Skip to main content

Soluble factors from biofilm of Candida albicans and Staphylococcus aureus promote cell death and inflammatory response

Abstract

Background

The objective of this study was to better understand the effects of soluble factors from biofilm of single- and mixed-species Candida albicans (C. albicans) and methicillin-sensitive Staphylococcus aureus (MSSA) cultures after 36 h in culture on keratinocytes (NOK-si and HaCaT) and macrophages (J774A.1). Soluble factors from biofilms of C. albicans and MSSA were collected and incubated with keratinocytes and macrophages, which were subsequently evaluated by cell viability assays (MTT). Lactate dehydrogenase (LDH) enzyme release was measured to assess cell membrane damage to keratinocytes. Cells were analysed by brightfield microscopy after 2 and 24 h of exposure to the soluble factors from biofilm. Cell death was detected by labelling apoptotic cells with annexin V and necrotic cells with propidium iodide (PI) and was visualized via fluorescence microscopy. Soluble factors from biofilm were incubated with J774A.1 cells for 24 h; the subsequent production of NO and the cytokines IL-6 and TNF-α was measured by ELISA.

Results

The cell viability assays showed that the soluble factors of single-species C. albicans cultures were as toxic as the soluble factors from biofilm of mixed cultures, whereas the soluble factors of MSSA cultures were less toxic than those of C. albicans or mixed cultures. The soluble factors from biofilm of mixed cultures were the most toxic to the NOK-si and HaCaT cells, as confirmed by analyses of PI labelling and cell morphology. Soluble factors from biofilm of single-species MSSA and mixed-species cultures induced the production of IL-6, NO and TNF-α by J744A.1 macrophages. The production of IL-6 and NO induced by the soluble factors from biofilm of mixed cultures was lower than that induced by the soluble factors from biofilm of single-species MSSA cultures, whereas the soluble factors from biofilm of C. albicans cultures induced only low levels of NO.

Conclusions

Soluble factors from 36-h-old biofilm of C. albicans and MSSA cultures promoted cell death and inflammatory responses.

Background

Biofilm represents a microbial community enclosed in a matrix of extracellular polymeric substances (EPS). Biofilm formation is a form of microbial growth in which interacting sessile cells are anchored to a solid substratum and to each other [1, 2]. Among the opportunistic pathogens in the oral cavity, Candida albicans (C. albicans) is most frequently isolated in denture bases (64.4%) [3]. Methicillin-sensitive Staphylococcus aureus (MSSA) has been recovered from 34.4% of denture users, and the combination of these two microorganisms is found in 8.8% of patients [3]. C. albicans and MSSA form a mutual alliance that promotes a positive synergism between the species [4, 5], which has been attributed to increased frequency and severity of infectious diseases [6] such as prosthetic stomatitis, the most common form of oral candidosis, with an overall incidence of 11–65% in users of complete prostheses [7,8,9]. Moreover, C. albicans and MSSA have also been co-isolated from individuals with various pathologies, as well as from the surfaces of various biomaterials such as catheters [10, 11].

These opportunistic pathogens can colonize mucous membranes, invade tissues and cause infection [12]. Their pathogenicity is attributed to several factors, such as the ability to develop biofilms, drug resistance, and the production of toxic metabolites and toxins [13]. C. albicans and MSSA biofilms are rich in proteases and phospholipase. When these microorganism biofilms are co-cultured, both phospholipase C (PL-C) and proteases (SAP) can be found [5]. Furthermore, the interaction between MSSA and C. albicans promotes a strong inflammatory response in polymicrobial infections and modulates the proteomic profiles of biofilm in co-cultures in vitro. These modulatory effects include the expression of several defined and putative virulent proteins, such as CodY, which regulates nutrient acquisition and toxin production [4].

The ability of microorganisms to induce cell damage is a key factor promoting proinflammatory responses leading to recruitment and activation of immune cells, such as neutrophils and macrophages [14]. Significantly higher levels of systemic and local interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α) and IL-1β have been found during the early stages of co-infection than during single infection, regardless of the morphogenesis of C. albicans [15]. The concentrations of IL-6 and IL-8 produced by HaCaT keratinocytes increased when incubated with C. albicans filtrates [16]. The pathogenicity of MSSA is due to its repertoire of toxins, exoenzymes, adhesins, and immune-modulating proteins [17]. Compared with monomicrobial peritonitis, polymicrobial peritonitis is associated with increased proinflammatory cytokines such as IL-6, keratinocyte chemoattractant, macrophage inflammatory protein-1α, monocyte chemoattractant protein-1, and granulocyte colony-stimulating factor [11]. Soluble factors from biofilm of MSSA are able to induce the production of IL-1β, IL-6, TNF-α, CXCL-8 and CXCL-1 in human keratinocytes as shown by ELISA [18]. The highest concentrations of IL-6 and TNF-α were detected in human mononuclear cells incubated in the presence of MSSA [19].

