease the fluconazole MICs for C. albicans and C. dubliniensis from resistant values to a susceptible dose-dependent range, while concentrations of 100–300 mM resulted in fluconazole susceptibility [76].
One of the first major breakthroughs in combination-based experiments with farnesol and antifungals was published by Katragkou et al. (2015), who found a significant synergy against C. albicans 48 h-old biofilms between fluconazole, amphotericin B, and micafungin in the presence of farnesol [26]. The highest synergistic effect was observed in the case of micafungin combined with farnesol using fractional inhibitory concentration index determination and Bliss independence analysis. Based on the Bliss model, the observed effects were 39–52% higher compared to the expected efficacy if the drugs had been acting independently [26]. It should be noted that synergism was observed only in the case of farnesol/micafungin and farnesol/fluconazole based on calculated fractional inhibitory concentration indices, suggesting the usage of multiple analytical approaches for investigation of drug-drug interaction [26].
Regarding non-albicans species, Kovács et al. (2016) showed that farnesol consistently enhanced the activity of caspofungin and micafungin, as concordantly shown in two independent experimental settings (chequerboard dilution and time–kill experiments) [27]. Fernández-Rivero et al. (2017) reported that a supraphysiological farnesol concentration (300 μM) improved the activity of amphotericin B against Candida tropicalis biofilms but did not affect anidulafungin [77]. Two recent studies by Nagy et al. concluded that farnesol significantly enhanced the activity of echinocandins and triazoles against one-day-old C. auris biofilms in vitro, suggesting an alternative approach to overcome the previously well-documented azole and echinocandin resistance of C. auris biofilms [45,78].
Animal experiments with farnesol raised several questions in terms of in vivo applicability of this compound. In one of the first in vivo studies, Navarathna et al. (2007) concluded that the physiological farnesol production may play a pivotal role as a virulence factor in fungal pathogenesis; furthermore, exogenous oral and intraperitoneal farnesol administration (20 mM) enhances the mortality of mice in their systemic mouse model [79]. Contrary to these results, Hisajima et al. (2008) observed a protective effect against C. albicans in their oral candidiasis mouse model [80]. It should be noted that there was a 1000-fold difference between the administered farnesol dosages (9 μM/mouse) in the experiments of Hisajima et al. (2008) [80] compared to experiments performed by Navarathna et al. (2007) (20 mM/mouse) [79]. In addition, they reported a potential gastrointestinal tract-related farnesol effect including moderate bodyweight reduction and reduced Candida faeces burden [80]. A cocktail of Candida-derived regulatory alcohols combined with nanomolar amounts of farnesol was reported to have a similar protective effect by Martins et al. (2012) in their murine model of disseminated candidiasis [81]. Bozó et al. (2016) did not find a farnesol-related protective effect against vaginal C. albicans infection [82], in contrast to the findings of Hisajima et al. (2008) [80]. However, both administered farnesol regimens enhanced the activity of 5 mg/kg daily fluconazole treatment against fluconazole-resistant C. albicans strain [82]. Similar fluconazole resistance reversion was observed in the case of planktonic cells by Jabra-Rizk et al. (2006) [76] and Cordeiro et al. (2013) [83]. Fernandes Costa et al. (2019) used nanoparticles containing farnesol alone or in combination with miconazole; nanoparticles containing farnesol inhibited yeast-to-hyphae transition at concentrations greater than or equal to 240 μM [84]. In addition, chitosan nanoparticles containing miconazole (33 mg/L) and farnesol (2.1 mM) inhibited fungal proliferation and decreased the pathogenicity of mouse vulvovaginitis infection [84]. Nagy et al. (2020) demonstrated that a daily treatment with 75 μM farnesol decreased the C. auris kidney burden in their immunocompromised systemic mouse model, especially when inocula was pre-exposed to farnesol [45].
The farnesol-exerted antifungal activity can be explained by the higher level of reactive oxygen species, especially in the case of non-albicans species [43,45]. Furthermore, farnesol has an amphiphilic property which allows for its integration into cell membranes, influencing membrane fluidity and integrity. In the case of Candida parapsilosis and C. dubliniensis, farnesol affected the cellular polarization and membrane permeability [61,76,85]. These observations can help further elucidate the antifungal effect.
Farnesol has a remarkable antibacterial effect alone or in combination with traditional antibacterial agents as demonstrated by in vitro investigations. Jabra-Rizk et al. (2006) observed that farnesol treatment (100 μM) increases the activity of gentamicin against Staphylococcus aureus biofilms [86]. Gomes et al. (2009) showed that farnesol exposure (300 μM) produced a relatively long post-antimicrobial effect (>8 h) against Staphylococcus epidermidis [87], while Pammi et al. (2011) observed that farnesol exposure at a concentration of 500 μM significantly inhibited the S. epidermidis biofilm formation in vitro [88]. A clear synergistic interaction was observed between farnesol and nafcillin or vancomycin against S. epidermidis sessile cells [88]. Additionally, it potentiates the activity of beta-lactam antibiotics against antibiotic-resistant bacterium species [89]. Castelo-Branco et al. (2012) showed a potent antimicrobial effect exerted by exogenous farnesol exposure against mature Burkholderia pseudomallei biofilms [90]. Additionally, it increased the activity of amoxicillin, ceftazidime, doxycycline, and sulfamethoxazole-trimethoprim, which are routinely administered for the treatment of melioidoses [91]. Farnesol also had a synergizing effect against ciprofloxacin-resistant Pseudomonas aeruginosa biofilms when used in combination with ciprofloxacin [92]. In vivo data also supports the antibacterial efficacy of farnesol. It has been observed that 6.7 mM farnesol treatment significantly decreased the S. epidermidis associated catheter infection and systemic dissemination [88].
Based on several studies, farnesol has a remarkable effect in Candida-bacterium mixed biofilms. C. albicans-derived farnesol has also been shown to have an effect on the response of S. aureus to antibiotics in mixed species biofilms. Farnesol exposure results in a significant decrease in staphyloxantin, which is an important virulence factor of this bacterium [42]. Černáková et al. (2018) showed that 200 μM farnesol has an inhibitory effect on C. albicans growth in mixed-species biofilms with Streptococcus mutans [93]. Cugini et al. (2010) examined the C. albicans-P. aeruginosa mixed species biofilms, where the C. albicans-derived farnesol enhanced P. aeruginosa quinolone signal production in a LasR-defective strain [94].