Therefore, the study investigated the effects of soluble factors derived from C. albicans and MSSA biofilms on epithelial cell death and macrophage inflammatory responses. To identify synergism in the pathogenicity and virulence of these microorganisms, the effects of biofilms from single- and mixed-species cultures were compared.

Results

Characterization of single- and mixed-species biofilms

The biofilm of mixed MSSA cultures showed higher log10CFU/mL than the biofilm of single cultures, although no such difference was observed between single-species and mixed-species C. albicans-derived biofilms (Table 1). The pH of soluble factors from biofilm of C. albicans (pH = 7.03), MSSA (pH = 6.89), and mixed cultures (pH = 7.00) remained near physiologic levels (pH = 7.0), showing that pH did not affect the cell viability rates.

Table 1- Growth of single and mixed C. albicans and MSSA biofilms after 36 h, performed three times in triplicate (n = 9 samples)

Protein content measurements by Bradford assay showed a lower total protein level of soluble factors in biofilm from C. albicans cultures (3.12 μg/mL ± 1.10) than from MSSA (21.44 μg/mL ± 0.53) or mixed cultures (19.21 μg/mL ± 1.35) (Fig. 1).

Fig. 1
figure 1

Total protein contained in the biofilm metabolites produced by single and mixed-species biofilms by the Bradford method. Different letters means significant differences (p˂0.05)

Cell viability

The cell viability of NOK-si cells started to decrease after 6 h of incubation with soluble factors from biofilm of single- C. albicans and mixed-species cultures. Cell viability after incubation with MSSA soluble factors remained unaltered, with minimum decrease after 12 h of incubation. Similar results were obtained when HaCaT cells were incubated with soluble factors from biofilm of single- C. albicans and mixed-species cultures. The greatest reductions in cell viability were achieved within 24 h for all groups (Fig. 2a and b).

Fig. 2
figure 2

NOK-si (a) and HaCat (b) cell viability after being in contact with soluble factors from biofilm for 2, 4, 6, 8, 12 and 24 h in three independent experiments, in triplicate for each experimental condition (n = 9). Error bars represent standard error. * versus C. albicans and mixed biofilm. # versus MSSA and mixed biofilm. Analysis by ANOVA, Tukey post-test, p ≥ 0.05. RPMI = Negative control, C. albicans = soluble factors from biofilm of C. albicans, MSSA = Soluble factors from biofilm of MSSA, Mixed = soluble factors from mixed biofilm, TL = lysis buffer

Cell membrane damage

The soluble factors from biofilms of mixed cultures significantly increased LDH release compared with those from single-species C. albicans and MSSA cultures after 8 h of incubation, reaching higher LDH levelsin 24 h. The soluble factors in biofilm from MSSA induced less LDH release (Fig. 3a and b).

Fig. 3
figure 3

LDH release from NOK-si (a) and HaCat (b) cells after incubation with soluble factors from biofilm of for 2, 4, 6, 8, 12 and 24 h. Three independent experiments were performed in triplicate for each experimental condition (n = 9). DMEM = negative control; RPMI = negative control; C. albicans = soluble factors from biofilm of C. albicans; MSSA = Soluble factors from biofilm of MSSA; Mixed = soluble factors from mixed biofilm. (Tukey post-hoc test at ρ >0.05). Different letters for results with significant difference in relation to growth conditions (C. albicans, MSSA and Mixed biofilm)

Cell morphology of keratinocytes

No changes in the morphology of NOK-si and HaCaT cells were observed after only 2 h of contact with soluble factors from biofilm of single-species C. albicans or MSSA cultures or mixed-species cultures. Major changes in morphology (i.e., loose and spherical cells, cellular debris, and decreased number of cells) were evident after 24 h of incubation with the soluble factors from biofilms of the three types of cultures (Fig. 4a and b).

Fig. 4
figure 4

a Images obtained from inverted microscope for the control group (DMEM and RPMI) and experimental groups (C. albicans, MSSA and Mixed) after 2 and 24 h of contact with NOK-si. b Images obtained from inverted microscope for the control group (DMEM and RPMI) and experimental groups (C. albicans, MSSA and Mixed) after 2 and 24 h of contact with HaCat. The bar in the images corresponds to 100 μm

Cell death by necrosis or apoptosis

Because we detected more LDH release from the NOK-si and HaCaT cells (which suggested necrosis) after incubation with the soluble factors from biofilm of mixed-species cultures than after incubation with the soluble factors from biofilm of single-species cultures, NOK-si and HaCaT cells were labelled with annexin V and PI. The cells that were incubetedwith the soluble factors from biofilm of mixed cultures were positively labelled with annexin V. However, more abundant PI staining of cells incubated with soluble factors from biofilm of mixed cultures than single-species cultures suggested that the microorganism synergism caused greater disruption in the integrity of epithelial cell membranes, resulting in cell death via late apoptosis or necrosis. Cells were also positively labelled with annexin V and, to a lesser extent, PI when exposed to the soluble factors from biofilm of single-species C. albicans cultures. Fewer cells positively labelled with annexin V and PI were observed when the cells were exposed to the soluble factors from biofilm of MSSA cultures (Fig. 5).

Fig. 5
figure 5

Detection of apoptotic cells (staining positively for annexin V in green) and necrotic cells (staining positively for PI in red) with soluble factors from C. albicans, MSSA and mixed biofilm. Magnification ×200

Production of cytokines by macrophages

Macrophages exposure to soluble factors from biofilms demonstrated unaltered cell viability (Fig. 6a). ELISA revealed that the J774A.1 macrophages stimulated with soluble factors from biofilms of MSSA cultures had greater production levels of NO (nitric oxide), IL-6 and TNF-α than those exposed to soluble factors from biofilms of C. albicans cultures. When stimulated with soluble factors from biofilms of mixed cultures, TNF-α production by J774A.1 cells remained at similarly high levels to those stimulated with soluble factors from biofilms of MSSA cultures, with the exception of IL-6 and NO, which were at higher levels in cells stimulated with soluble factors from biofilms of MSSA cultures (Fig. 6b).

Fig. 6
figure 6

Macrophage response after challenge with soluble-factors produced by C. albicans and MSSA single- and mixed-species biofilms. a Toxicity percentage according to soluble factors from biofilm in contact with the J774A.1 macrophage, tested on three occasions performed in triplicate (n = 9 samples). b IL-6 (A), NO (B) and TNF-α (C) production from J774A.1 macrophages after 24 h stimulation with soluble factors from MSSA, C. albicans and mixed biofilm. Error bars represent standard deviation. Symbols represent statistical differences. IL-6, # versus Mixed; NO e, * versus C. albicans; # versus Mixed; TNF-alpha, * versus C. albicans. Analysis by ANOVA, Tukey post-test, p ≥ 0.05. CLT = control (DMEM), C. albicans = C. albicans soluble-factors, MSSA = MSSA soluble-factors, Mixed = mixed soluble-factors. N.D. = not detectable

Discussion

The present study demonstrated that soluble factors from biofilm of mixed-species cultures were more pathogenic to epithelial cells than those from biofilms of single-species cultures. Moreover, soluble factors from biofilm of single-species C. albicans cultures promoted modest cytokine production by J774A.1 macrophages, whereas soluble factors from biofilm of single MSSA cultures yielded higher levels of cytokines.

Biofilm characterization revealed higher log10CFU/mL MSSA in biofilms of mixed-species cultures than in those of single-species cultures (Table 1). Prostaglandin E2 produced by C. albicans and present in biofilms of mixed-species cultures consisting of C. albicans and MSSA, is known to stimulate the growth of MSSA [20]. Moreover, MSSA has an affinity for C. albicans hyphae [4, 5] and accordingly, more MSSA biomass is retained after washing, which may account for the higher log10CFU/mL MSSA in biofilms from mixed–species cultures. Additionally, the biofilm of single-species MSSA cultures and the biofilm of mixed-species cultures induced higher levels of total protein production than the biofilm of single-species C. albicans cultures (Fig. 1). Although no differences of total protein content were observed between the soluble factors from biofilm of MSSA cultures and mixed cultures, differential in-gel electrophoresis has shown significant differences in the production of 27 proteins by MSSA and C. albicans during co-culture biofilm growth, including CodY, which regulates toxin production and nutrient acquisition [4].

The soluble factors from biofilms of mixed-species cultures were more damaging to both NOK-si and HaCaT cells than the soluble factors from biofilms of single C. albicans and MSSA cultures. This observation, however, was not evident from the MTT assay, wherein the decreased rates of cell viability were similar between soluble factors from biofilm of single C. albicans and mixed cultures. Nevertheless, the LDH assay revealed greater LDH release from both cell types after incubation with the soluble factors from biofilm of mixed cultures. Hydrolytic enzymes, such as phospholipases (PL-C) and proteinase, are metabolites known to be present in C. albicans biofilms [21, 22]. Moreover, greater proteinase production has been reported in biofilms of single-species MSSA cultures, whereas biofilms of single-species C. albicans cultures presented greater levels of PL-C. When the two microorganisms were co-cultured, both enzymes were produced [5]. The activation of phosphatidylinositol-specific PL-C increases cellular calcium levels and may in turn participate in apoptosis [23]. Thus, PL-C production may be associated with a greater number of apoptotic keratinocyte cells following exposure to soluble factors from biofilms of single-species C. albicans and mixed-species cultures (Fig. 5). However, future studies using inhibitors of these enzymes in mixed cultures and monocultures would be required to confirm these hypotheses.