5. Tyrosol

5.1. Physiological Effect of Tyrosol in Candida Species

Tyrosol (2-(4-hydoxyphenyl)-ethanol) is a tyrosine-derived molecule which is synthetized via either tyramine or 4-hydroxyphenylacetaldehyde [95,96]. In the case of C. albicans, it is released into the growth medium continuously during the exponential growth phase and is capable of decreasing the duration of the lag phase before cells begin germination. The accumulation of tyrosol in the culture medium is proportional to the rise of fungal cell number. While the molecule stimulates filamentation, it exclusively promotes germ tube formation in conditions that normally induce these physiological processes [95,96]. Tyrosol exposure influences cell cycle regulation, DNA replication, and chromosome segregation in C. albicans [95]. Additionally, it was shown to have an inhibitory effect on neutrophil granulocytes by interfering with the oxidative stress response of these phagocytes [97,98]. Significantly more tyrosol was secreted by C. albicans (range: 21.01 ± 0.76 to 53.40 ± 1.73 μM/1.6 × 107–5.3 × 107 cells/mL) and C. tropicalis (range: 41.21 ± 1.21 to 48.63 ± 3.83 μM/2.6 × 107–2.7 × 107 cells/mL) than by Candida glabrata (range: 1.3 ± 0.17 to 3.26±0.33 μM/2.7×107–5.5×107 cells/mL) or C. parapsilosis (range: 1.59 ± 0.29 to 3.04 ± 0.43 μM/1.7 × 107–2.3 × 107 cells/mL), suggesting a possible link with virulence [99]. Tyrosol plays a pivotal role in biofilm production, where it can stimulate hypha production of C. albicans, especially between two and six hours of biofilm development. C. albicans biofilms released at least 50% more tyrosol when compared to planktonic cells [96].
Regarding non-albicans species, tyrosol has been recognized as inducing the biofilm-forming ability of C. auris to grow as yeast or pseudohyphae [96]. Based on RNA-Seq analysis, tyrosol treatment resulted in 261 and 181 differentially expressed genes with at least a 1.5-fold increase or decrease in expression in C. parapsilosis, respectively; however, the initial adherence was not affected by the presence of tyrosol [43]. Interestingly, the ortholog of the C. albicans CZF1 gene, which is a key transcription factor of biofilm development in C. parapsilosis, was upregulated following tyrosol exposure [43,100]. Nevertheless, Jakab et al. (2019) did not observe higher rates of biofilm formation in the presence of tyrosol [43]. In C. parapsilosis, tyrosol exposure overexpressed the active efflux pumps and caused an enhanced oxidative stress response, while inhibiting growth, ribosome biogenesis, and virulence. Surprisingly, its metabolism was modulated toward glycolysis and ethanol fermentation [43]. Monteiro et al. (2015) reported that tyrosol exposure did not induce increased adhesion in C. glabrata [101].
Regarding tyrosol related toxic effect, initial cytotoxicity was observed at concentrations of >10 mM, 3 mM, 5 mM and >15 mM for human gingival fibroblasts (GN61), human gingival epithelial cells (S-G), human salivary gland carcinoma cells (HSG1) and colon adenocarcinomas cell line (Caco-2), respectively [43,102].

5.2. Antimicrobial Activity of Tyrosol

Tyrosol is a relatively understudied molecule compared to farnesol in terms of potential antifungal or anti-biofilm activity; despite this, a few studies have examined the potential use of tyrosol in monotherapy or in combination with traditional antifungal agents against Candida species [36,72]. Arias et al. (2016) showed that tyrosol treatment at concentrations ranging from 100 to 200 mM exerted a significant reduction in metabolic activity against C. albicans and C. glabrata two-day-old oral biofilms, which was proportional to a reduction in cell number [103]. Do Vale et al. (2017) showed that tyrosol alone at concentrations of 50 and 90 mM demonstrated inhibition of the planktonic growth of C. albicans and C. glabrata cells, respectively [104]. However, tyrosol does not significantly reduce metabolic activity or the number of cells for one-day-old oral biofilms; in addition, the nature of interaction of tyrosol with chlorohexidine gluconate was indifferent. Nevertheless, 1.25 mM tyrosol with 0.00725 mM chlorhexidine gluconate showed a synergistic interaction in reducing the number of hyphae formed [104]. A combination of tyrosol and farnesol has been explored for oral Candida isolates for both planktonic and sessile growth. This combination showed synergy against C. glabrata, indicating that this combination may contribute to the development of oral care products against Candida species [105].
In another study, tyrosol showed anti-biofilm activity against denture-derived C. albicans isolates. However, it has been shown that the single use of tyrosol cannot decrease hydrolytic enzymes on oral C. albicans [106]. Shanmughapriya et al. (2014) observed that tyrosol treatment caused a 25% and a 50% reduction in intrauterine device-derived Candida krusei and C. tropicalis biofilm production at concentrations of 40 μM and 80 μM, respectively [107]. In addition, amphotericin B combined with tyrosol showed a remarkable inhibitory effect against these non-albicans biofilms. A concentration of 4 mg/L amphotericin B in the presence of 80 μM tyrosol exerted approximately 90% inhibition in biofilm formation [107]. Cordeiro et al. (2015) showed that the addition of tyrosol significantly reduced the MICs for amphotericin B, fluconazole, and itraconazole against planktonic C. albicans and C. tropicalis [108]. Furthermore, exogenous tyrosol alone was able to significantly reduce the biofilm formation of these species at concentrations ranging from 125 to 250 mM. At these concentrations, tyrosol decreased the metabolic activity of growing biofilms by approximately 24 and 30% for C. albicans and C. tropicalis, respectively. Reduction of metabolic activity was more pronounced when tyrosol was combined with traditional antifungal drugs including amphotericin B, fluconazole, and itraconazole. It should be noted that application of amphotericin B with tyrosol markedly decreased the metabolic activity of mature biofilms (35%) [108]. Kovács et al. (2017) reported that tyrosol may be used as an adjuvant agent with caspofungin or micafungin in alternative treatment strategies [109]. Regarding the in vivo antifungal effect of tyrosol, Jakab et al. (2019) reported that daily treatment with 15 mM tyrosol decreased the fungal tissue burden in their immunocompromised mouse model [43]. In this study, the expression of ALS6, which has a pivotal role in adhesion, was significantly reduced by tyrosol treatment. Furthermore, downregulation of the expression of FAD2 and FAD3 may also contribute to decreased virulence and kidney fungal burden. The well-documented antifungal effects exerted by tyrosol may be explained by the enhanced oxidative stress and the inhibition of virulence-related genes, growth, and ribosome biogenesis. In addition, tyrosol can alter the metabolism of Candida cells toward fermentation [43].
Data on the potential antibacterial effects of tyrosol remain scarce. Arias et al. (2016) found a potential anti-biofilm activity of tyrosol against S. mutans in single and mixed species biofilms with C. albicans or C. glabrata developed on acrylic resin and hydroxyapatite surfaces [103]. Their results may contribute to the development of innovative topical therapies focusing on biofilm-associated oral diseases. Abdel-Rhman et al. (2016) reported substantial antibacterial activity of tyrosol against S. aureus; moreover, tyrosol increased susceptibility to gentamicin, amikacin, and ciprofloxacin at subinhibitory concentrations ranging from 3.5 to 14.3 mM [110]. Tyrosol treatment can also influence S. aureus virulence, decreasing the production of protease and lipase enzymes and limiting the ability to form biofilms [110]. In the case of P. aeruginosa, tyrosol strongly inhibited haemolysin and protease production [111].