The LDH results demonstrated that soluble factors from 36-h-old biofilms were harmful to NOK-si and HaCaT cells, regardless of the source of the soluble factors. However, soluble factors from biofilm of mixed-species cultures produced more cell membrane damage, indicating the increased pathogenicity of the soluble factors from biofilm of mixed-species cultures (Fig. 3a and b). This more pathogenic behaviour was also demonstrated in the annexin V/PI assays. Under such conditions, the NOK-si and HaCaT cells were double-stained with annexin V and PI. In contrast, when the cells were exposed to soluble factors from biofilm of single-species cultures, few cells were positively stained with PI, although positive PI labelling was more frequent in the C. albicans group (Fig. 5). Additionally, C. albicans secretes farnesol [24], which is capable to induce apoptosis via activation of caspase, the production of reactive oxygen species (ROS) and the disruption of mitochondrial integrity, resulting in cell death [10, 25, 26].

Interestingly, higher levels of IL-6 production by J774A.1 cells were induced in the presence of soluble factors from biofilm of MSSA cultures. Lower levels of this cytokine were also detected when macrophages were exposed to soluble factors from biofilm of mixed cultures. MSSA toxins (e.g. exotoxins, protein A and α-toxins) do not possess direct cell damaging action, but have a potent effect on cells of the immune system by inducing the overproduction of cytokines [27]. Previous studies have shown that lipoteichoic acid is a potent stimulus that induces IL-6 production by monocyte-like cell lines [28, 29]. IL-6 can inhibit apoptosis during the inflammatory process, keeping cells alive even in highly toxic environments [30]. Through an uncertain mechanism, the soluble factors from biofilm of C. albicans interacted with those of S. aureus, causing the soluble factors from biofilm of mixed cultures to induce lower levels of IL-6 production by macrophages. However, high levels of TNF-α can trigger the extrinsic apoptotic pathway through recruitment TNF receptor 1 (TNFR1) along with TRADD and other molecules [31]. Furthermore, post-translational modifications can promote RIP1 and TRADD dissociation, leading to the exposure of their death domains (DD), which bind to FADD and recruit caspases-8 and -10, resulting in apoptosis [31]. While we did not perform experiments to determine whether supernatant from macrophages could affect cell death in NOK-si and HaCaT cells, our results suggest that the different amounts of TNF- α and IL-6 generated by macrophages in this microenvironment could affect the cell death of keratinocytes.

The presence of C. albicans has been shown to be capable of blocking nitric oxide (NO) production by macrophages [32]. In this study, the soluble factors from biofilm of single-species C. albicans cultures likely mediated the same effect. The same behaviour was observed for NO production by J774A.1 cells after exposure to soluble factors from biofilm for 24 h. NO production was observed when the macrophages were incubated with soluble-factors from biofilm of single-species MSSA cultures, and less NO production was observed when the macrophages were incubated with soluble-factors from biofilm of mixed-species cultures than single-species MSSA cultures. In contrast, soluble factors from single-species C. albicans cultures induced low levels of NO production by J774A.1 cells (Fig. 6b).

The present study showed that C. albicans interacted with S. aureus in the microenvironment of 36-h-old biofilm of mixed-species cultures such that the soluble factors of this biofilm were less able to induce IL-6 and NO production than the soluble factors from biofilm of 36-h-old S. aureus cultures. This behaviour was not observed for TNF production, which may be due to the lower production of NO induced by the soluble factors from biofilm of mixed-species cultures. Indeed, the induction of NO production has been related to higher levels of TNF-α [33].

This field requires further study to determine the biochemistry of these soluble factors, the mechanisms involved in human cell death following exposure to soluble factors from biofilm of single- and mixed-species cultures, and the inflammatory pathways induced by these factors.

Conclusion

The results of the present study show that soluble factors from 36-h-old biofilms of C. albicans and S. aureus cultures promote cell death and inflammatory responses. During the growth of the biofilms, the presence of C. albicans evidently enhanced the damage to keratinocytes caused by soluble factors from biofilm of mixed cultures, triggering major necrotic cell death. However, the soluble factors from biofilm of mixed cultures were less capable of inducing the production of proinflammatory cytokines than the soluble factors from biofilm of S. aureus cultures.