6. Future Remarks

Paradoxically, medical advancement has resulted in an increasing number of immunocompromised individuals susceptible to Candida infections. The incidence and mortality rate related to systemic Candida infections has remained unchanged, despite the advances in the field of antifungal therapy. Based on recent comprehensive epidemiological studies, the high incidence and mortality may be attributed to sessile Candida populations, namely biofilms, which show high resistance against environmental factors, immune responses, and traditional antifungal therapy. Although there is no definitive solution or highly effective therapeutic recommendation against Candida biofilms, there are many promising therapeutic strategies including antifungal “lock” therapy, photodynamic inactivation, and the use of natural products or synthetic peptides with antifungal activity. A further solution may be the utilization of quorum-sensing molecules alone or in combination with traditional antifungal agents; however, there are numerous open questions as to their exact action or the interaction between quorum-sensing molecules and the host. In addition, the full understanding of quorum sensing in non-albicans species has remained unelucidated. In this review, we provided an overview on the current status of studies focusing on anti-biofilm activity of farnesol and tyrosol. Hopefully, these in vitro and in vivo results can be implemented in therapeutic practice as soon as possible to overcome Candida biofilm-related infections.

Author Contributions

Conceptualization, methodology and writing were performed by R.K. and L.M. All authors have read and agreed to the published version of the manuscript.

Funding

R.K. was supported by the EFOP-3.6.3-VEKOP-16-2017-00009 program and by a FEMS Research and Training Grant (FEMS-GO-2019-502).

Conflicts of Interest

L.M. received conference travel grants from Cidara, MSD, Astellas and Pfizer. All other authors report no conflicts of interest.