Methods

Microbial strains and growth conditions

C. albicans SC5314 and MSSA ATCC25923 microorganisms were used to produce single and dual species biofilms, in accordance with the methodology described by Zago et al. [5]. After incubation, seven freshly grown colonies of MSSA were transferred to 10 mL of TSB medium for pre-inoculum growth at 37 °C for 18 h. For C. albicans, 10 freshly grown colonies were transferred to 10 mL of Yeast Nitrogen Base broth culture media (YNB; Difco, Becton Dickinson, Sparks, MD, USA) supplemented with 100 mM glucose for pre-inoculum growth at 37 °C for 16 h. Thereafter, the dilution of the inoculum was performed, and cultures were incubated until the mid-exponential growth phase. The cells of the resultant cultures were harvested and washed twice with sterile phosphate buffered saline solution (PBS, pH 7.2). Microorganisms were resuspended in RPMI-1640 culture medium (Sigma-Aldrich, St. Louis, MO, USA) supplemented with HEPES (25 mM), L-glutamine (2.0 mM) and sodium bicarbonate (2.0 g/L) (Sigma-Aldrich, St. Louis, MO, USA) [34]. The optical densities of the suspensions were standardized to 1 × 107 CFU/mL for both microorganisms.

Adhesion and biofilm formation

Biofilm formation was carried out in 24-well microplates (TPP Techno Plastic Products AG, Switzerland) [35]. Adhesion and biofilm formation was realized according to protocol recommended by Zago et al. [5]. Experiments were performed in three replicates and repeated in three independent assays. pH was measured using a benchtop pH meter (QX 1500 Plus-Qualxtron, São Paulo, Brazil).

To obtain the soluble factors from the 36-h-old biofilms, media (supernatants) and the microorganisms attached to the wells were removed and filtered through a 0.22-μm low-protein-binding filter (SFCA, Corning, Germany). For epithelial cells (NOK-si and HaCaT), the undiluted soluble factors from biofilm were applied for 2, 4, 6, 8, 12 and 24 h.

Keratinocyte cell cultures

NOK-si was kindly provided by Prof. Dr. Carlos Rossa Junior (Department of Periodontology, School of Dentistry of Araraquara-UNESP, Brazil) [36], and HaCaT (BCRJ 0341) and J774A.1 macrophages (BCRJ 0121) were purchased from the Rio de Janeiro Cell Bank (BCRJ, RJ, Brazil). The cells were cultured in Dulbecco’s Modified Eagles Medium (DMEM, Gibco BRL, Grand Island, NJ, USA) containing 10% fetal bovine serum (FBS, GIBCO, Grand Island, NY), 100 IU/mL penicillin, 100 mg/mL streptomycin (Sigma Chemical Co., St. Louis, MO, USA) and 2 mL/L glutamine (GIBCO, Grand Island, NY). The cells were maintained at 37 °C under 5% CO2 and 80% humidity. The cells were grown until confluence (90%), counted in a Neubauer chamber (magnification ×10) and plated (4.5 × 104 cells/well for NOK-si and HaCaT and 1.0 × 105 cells/well for J774A.1). Cells were used between the 3rd and 8th passages.

Proteins contained in the supernatant

The total level of soluble protein contained in the soluble factors from biofilm was measured by Bradford protein assay [37], using bovine serum albumin (BSA, Sigma-Aldrich, St. Louis, MO, USA) as the standard. Spectrophotometric measurements were performed at 595 nm (400 EZ Reader; Biochrom, Cambridge, UK).

Cell viability

For cellular metabolism analyses, the mitochondrial activity of the keratinocytes was measured using the MTT assay [3-(4.5-dimethylthiazole-2-yl) 2.5-diphenyl tetrazolium bromide] (Sigma-Aldrich, St. Louis, MO, USA) [38], which was performed at 2, 4, 6, 8, 12 and 24 h after incubation at 37 °C in 5% CO2 in RPMI (negative control), C. albicans, MSSA and mixed soluble factors from biofilm, and lysis buffer containing Triton X-100 (LB, positive control). After each period of contact, the cells were washed with PBS. Then, 250 μL of MTT (2.5 mg/mL) were added to each well and the plates were incubated for 4 h. Next, MTT was removed and the formed formazan crystals were solubilized in 250 μL of 2-propanol. Spectrophotometric measurements were performed at 562 nm (Reader 400 EZ; Biochrom, Cambridge, UK).

Cell membrane damage

The release of the enzyme lactate dehydrogenase (LDH) was determined after 2, 4, 6, 8, 12 and 24 h of contact with the soluble factors using the CytoTox-96 nonradioactive cytotoxicity assay (Promega, Madison, WI) according to the manufacturer’s recommendations. Initially, 100 μL of each control (RPMI, DMEM and lysis buffer containing TritonX-100) and the experimental samples (soluble factors from biofilm) were added in triplicate to a 96-well plate (Thermo Scientific; #31125). Next, 100 μL of the CytoTox-ONE TM reagent was added, and the plate was incubated for 10 min. Subsequently, 50 μL of a “stop” solution were added to each well, and fluorescence was then measured with a filter combination of 544 nm/590 nm (Fluoroskan, FL Ascent, Labsystems, Helsinki, Finland). Culture medium and soluble factors from biofilm were used as blanks.

Evaluation of keratinocyte morphology

After 2 and 24 h of contact with soluble factors from biofilm, keratinocytes were analysed and photographed by brightfield microscopy using a Leica DMI 3000B microscope (Leica Microsystems, Wetzlar, Germany) soluble factors from biofilm.

Cell death assay

The type of NOK-si and HaCaT cell death (apoptosis/necrosis) induced by soluble factors from biofilm of MSSA, C. albicans and mixed cultures was investigated using annexin V/Alexa Fluor 488 and PI (594 nm) (Molecular Probes, Invitrogen). Annexin V binds specifically to phosphatidylserine residues on cell membranes during apoptosis. PI intercalates with the broken DNA, a typical process of cell necrosis [39, 40]. After 8 h (in the range of 8–12 h, approximately 10 h) of contact with the soluble factors from biofilm of single- and mixed-species cultures, the cells were washed twice with binding buffer (10 mM HEPES/NaOH, 140 mM NaCl, 2.5 mM CaCl2, pH = 7.4) and incubated with annexin V (10 μL/well) and 2 μL of PI (100 μg/mL) for 20 min at room temperature. The cells were then washed with PBS and maintained in 10% DMEM for analysis with an inverted fluorescence microscope (Leica DMI 3000B; Leica Microsystems, Wetzlar, Germany).

Cytokines produced by macrophages

J774A.1 macrophages were continuously exposed to soluble factors from biofilm of C. albicans, MSSA or mixed cultures for 24 h, at a ratio of 1:4 (soluble factors from biofilm: cell culture medium) to avoid cell death (as determined experimentally by MTT assays; Fig. 6a). Subsequently, macrophage supernatants were collected, and cytokine (IL-6, TNF-α and NO) production was measured by enzyme-linked immunosorbent assay (ELISA; BD Biosciences, San Jose, CA).

Statistical analysis

The normality and homogeneity of variances were evaluated using the Shapiro-Wilk and Levene tests. The results were evaluated using one-way analysis of variance (ANOVA) followed by Tukey’s test. A 5% significance level was adopted for all tests performed (p˂0.05). All studies were performed in triplicate for each experimental condition and repeated on three independent occasions (n = 9).

Abbreviations

BHI:

Brain heart infusion

BSA:

Bovine serum albumin

C. albicans :

Candida albicans

CFUs:

Colony-forming units

DMEM:

Dulbecco’s modified eagle’s

ELISA:

Enzyme-linked immunosorbent assay

EPS:

Extracellular polymeric substances

FBS:

Fetal bovine serum

HaCat:

Human adult skin keratinocytes propagated under low Ca2+ conditions and elevated temperature

IL-6:

Interleucin-6

J774A.1:

Macrophages

LDH:

Lactate dehydrogenase

MSSA:

Methicillin-sensitive Staphylococcus aureus

MTT:

3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

NO:

Nitric oxide

NOK-si:

Spontaneously immortalized normal oral keratinocytes

OD:

Optical density

PBS:

Phosphate buffered saline

PI:

Propidium iodide

PL-C:

Phospholipase C

RPMI:

Roswell Park Memorial Institute

SAP:

Proteases

SDA:

Sabouraud dextrose agar

TNF-α:

Tumour necrosis factor alpha

TSB:

Tryptic soy broth medium

YEPD:

Yeast peptone glucose medium

References

  1. Socransky SS, Haffajee AD. Periodontal microbial ecology. Periodontol. 2005;38:135–87.

    Article  Google Scholar 

  2. Nobile CJ, Mitchell AP. Microbial biofilms: e pluribus unum. Curr Biol. 2007;17:349–53.

    Article  Google Scholar 

  3. Ribeiro DG, Pavarina AC, Dovigo LN, Machado AL, Giampaolo ET, Vergani CE. Prevalence of Candida spp. associated with bacteria species on complete dentures. Gerodontology. 2012;29:203–8.

    Article  PubMed  Google Scholar 

  4. Peters BM, Jabra-Rizk MA, Scheper MA, Leid JG, Costerton JW, Shirtliff ME. Microbial interactions and differential protein expression in Staphylococcus aureus -Candida albicans dual-species biofilms. FEMS Immunol Med Microbiol. 2010;59(3):493–503.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Zago CE, Silva S, Sanitá PV, Barbugli PA, Dias CM, Lordello VB, et al. Dynamics of biofilm formation and the interaction between Candida albicans and methicillin-susceptible (MSSA) and -resistant Staphylococcus aureus (MRSA). PLoS One. 2015;10(4):e0123206.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Morales DK, Hogan DA. Candida albicans Interactions with bacteria in the context of human health and disease. PLoS Pathog. 2010;6(4):e1000886.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Jeganathan S, Lin CC. Denture stomatitis--a review of the aetiology, diagnosis and management. Aust Dent J. 1992;37(2):107–14.