References

  1. Hawksworth, D.L.; Lücking, R. Fungal Diversity Revisited: 2.2 to 3.8 Million Species. Microbiol. Spectr. 2017, 5. [Google Scholar] [CrossRef]
  2. Magill, S.S.; O’Leary, E.; Janelle, S.J.; Thompson, D.L.; Dumyati, G.; Nadle, J.; Wilson, L.E.; Kainer, M.A.; Lynfield, R.; Greissman, S.; et al. Changes in Prevalence of Health Care-Associated Infections in U.S. Hospitals. N. Engl. J. Med. 2018, 379, 1732–1744. [Google Scholar] [CrossRef] [PubMed]
  3. Quindós, G. Epidemiology of candidaemia and invasive candidiasis. A changing face. Rev. Iberoam. Micol. 2014, 31, 42–48. [Google Scholar] [CrossRef] [PubMed]
  4. Wiederhold, N.P. Antifungal resistance: Current trends and future strategies to combat. Infect. Drug Resist. 2017, 10, 249–259. [Google Scholar] [CrossRef] [Green Version]
  5. Kontoyiannis, D.P. Antifungal Resistance: An Emerging Reality and A Global Challenge. J. Infect. Dis. 2017, 216 (Suppl. 3), S431–S435. [Google Scholar] [CrossRef] [Green Version]
  6. Casadevall, A.; Kontoyiannis, D.P.; Robert, V. On the Emergence of Candida auris: Climate Change, Azoles, Swamps, and Birds. MBio 2019, 10, e01397-19. [Google Scholar] [CrossRef] [Green Version]
  7. Friedman, D.Z.P.; Schwartz, I.S. Emerging Fungal Infections: New Patients, New Patterns, and New Pathogens. J. Fungi 2019, 5, 67. [Google Scholar] [CrossRef] [Green Version]
  8. Silva, S.; Rodrigues, C.F.; Araújo, D.; Rodrigues, M.E.; Henriques, M. Candida Species Biofilms’ Antifungal Resistance. J. Fungi 2017, 3, 8. [Google Scholar] [CrossRef] [Green Version]
  9. De Barros, P.P.; Rossoni, R.D.; De Souza, C.M.; Scorzoni, L.; Fenley, J.C.; Junqueira, J.C. Candida Biofilms: An Update on Developmental Mechanisms and Therapeutic Challenges [published online ahead of print, 2020 Apr 10]. Mycopathologia 2020. [Google Scholar] [CrossRef]
  10. Cavalheiro, M.; Teixeira, M.C. Candida Biofilms: Threats, Challenges, and Promising Strategies. Front. Med. 2018, 5, 28. [Google Scholar] [CrossRef] [Green Version]
  11. Nett, J.E. Future directions for anti-biofilm therapeutics targeting Candida. Expert Rev. Anti Infect. Ther. 2014, 12, 375–382. [Google Scholar] [CrossRef] [PubMed]
  12. Davis, M.R.; Donnelley, M.A.; Thompson, G.R. Ibrexafungerp: A novel oral glucan synthase inhibitor [published online ahead of print, 2019 Jul 25]. Med. Mycol. 2019, myz083. [Google Scholar] [CrossRef]
  13. Pfaller, M.A.; Carvalhaes, C.; Messer, S.A.; Rhomberg, P.R.; Castanheira, M. Activity of a Long-Acting Echinocandin, Rezafungin, and Comparator Antifungal Agents Tested against Contemporary Invasive Fungal Isolates (SENTRY Program, 2016 to 2018). Antimicrob. Agents Chemother. 2020, 64, e00099-20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Nield, B.; Larsen, S.R.; Van Hal, S.J. Clinical experience with new formulation SUBA®-itraconazole for prophylaxis in patients undergoing stem cell transplantation or treatment for haematological malignancies. J. Antimicrob. Chemother. 2019, 74, 3049–3055. [Google Scholar] [CrossRef]
  15. Warrilow, A.G.; Hull, C.M.; Parker, J.E.; Garvey, E.P.; Hoekstra, W.J.; Moore, W.R.; Schotzinger, R.J.; Kelly, D.E.; Kelly, S.L. The clinical candidate VT-1161 is a highly potent inhibitor of Candida albicans CYP51 but fails to bind the human enzyme. Antimicrob. Agents Chemother. 2014, 58, 7121–7127. [Google Scholar] [CrossRef] [Green Version]
  16. Samaranayake, Y.H.; Ye, J.; Yau, J.Y.; Cheung, B.P.; Samaranayake, L.P. In vitro method to study antifungal perfusion in Candida biofilms. J. Clin. Microbiol. 2005, 43, 818–825. [Google Scholar] [CrossRef] [Green Version]
  17. Kuhn, D.M.; George, T.; Chandra, J.; Mukherjee, P.K.; Ghannoum, M.A. Antifungal susceptibility of Candida biofilms: Unique efficacy of amphotericin B lipid formulations and echinocandins. Antimicrob. Agents Chemother. 2002, 46, 1773–1780. [Google Scholar] [CrossRef] [Green Version]
  18. Pappas, P.G.; Kauffman, C.A.; Andes, D.R.; Clancy, C.J.; Marr, K.A.; Ostrosky-Zeichner, L.; Reboli, A.C.; Schuster, M.G.; Vazquez, J.A.; Walsh, T.J.; et al. Executive Summary: Clinical Practice Guideline for the Management of Candidiasis: 2016 Update by the Infectious Diseases Society of America. Clin. Infect. Dis. 2016, 62, 409–417. [Google Scholar] [CrossRef] [Green Version]
  19. Walraven, C.J.; Lee, S.A. Antifungal lock therapy. Antimicrob. Agents Chemother. 2013, 57, 1–8. [Google Scholar] [CrossRef] [Green Version]
  20. Kovács, R.; Nagy, F.; Tóth, Z.; Bozó, A.; Balázs, B.; Majoros, L. Synergistic effect of nikkomycin Z with caspofungin and micafungin against Candida albicans and Candida parapsilosis biofilms. Lett. Appl. Microbiol. 2019, 69, 271–278. [Google Scholar] [CrossRef] [Green Version]
  21. Wall, G.; Chaturvedi, A.K.; Wormley, F.L., Jr.; Wiederhold, N.P.; Patterson, H.P.; Patterson, T.F.; Lopez-Ribot, J.L. Screening a Repurposing Library for Inhibitors of Multidrug-Resistant Candida auris Identifies Ebselen as a Repositionable Candidate for Antifungal Drug Development. Antimicrob. Agents Chemother. 2018, 62, e01084-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Pedroso, R.; Balbino, B.L.; Andrade, G.; Dias, M.; Alvarenga, T.A.; Pedroso, R.; Pimenta, L.P.; Lucarini, R.; Pauletti, P.M.; Januário, A.H.; et al. In Vitro and In Vivo Anti-Candida spp. Activity of Plant-Derived Products. Plants 2019, 8, 494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Souza, C.M.; Pereira Junior, S.A.; Moraes, T.; Damasceno, J.L.; Amorim Mendes, S.; Dias, H.J.; Stefani, R.; Tavares, D.C.; Martins, C.H.; Crotti, A.E.; et al. Antifungal activity of plant-derived essential oils on Candida tropicalis planktonic and biofilms cells. Med. Mycol. 2016, 54, 515–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Sherry, L.; Jose, A.; Murray, C.; Williams, C.; Jones, B.; Millington, O.; Bagg, J.; Ramage, G. Carbohydrate Derived Fulvic Acid: An in vitro Investigation of a Novel Membrane Active Antiseptic Agent against Candida albicans Biofilms. Front. Microbiol. 2012, 3, 116. [Google Scholar] [CrossRef] [Green Version]
  25. Ramage, G.; Saville, S.P.; Wickes, B.L.; López-Ribot, J.L. Inhibition of Candida albicans biofilm formation by farnesol, a quorum-sensing molecule. Appl. Environ. Microbiol. 2002, 68, 5459–5463. [Google Scholar] [CrossRef] [Green Version]
  26. Katragkou, A.; McCarthy, M.; Alexander, E.L.; Antachopoulos, C.; Meletiadis, J.; Jabra-Rizk, M.A.; Petraitis, V.; Roilides, E.; Walsh, T.J. In vitro interactions between farnesol and fluconazole, amphotericin B or micafungin against Candida albicans biofilms. J. Antimicrob. Chemother. 2015, 70, 470–478. [Google Scholar] [CrossRef]
  27. Kovács, R.; Bozó, A.; Gesztelyi, R.; Domán, M.; Kardos, G.; Nagy, F.; Tóth, Z.; Majoros, L. Effect of caspofungin and micafungin in combination with farnesol against Candida parapsilosis biofilms. Int. J. Antimicrob. Agents. 2016, 47, 304–310. [Google Scholar] [CrossRef] [Green Version]
  28. Weiland-Bräuer, N.; Malek, I.; Schmitz, R.A. Metagenomic quorum quenching enzymes affect biofilm formation of Candida albicans and Staphylococcus epidermidis. PLoS ONE 2019, 14, e0211366. [Google Scholar] [CrossRef]
  29. Nobile, C.J.; Johnson, A.D. Candida albicans Biofilms and Human Disease. Annu. Rev. Microbiol. 2015, 69, 71–92. [Google Scholar] [CrossRef] [Green Version]
  30. Harriott, M.M.; Lilly, E.A.; Rodriguez, T.E.; Fidel, P.L.; Noverr, M.C. Candida albicans forms biofilms on the vaginal mucosa. Microbiology. 2010, 156 Pt 12, 3635–3644. [Google Scholar] [CrossRef] [Green Version]
  31. Ganguly, S.; Mitchell, A.P. Mucosal biofilms of Candida albicans. Curr. Opin. Microbiol. 2011, 14, 380–385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kalan, L.; Loesche, M.; Hodkinson, B.P.; Heilmann, K.; Ruthel, G.; Gardner, S.E.; Grice, E.A. Redefining the Chronic-Wound Microbiome: Fungal Communities Are Prevalent, Dynamic, and Associated with Delayed Healing. MBio 2016, 7, e01058-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Ramage, G.; VandeWalle, K.; López-Ribot, J.L.; Wickes, B.L. The filamentation pathway controlled by the Efg1 regulator protein is required for normal biofilm formation and development in Candida albicans. FEMS Microbiol. Lett. 2002, 214, 95–100. [Google Scholar] [CrossRef] [Green Version]
  34. Lewis, K. Persister cells, dormancy and infectious disease. Nat. Rev. Microbiol. 2007, 5, 48–56. [Google Scholar] [CrossRef] [PubMed]
  35. Ramage, G.; Bachmann, S.; Patterson, T.F.; Wickes, B.L.; López-Ribot, J.L. Investigation of multidrug efflux pumps in relation to fluconazole resistance in Candida albicans biofilms. J. Antimicrob. Chemother. 2002, 49, 973–980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Mehmood, A.; Liu, G.; Wang, X.; Meng, G.; Wang, C.; Liu, Y. Fungal Quorum-Sensing Molecules and Inhibitors with Potential Antifungal Activity: A Review. Molecules 2019, 24, 1950. [Google Scholar] [CrossRef] [Green Version]
  37. Albuquerque, P.; Casadevall, A. Quorum sensing in fungi—A review. Med. Mycol. 2012, 50, 337–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Hornby, J.M.; Jensen, E.C.; Lisec, A.D.; Tasto, J.J.; Jahnke, B.; Shoemaker, R.; Dussault, P.; Nickerson, K.W. Quorum sensing in the dimorphic fungus Candida albicans is mediated by farnesol. Appl. Environ. Microbiol. 2001, 67, 2982–2992. [Google Scholar] [CrossRef] [Green Version]
  39. Sorrentino, F.; Roy, I.; Keshavarz, T. Impact of linoleic acid supplementation on lovastatin production in Aspergillus terreus cultures. Appl. Microbiol. Biotechnol. 2010, 88, 65–73. [Google Scholar] [CrossRef]
  40. Raina, S.; Odell, M.; Keshavarz, T. Quorum sensing as a method for improving sclerotiorin production in Penicillium sclerotiorum. J. Biotechnol. 2010, 148, 91–98. [Google Scholar] [CrossRef]
  41. Wongsuk, T.; Pumeesat, P.; Luplertlop, N. Fungal quorum sensing molecules: Role in fungal morphogenesis and pathogenicity. J. Basic Microbiol. 2016, 56, 440–447. [Google Scholar] [CrossRef] [PubMed]
  42. Vila, T.; Kong, E.F.; Ibrahim, A.; Piepenbrink, K.; Shetty, A.C.; McCracken, C.; Bruno, V.; Jabra-Rizk, M.A. Candida albicans quorum-sensing molecule farnesol modulates staphyloxanthin production and activates the thiol-based oxidative-stress response in Staphylococcus aureus. Virulence 2019, 10, 625–642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Jakab, Á.; Tóth, Z.; Nagy, F.; Nemes, D.; Bácskay, I.; Kardos, G.; Emri, T.; Pócsi, I.; Majoros, L.; Kovács, R. Physiological and Transcriptional Responses of Candida parapsilosis to Exogenous Tyrosol. Appl. Environ. Microbiol. 2019, 85, e01388-19. [Google Scholar] [CrossRef] [Green Version]
  44. Shirtliff, M.E.; Krom, B.P.; Meijering, R.A.; Peters, B.M.; Zhu, J.; Scheper, M.A.; Harris, M.L.; Jabra-Rizk, M.A. Farnesol-induced apoptosis in Candida albicans. Antimicrob. Agents Chemother. 2009, 53, 2392–2401. [Google Scholar] [CrossRef] [Green Version]
  45. Nagy, F.; Vitális, E.; Jakab, Á.; Borman, A.M.; Forgács, L.; Tóth, Z.; Majoros, L.; Kovács, R. In vitro and in vivo Effect of Exogenous Farnesol Exposure against Candida auris. Front. Microbiol. 2020, 11, 957. [Google Scholar] [CrossRef] [PubMed]
  46. Hornby, J.M.; Kebaara, B.W.; Nickerson, K.W. Farnesol biosynthesis in Candida albicans: Cellular response to sterol inhibition by zaragozic acid B. Antimicrob. Agents Chemother. 2003, 47, 2366–2369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Westwater, C.; Balish, E.; Schofield, D.A. Candida albicans-conditioned medium protects yeast cells from oxidative stress: A possible link between quorum sensing and oxidative stress resistance. Eukaryot Cell 2005, 4, 1654–1661. [Google Scholar] [CrossRef] [Green Version]