    Article  CAS  PubMed  Google Scholar 

  8. Marchini L, Tamashiro E, Nascimento DF, Cunha VP. Self-reported denture hygiene of a sample of edentulous attendees at a university dental clinic and the relationship to the condition of the oral tissues. Gerodontology. 2004;24(4):226–8.

    Article  Google Scholar 

  9. Coelho CM, Sousa YT, Daré AM. Denture-related oral mucosal lesions in a Brazilian school of dentistry. J Oral Rehabil. 2004;31(2):135–9.

    Article  CAS  PubMed  Google Scholar 

  10. Shirtliff ME, Krom BP, Meijering RA, et al. Farnesol-induced apoptosis in Candida albicans. Antimicrob Agents Chemother. 2009;53:2392–401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Peters BM, Noverr MC. Candida albicans-Staphylococcus aureus polymicrobial peritonitis modulates host innate immunity. Infect Immun. 2013;81(6):2178–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Calderone RA, Fonzi WA. VirμLence factors of Candida albicans. Trends Microbiol. 2001;9:327–35.

    Article  CAS  PubMed  Google Scholar 

  13. Palavecino E. Community-acquired methicillin-resistant Staphylococcus aureus infections. Clin Lab Med. 2004;24:403–18.

    Article  PubMed  Google Scholar 

  14. Villar CC, Zhao XR. Candida albicans Induces early apoptosis followed by secondary necrosis in oral epithelial cells. Mol Oral Microbiol. 2010;25(3):215–25.

    Article  CAS  PubMed  Google Scholar 

  15. Nash EE, Peters BM, Palmer GE, Fidel PL, Noverr MC. Morphogenesis is not required for Candida albicans-Staphylococcus aureus intra-abdominal infection mediated dissemination and lethal sepsis. Infect Immun. 2014;82(8):3426–35.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Wollina U, Künkel W, Bulling L, Fünfstück C, Knöll B, Vennewald I, et al. Candida albicans-induced inflammatory response in human keratinocytes. Mycoses. 2004;47(5–6):193–9.

    Article  CAS  PubMed  Google Scholar 

  17. Fournier B, Philpott DJ. Recognition of Staphylococcus aureus by the innate immune system. Clin Microbiol Rev. 2005;18(3):521–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Secor PR, James GA, Fleckman P, Olerud JE, McInnerney K, Stewart PS. Staphylococcus aureus Biofilm and Planktonic cultures differentially impact gene expression, mapk phosphorylation, and cytokine production in human keratinocytes. BMC Microbiol. 2011;11:143.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Knop J, Hanses F, Leist T, Archin NM, Buchholz S, Gläsner J, et al. Staphylococcus aureus Infection in humanized mice: a new model to study Pathogenicity associated with human immune response. J Infect Dis. 2015;212(3):435–44.

    Article  CAS  PubMed  Google Scholar 

  20. Krause J, Geginat G, Tammer I. Prostaglandin E2 from Candida albicans stimulates the growth of Staphylococcus aureus in mixed biofilms. Seneviratne CJ. PLoS One. 2015;10(8): e0135404.

  21. Lyon JP, Resende MA. Correlation between adhesion, enzyme production, and susceptibility to fluconazole in Candida albicans obtained from denture wearers. Oral Med Oral Pathol Oral Radiol Endod. 2006;102:632–8.

    Article  Google Scholar 

  22. Pinto E, Ribeiro IC, Ferreira NJ, Fortes CE, Fonseca PA, Figueiral MH. Correlation between enzyme production, germ tube formation and susceptibility to fluconazole in Candida species isolated from patients with denture-related stomatitis and control individuals. J Oral Pathol Med. 2008;37:587–92.

    Article  CAS  PubMed  Google Scholar 

  23. Min BM, Woo KM, Lee G, Park NH. Terminal differentiation of normal human oral keratinocytes is associated with enhanced cellular TGF-beta and phospholipase C-gamma 1 levels and apoptotic cell death. Exp Cell Res. 1999;249(2):377–85.

    Article  CAS  PubMed  Google Scholar 

  24. Ramage G, Saville SP, Wickes BL, López-Ribot JL. Inhibition of Candida albicans biofilm formation by farnesol, a quorum sensing molecule. Appl Environ Microbiol. 2002;68(11):5459–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Henneberry AL, Wright MM, McMaster CR. The major sites of cellular phospholipid synthesis and molecular determinants of fatty acid and lipid head group specificity. Mol Biol Cell. 2002;13(9):3148–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wiseman DA, Werner SR, Crowell PL. Cell cycle arrest by the isoprenoids perillyl alcohol, geraniol, and farnesol is mediated by p21(Cip1) and p27(Kip1) in human pancreatic adenocarcinoma cells. J Pharmacol Exp Ther. 2007;320:1163–70.