  48. Weber, K.; Sohr, R.; Schulz, B.; Fleischhacker, M.; Ruhnke, M. Secretion of E,E-farnesol and biofilm formation in eight different Candida species [published correction appears in Antimicrob Agents Chemother. 2009 Feb;53(2):848]. Antimicrob. Agents Chemother. 2008, 52, 1859–1861. [Google Scholar] [CrossRef] [Green Version]
  49. Nickerson, K.W.; Atkin, A.L.; Hornby, J.M. Quorum sensing in dimorphic fungi: Farnesol and beyond. Appl Environ Microbiol. 2006, 72, 3805–3813. [Google Scholar] [CrossRef] [Green Version]
  50. Cao, Y.Y.; Cao, Y.B.; Xu, Z.; Ying, K.; Li, Y.; Xie, Y.; Zhu, Z.Y.; Chen, W.S.; Jiang, Y.Y. cDNA microarray analysis of differential gene expression in Candida albicans biofilm exposed to farnesol. Antimicrob. Agents Chemother. 2005, 49, 584–589. [Google Scholar] [CrossRef] [Green Version]
  51. Sharma, M.; Prasad, R. The quorum-sensing molecule farnesol is a modulator of drug efflux mediated by ABC multidrug transporters and synergizes with drugs in Candida albicans. Antimicrob. Agents Chemother. 2011, 55, 4834–4843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Léger, T.; Garcia, C.; Ounissi, M.; Lelandais, G.; Camadro, J.M. The metacaspase (Mca1p) has a dual role in farnesol-induced apoptosis in Candida albicans. Mol. Cell Proteom. 2015, 14, 93–108. [Google Scholar] [CrossRef] [Green Version]
  53. Enjalbert, B.; Whiteway, M. Release from quorum-sensing molecules triggers hyphal formation during Candida albicans resumption of growth. Eukaryot Cell 2005, 4, 1203–1210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Uppuluri, P.; Mekala, S.; Chaffin, W.L. Farnesol-mediated inhibition of Candida albicans yeast growth and rescue by a diacylglycerol analogue. Yeast 2007, 24, 681–693. [Google Scholar] [CrossRef] [PubMed]
  55. Weber, K.; Schulz, B.; Ruhnke, M. The quorum-sensing molecule E,E-farnesol—Its variable secretion and its impact on the growth and metabolism of Candida species. Yeast 2010, 27, 727–739. [Google Scholar] [CrossRef] [Green Version]
  56. Langford, M.L.; Hasim, S.; Nickerson, K.W.; Atkin, A.L. Activity and toxicity of farnesol towards Candida albicans are dependent on growth conditions. Antimicrob. Agents Chemother. 2010, 54, 940–942. [Google Scholar] [CrossRef] [Green Version]
  57. Polke, M.; Leonhardt, I.; Kurzai, O.; Jacobsen, I.D. Farnesol signalling in Candida albicans-more than just communication. Crit. Rev. Microbiol. 2018, 44, 230–243. [Google Scholar] [CrossRef]
  58. Piispanen, A.E.; Grahl, N.; Hollomon, J.M.; Hogan, D.A. Regulated proteolysis of Candida albicans Ras1 is involved in morphogenesis and quorum sensing regulation. Mol. Microbiol. 2013, 89, 166–178. [Google Scholar] [CrossRef] [Green Version]
  59. Lindsay, A.K.; Deveau, A.; Piispanen, A.E.; Hogan, D.A. Farnesol and cyclic AMP signaling effects on the hypha-to-yeast transition in Candida albicans. Eukaryot Cell 2012, 11, 1219–1225. [Google Scholar] [CrossRef] [Green Version]
  60. Lu, Y.; Su, C.; Unoje, O.; Liu, H. Quorum sensing controls hyphal initiation in Candida albicans through Ubr1-mediated protein degradation. Proc. Natl. Acad. Sci. USA 2014, 111, 1975–1980. [Google Scholar] [CrossRef] [Green Version]
  61. Rossignol, T.; Logue, M.E.; Reynolds, K.; Grenon, M.; Lowndes, N.F.; Butler, G. Transcriptional response of Candida parapsilosis following exposure to farnesol [published correction appears in Antimicrob Agents Chemother. 2008 Jun;52(6):2296]. Antimicrob. Agents Chemother. 2007, 51, 2304–2312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Fairn, G.D.; Macdonald, K.; McMaster, C.R. A chemogenomic screen in Saccharomyces cerevisiae uncovers a primary role for the mitochondria in farnesol toxicity and its regulation by the Pkc1 pathway. J. Biol. Chem. 2007, 282, 4868–4874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Lorek, J.; Pöggeler, S.; Weide, M.R.; Breves, R.; Bockmühl, D.P. Influence of farnesol on the morphogenesis of Aspergillus niger. J. Basic Microbiol. 2008, 48, 99–103. [Google Scholar] [CrossRef] [PubMed]
  64. Wang, X.; Wang, Y.; Zhou, Y.; Wei, X. Farnesol induces apoptosis-like cell death in the pathogenic fungus Aspergillus flavus. Mycologia 2014, 106, 881–888. [Google Scholar] [CrossRef] [PubMed]
  65. Semighini, C.P.; Hornby, J.M.; Dumitru, R.; Nickerson, K.W.; Harris, S.D. Farnesol-induced apoptosis in Aspergillus nidulans reveals a possible mechanism for antagonistic interactions between fungi. Mol. Microbiol. 2006, 59, 753–764. [Google Scholar] [CrossRef] [Green Version]
  66. Liu, P.; Luo, L.; Guo, J.; Liu, H.; Wang, B.; Deng, B.; Long, C.A.; Cheng, Y. Farnesol induces apoptosis and oxidative stress in the fungal pathogen Penicillium expansum. Mycologia 2010, 102, 311–318. [Google Scholar] [CrossRef]
  67. Semighini, C.P.; Murray, N.; Harris, S.D. Inhibition of Fusarium graminearum growth and development by farnesol. FEMS Microbiol. Lett. 2008, 279, 259–264. [Google Scholar] [CrossRef] [Green Version]
  68. Derengowski, L.S.; De-Souza-Silva, C.; Braz, S.V.; Mello-De-Sousa, T.M.; Báo, S.N.; Kyaw, C.M.; Silva-Pereira, I. Antimicrobial effect of farnesol, a Candida albicans quorum sensing molecule, on Paracoccidioides brasiliensis growth and morphogenesis. Ann. Clin. Microbiol. Antimicrob. 2009, 8, 13. [Google Scholar] [CrossRef] [Green Version]
  69. Hargarten, J.C.; Moore, T.C.; Petro, T.M.; Nickerson, K.W.; Atkin, A.L. Candida albicans Quorum Sensing Molecules Stimulate Mouse Macrophage Migration. Infect. Immun. 2015, 83, 3857–3864. [Google Scholar] [CrossRef] [Green Version]
  70. Scheper, M.A.; Shirtliff, M.E.; Meiller, T.F.; Peters, B.M.; Jabra-Rizk, M.A. Farnesol, a fungal quorum-sensing molecule triggers apoptosis in human oral squamous carcinoma cells. Neoplasia 2008, 10, 954–963. [Google Scholar] [CrossRef] [Green Version]
  71. Wang, Z.; Chen, H.T.; Roa, W.; Finlay, W. Farnesol for aerosol inhalation: Nebulization and activity against human lung cancer cells. J. Pharm. Pharm. Sci. 2003, 6, 95–100. [Google Scholar] [PubMed]