    Article  CAS  PubMed  Google Scholar 

  27. Ezepchuk YV, Leung DY, Middleton MH, Bina P, Reiser R, Norris DA. Staphylococcal toxins and protein a differentially induce cytotoxicity and release of tumor necrosis factor-alpha from human keratinocytes. J Invest Dermatol. 1996;107:603–9.

    Article  CAS  PubMed  Google Scholar 

  28. Ellingsen E, Morath S, Flo T, Schromm A, Hartung T, Thiemermann C, et al. Induction of cytokine production in human T cells and monocytes by highly purified lipoteichoic acid: involvement of toll-like receptors and CD14. Med Sci Monit. 2002;8(5):149–56.

    Google Scholar 

  29. Kim H, Jung BJ, Kim JY, Chung DK. Differential effects of low and high doses of lipoteichoic acid on lipopolysaccharide-induced interleukin-6 production. Inflamm Res. 2014;63(6):419–28.

    Article  CAS  PubMed  Google Scholar 

  30. Hodge DR, Hurt EM, Farrar WL. The role of IL-6 and STAT3 in inflammation and cancer. Eur J Cancer. 2005;41(16):2502–12.

    Article  CAS  PubMed  Google Scholar 

  31. Koff JL, Ramachandiran S, Bernal-Mizrachi L. A time to kill: targeting apoptosis in cancer. Int J Mol Sci. 2015;16(2):2942–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Collette JR, Zhou H, Lorenz MC. Candida albicans Suppresses nitric oxide generation from macrophages via a secreted molecule. PLoS One. 2014;9(4):e96203.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Viard-Leveugle I, Gaide O, Jankovic D, Feldmeyer L, Kerl K, Pickard C, et al. TNF-α and IFN-γ are potential inducers of Fas-mediated keratinocyte apoptosis through activation of inducible nitric oxide synthase in toxic epidermal necrolysis. J Invest Dermatol. 2013;133(2):489–98.

    Article  CAS  PubMed  Google Scholar 

  34. Dias KC, Barbugli PA, Vergani CE. Influence of different buffers (HEPES/MOPS) on keratinocyte cell viability and microbial growth. J Microbiol Methods. 2016;125:40–2.

    Article  CAS  Google Scholar 

  35. Pereira CA, Romeiro RL, Costa AC, Machado AK, Junqueira JC, Jorge AO. Susceptibility of Candida albicans, Staphylococcus aureus, and Streptococcus mutans biofilms to photodynamic inactivation: an in vitro study. Lasers Med Sci. 2011;26(3):341–8.

    Article  PubMed  Google Scholar 

  36. Castilho RM, Squarize CH, LeelahavanichkμL K, Zheng Y, Bugge T, Gutkind JS. Rac1 is required for epithelial stem cell function during dermal and oral mucosal wound healing but not for tissue homeostasis in mice. PLoS One. 2010;5(5):e10503.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976;72:248–54.

    Article  CAS  PubMed  Google Scholar 

  38. Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotocity assays. J Immunol Methods. 1983;65(1–2):55–63.

    Article  CAS  PubMed  Google Scholar 

  39. Leist M, Nicotera P. The shape of cell death. Biochem Biophys Res Commun. 1997;236:1–9.

    Article  CAS  PubMed  Google Scholar 

  40. Hu LF, Lu M, Wu ZY, Wong PT, Bian JS. Hydrogen sulfide inhibits rotenone induced apoptosis via preservation of mitochondrial function. Mol Pharmacol. 2009;75(1):27–34.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This work was supported the CNPq (the Brazilian National Council for Scientific and Technological Development) (process number 163551/2012–0, 400,658/2012–7 and 471,540/2012–9). The study sponsor (CNPq) had no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; and in the decision to submit the paper for publication.

Availability of data and materials

The datasets used and/or analysed during the current study available from the corresponding author on reasonable request.

Authors’ contributions

Conceived and designed the experiments: CEV KCD PAB AIM. Performed the experiments: KCD PAB FP VBL LAP. Analyzed the data: CEV KCD PAB AIM. Contributed reagents/materials/analysis tools: CEV AIM PAB. Wrote the paper: KCD PAB FP VBL LAP AIM CEV. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.

Consent for publication

Not applicable

Ethics approval and consent to participate

Not applicable

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Carlos Eduardo Vergani.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

de Carvalho Dias, K., Barbugli, P.A., de Patto, F. et al. Soluble factors from biofilm of Candida albicans and Staphylococcus aureus promote cell death and inflammatory response. BMC Microbiol 17, 146 (2017). https://0-doi-org.brum.beds.ac.uk/10.1186/s12866-017-1031-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12866-017-1031-5

Keywords