  72. Rodrigues, C.F.; Černáková, L. Farnesol and Tyrosol: Secondary Metabolites with a Crucial quorum-sensing Role in Candida Biofilm Development. Genes 2020, 11, 444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Dižová, S.; Černáková, L.; Bujdáková, H. The impact of farnesol in combination with fluconazole on Candida albicans biofilm: Regulation of ERG20, ERG9, and ERG11 genes. Folia Microbiol. 2018, 63, 363–371. [Google Scholar] [CrossRef] [PubMed]
  74. Chen, S.; Xia, J.; Li, C.; Zuo, L.; Wei, X. The possible molecular mechanisms of farnesol on the antifungal resistance of C. albicans biofilms: The regulation of CYR1 and PDE2. BMC Microbiol. 2018, 18, 203. [Google Scholar] [CrossRef] [Green Version]
  75. Yu, L.H.; Wei, X.; Ma, M.; Chen, X.J.; Xu, S.B. Possible inhibitory molecular mechanism of farnesol on the development of fluconazole resistance in Candida albicans biofilm. Antimicrob. Agents Chemother. 2012, 56, 770–775. [Google Scholar] [CrossRef] [Green Version]
  76. Jabra-Rizk, M.A.; Shirtliff, M.; James, C.; Meiller, T. Effect of farnesol on Candida dubliniensis biofilm formation and fluconazole resistance. FEMS Yeast Res. 2006, 6, 1063–1073. [Google Scholar] [CrossRef] [Green Version]
  77. Fernández-Rivero, M.E.; Del Pozo, J.L.; Valentín, A.; De Diego, A.M.; Pemán, J.; Cantón, E. Activity of Amphotericin B and Anidulafungin Combined with Rifampicin, Clarithromycin, Ethylenediaminetetraacetic Acid, N-Acetylcysteine, and Farnesol against Candida tropicalis Biofilms. J. Fungi 2017, 3, 16. [Google Scholar] [CrossRef] [Green Version]
  78. Nagy, F.; Tóth, Z.; Daróczi, L.; Székely, A.; Borman, A.M.; Majoros, L.; Kovács, R. Farnesol increases the activity of echinocandins against Candida auris biofilms. Med. Mycol. 2020, 58, 404–407. [Google Scholar] [CrossRef]
  79. Navarathna, D.H.; Hornby, J.M.; Krishnan, N.; Parkhurst, A.; Duhamel, G.E.; Nickerson, K.W. Effect of farnesol on a mouse model of systemic candidiasis, determined by use of a DPP3 knockout mutant of Candida albicans. Infect. Immun. 2007, 75, 1609–1618. [Google Scholar] [CrossRef] [Green Version]
  80. Hisajima, T.; Maruyama, N.; Tanabe, Y.; Ishibashi, H.; Yamada, T.; Makimura, K.; Nishiyama, Y.; Funakoshi, K.; Oshima, H.; Abe, S. Protective effects of farnesol against oral candidiasis in mice. Microbiol. Immunol. 2008, 52, 327–333. [Google Scholar] [CrossRef]
  81. Martins, M.; Lazzell, A.L.; Lopez-Ribot, J.L.; Henriques, M.; Oliveira, R. Effect of exogenous administration of Candida albicans autoregulatory alcohols in a murine model of hematogenously disseminated candidiasis. J. Basic Microbiol. 2012, 52, 487–491. [Google Scholar] [CrossRef]
  82. Bozó, A.; Domán, M.; Majoros, L.; Kardos, G.; Varga, I.; Kovács, R. The in vitro and in vivo efficacy of fluconazole in combination with farnesol against Candida albicans isolates using a murine vulvovaginitis model. J. Microbiol. 2016, 54, 753–760. [Google Scholar] [CrossRef] [PubMed]
  83. Cordeiro, R.A.; Teixeira, C.E.; Brilhante, R.S.; Castelo-Branco, D.S.; Paiva, M.A.; Giffoni Leite, J.J.; Lima, D.T.; Monteiro, A.J.; Sidrim, J.J.; Rocha, M.F.; et al. Minimum inhibitory concentrations of amphotericin B, azoles and caspofungin against Candida species are reduced by farnesol. Med. Mycol. 2013, 51, 53–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Fernandes Costa, A.; Evangelista Araujo, D.; Santos Cabral, M.; Teles Brito, I.; Borges de Menezes Leite, L.; Pereira, M.; Correa Amaral, A. Development, characterization, and in vitro-in vivo evaluation of polymeric nanoparticles containing miconazole and farnesol for treatment of vulvovaginal candidiasis. Med. Mycol. 2019, 57, 52–62. [Google Scholar] [CrossRef]
  85. Funari, S.S.; Prades, J.; Escribá, P.V.; Barceló, F. Farnesol and geranylgeraniol modulate the structural properties of phosphatidylethanolamine model membranes. Mol. Membr. Biol. 2005, 22, 303–311. [Google Scholar] [CrossRef] [PubMed]
  86. Jabra-Rizk, M.A.; Meiller, T.F.; James, C.E.; Shirtliff, M.E. Effect of farnesol on Staphylococcus aureus biofilm formation and antimicrobial susceptibility. Antimicrob. Agents Chemother. 2006, 50, 1463–1469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Gomes, F.I.; Teixeira, P.; Azeredo, J.; Oliveira, R. Effect of farnesol on planktonic and biofilm cells of Staphylococcus epidermidis. Curr. Microbiol. 2009, 59, 118–122. [Google Scholar] [CrossRef] [Green Version]
  88. Pammi, M.; Liang, R.; Hicks, J.M.; Barrish, J.; Versalovic, J. Farnesol decreases biofilms of Staphylococcus epidermidis and exhibits synergy with nafcillin and vancomycin. Pediatr. Res. 2011, 70, 578–583. [Google Scholar] [CrossRef] [Green Version]
  89. Kim, C.; Hesek, D.; Lee, M.; Mobashery, S. Potentiation of the activity of β-lactam antibiotics by farnesol and its derivatives. Bioorg. Med. Chem. Lett. 2018, 28, 642–645. [Google Scholar] [CrossRef]
  90. Brilhante, R.S.; Valente, L.G.; Rocha, M.F.; Bandeira, T.J.; Cordeiro, R.A.; Lima, R.A.; Leite, J.J.; Ribeiro, J.F.; Pereira, J.F.; Castelo-Branco, D.S.; et al. Sesquiterpene farnesol contributes to increased susceptibility to β-lactams in strains of Burkholderia pseudomallei. Antimicrob. Agents Chemother. 2012, 56, 2198–2200. [Google Scholar] [CrossRef] [Green Version]
  91. Castelo-Branco, D.S.; Riello, G.B.; Vasconcelos, D.C.; Guedes, G.M.; Serpa, R.; Bandeira, T.J.; Monteiro, A.J.; Cordeiro, R.A.; Rocha, M.F.; Sidrim, J.J.; et al. Farnesol increases the susceptibility of Burkholderia pseudomallei biofilm to antimicrobials used to treat melioidosis. J. Appl. Microbiol. 2016, 120, 600–606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Bandara, H.M.; Herpin, M.J.; Kolacny, D., Jr.; Harb, A.; Romanovicz, D.; Smyth, H.D. Incorporation of Farnesol Significantly Increases the Efficacy of Liposomal Ciprofloxacin against Pseudomonas aeruginosa Biofilms in Vitro. Mol. Pharm. 2016, 13, 2760–2770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Černáková, L.; Jordao, L.; Bujdáková, H. Impact of farnesol and Corsodyl® on Candida albicans forming dual biofilm with Streptococcus mutans. Oral. Dis. 2018, 24, 1126–1131. [Google Scholar] [CrossRef] [PubMed]
  94. Cugini, C.; Morales, D.K.; Hogan, D.A. Candida albicans-produced farnesol stimulates Pseudomonas quinolone signal production in LasR-defective Pseudomonas aeruginosa strains. Microbiology 2010, 156, 3096–3107. [Google Scholar] [CrossRef] [Green Version]
  95. Chen, H.; Fujita, M.; Feng, Q.; Clardy, J.; Fink, G.R. Tyrosol is a quorum-sensing molecule in Candida albicans. Proc. Natl. Acad. Sci. USA 2004, 101, 5048–5052. [Google Scholar] [CrossRef] [Green Version]
  96. Alem, M.A.; Oteef, M.D.; Flowers, T.H.; Douglas, L.J. Production of tyrosol by Candida albicans biofilms and its role in quorum sensing and biofilm development. Eukaryot Cell. 2006, 5, 1770–1779. [Google Scholar] [CrossRef] [Green Version]
  97. Bigagli, E.; Cinci, L.; Paccosi, S.; Parenti, A.; D’Ambrosio, M.; Luceri, C. Nutritionally relevant concentrations of resveratrol and hydroxytyrosol mitigate oxidative burst of human granulocytes and monocytes and the production of pro-inflammatory mediators in LPS-stimulated RAW 264.7 macrophages. Int. Immunopharmacol. 2017, 43, 147–155. [Google Scholar] [CrossRef]
  98. Rigacci, S.; Stefani, M. Nutraceutical Properties of Olive Oil Polyphenols. An Itinerary from Cultured Cells through Animal Models to Humans. Int. J. Mol. Sci. 2016, 17, 843. [Google Scholar] [CrossRef] [Green Version]
  99. Cremer, J.; Vatou, V.; Braveny, I. 2,4-(hydroxyphenyl)-ethanol, an antioxidative agent produced by Candida spp., impairs neutrophilic yeast killing in vitro. FEMS Microbiol. Lett. 1999, 170, 319–325. [Google Scholar] [CrossRef]
  100. Holland, L.M.; Schröder, M.S.; Turner, S.A.; Taff, H.; Andes, D.; Grózer, Z.; Gácser, A.; Ames, L.; Haynes, K.; Higgins, D.G.; et al. Comparative phenotypic analysis of the major fungal pathogens Candida parapsilosis and Candida albicans. PLoS Pathog. 2014, 10, e1004365. [Google Scholar] [CrossRef] [Green Version]
  101. Monteiro, D.R.; Feresin, L.P.; Arias, L.S.; Barão, V.A.; Barbosa, D.B.; Delbem, A.C. Effect of tyrosol on adhesion of Candida albicans and Candida glabrata to acrylic surfaces. Med. Mycol. 2015, 53, 656–665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Babich, H.; Visioli, F. In vitro cytotoxicity to human cells in culture of some phenolics from olive oil. Farmaco 2003, 58, 403–407. [Google Scholar] [CrossRef]
  103. Arias, L.S.; Delbem, A.C.; Fernandes, R.A.; Barbosa, D.B.; Monteiro, D.R. Activity of tyrosol against single and mixed-species oral biofilms. J. Appl. Microbiol. 2016, 120, 1240–1249. [Google Scholar] [CrossRef] [Green Version]
  104. Do Vale, L.R.; Delbem, A.; Arias, L.S.; Fernandes, R.A.; Vieira, A.; Barbosa, D.B.; Monteiro, D.R. Differential effects of the combination of tyrosol with chlorhexidine gluconate on oral biofilms. Oral Dis. 2017, 23, 537–541. [Google Scholar] [CrossRef] [PubMed]
  105. Monteiro, D.R.; Arias, L.S.; Fernandes, R.A.; Deszo da Silva, L.F.; De Castilho, M.; Da Rosa, T.O.; Vieira, A.; Straioto, F.G.; Barbosa, D.B.; Delbem, A. Antifungal activity of tyrosol and farnesol used in combination against Candida species in the planktonic state or forming biofilms. J. Appl. Microbiol. 2017, 123, 392–400. [Google Scholar] [CrossRef] [PubMed]
  106. Monteiro, D.R.; Arias, L.S.; Fernandes, R.A.; Straioto, F.G.; Barros Barbosa, D.; Pessan, J.P.; Delbem, A. Role of tyrosol on Candida albicans, Candida glabrata and Streptococcus mutans biofilms developed on different surfaces. Am. J. Dent. 2017, 30, 35–39. [Google Scholar]
  107. Shanmughapriya, S.; Sornakumari, H.; Lency, A.; Kavitha, S.; Natarajaseenivasan, K. Synergistic effect of amphotericin B and tyrosol on biofilm formed by Candida krusei and Candida tropicalis from intrauterine device users. Med. Mycol. 2014, 52, 853–861. [Google Scholar] [CrossRef] [Green Version]
  108. Cordeiro, R.; Teixeira, C.E.; Brilhante, R.S.; Castelo-Branco, D.S.; Alencar, L.P.; De Oliveira, J.S.; Monteiro, A.J.; Bandeira, T.J.; Sidrim, J.J.; Moreira, J.L.; et al. Exogenous tyrosol inhibits planktonic cells and biofilms of Candida species and enhances their susceptibility to antifungals. FEMS Yeast Res. 2015, 15, fov012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Kovács, R.; Tóth, Z.; Nagy, F.; Daróczi, L.; Bozó, A.; Majoros, L. Activity of exogenous tyrosol in combination with caspofungin and micafungin against Candida parapsilosis sessile cells. J. Appl. Microbiol. 2017, 122, 1529–1536. [Google Scholar] [CrossRef]
  110. Abdel-Rhman, S.H.; Rizk, D.E. Effect of tyrosol on Staphylococcus aureus antimicrobial susceptibility, biofilm formation and virulence factors. Afr. J. Microbial. Res. 2016, 10, 687–693. [Google Scholar] [CrossRef]
  111. Abdel-Rhman, S.H.; El-Mahdy, A.M.; El-Mowafy, M. Effect of Tyrosol and Farnesol on Virulence and Antibiotic Resistance of Clinical Isolates of Pseudomonas aeruginosa. Biomed. Res. Int. 2015, 2015, 456463. [Google Scholar] [CrossRef] [PubMed] [Green Version]

Share and Cite

MDPI and ACS Style

Kovács, R.; Majoros, L. Fungal Quorum-Sensing Molecules: A Review of Their Antifungal Effect against Candida Biofilms. J. Fungi 2020, 6, 99. https://doi.org/10.3390/jof6030099

AMA Style

Kovács R, Majoros L. Fungal Quorum-Sensing Molecules: A Review of Their Antifungal Effect against Candida Biofilms. Journal of Fungi. 2020; 6(3):99. https://doi.org/10.3390/jof6030099

Chicago/Turabian Style

Kovács, Renátó, and László Majoros. 2020. "Fungal Quorum-Sensing Molecules: A Review of Their Antifungal Effect against Candida Biofilms" Journal of Fungi 6, no. 3: 99. https://doi.org/10.3390/jof6030099

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop