Next Article in Journal
PHI-1, an Endogenous Inhibitor Protein for Protein Phosphatase-1 and a Pan-Cancer Marker, Regulates Raf-1 Proteostasis
Previous Article in Journal
Ca2+ Signaling and Src Functions in Tumor Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Protective Properties of S-layer Protein 2 from Lactobacillus crispatus 2029 against Candida albicans Infections

by
Vyacheslav M. Abramov
1,2,*,
Igor V. Kosarev
1,2,
Andrey V. Machulin
3,
Tatiana V. Priputnevich
2,
Evgenia I. Deryusheva
4,
Alexander N. Panin
1,
Irina O. Chikileva
5,
Tatiana N. Abashina
3,
Vyacheslav G. Melnikov
6,
Nataliya E. Suzina
3,
Ilia N. Nikonov
7,
Anna A. Akhmetzyanova
1,
Valentin S. Khlebnikov
8,
Vadim K. Sakulin
8,
Raisa N. Vasilenko
8,
Vladimir A. Samoilenko
3,
Alexey B. Gordeev
2,
Gennady T. Sukhikh
2,
Vladimir N. Uversky
9 and
Andrey V. Karlyshev
10
1
Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
2
Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
3
Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
4
Institute for Biological Instrumentation, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
5
Laboratory of Cell Immunity, Blokhin National Research Center of Oncology, Ministry of Health RF, 115478 Moscow, Russia
6
Gabrichevsky Research Institute for Epidemiology and Microbiology, 125212 Moscow, Russia
7
Federal State Educational Institution of Higher Professional Education Moscow State Academy of Veterinary Medicine and Biotechnology Named after K.I. Skryabin, 109472 Moscow, Russia
8
Institute of Immunological Engineering, 142380 Lyubuchany, Russia
9
Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
10
Department of Biomolecular Sciences, School of Life Sciences, Chemistry and Pharmacy, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston upon Thames KT1 2EE, UK
*
Author to whom correspondence should be addressed.
Biomolecules 2023, 13(12), 1740; https://doi.org/10.3390/biom13121740
Submission received: 10 October 2023 / Revised: 27 November 2023 / Accepted: 3 December 2023 / Published: 4 December 2023

Abstract

:
Previously, the protective role of the S-layer protein 2 (Slp2) of the vaginal Lactobacillus crispatus 2029 (LC2029) strain against foodborne pathogens Campylobacter jejuni, Salmonella enterica serovar Enteritidis, and Escherichia coli O157:H was demonstrated. We demonstrate the new roles of the Slp2-positive LC2029 strain and soluble Slp2 against C. albicans infections. We show that LC2029 bacteria can adhere to the surface of the cervical epithelial HeLa cells, prevent their contact with C. albicans, and block yeast transition to a pathogenic hyphal form. Surface-bound Slp2 provides the ability for LC2029 to co-aggregate with various C. albicans strains, including clinical isolates. C. albicans-induced necrotizing epithelial damage is reduced by colonization with the Slp2-positive LC2029 strain. Slp2 inhibits the adhesion of various strains of C. albicans to different human epithelial cells, blocks yeast transition to a pathogenic hyphal form, and prevents the colonization and pathogenic infiltration of mucosal barriers. Only Slp2 and LC2029 bacteria stimulate the production of protective human β-defensin 3 in various epithelial cells. These findings support the anti-Candida albicans potential of the probiotic LC2029 strain and Slp2 and form the basis for further research on their ability to prevent and manage invasive Candida infections.

1. Introduction

Candida albicans is a member of the normal human microbiome. In most individuals, C. albicans is a lifelong, harmless commensal. Under certain circumstances, C. albicans turns into a fungal pathobiont capable of causing epithelial cell damage. C. albicans is a major fungal pathogen in humans and is the most pathogenic species of the Candida genus [1]. C. albicans exists in the form of yeast (blastopore) or hyphae, i.e., single, oval-shaped cells or elongated, tubular, and branching filaments that form the mycelium [2,3]. Its pathogenicity is directly correlated with adhesive properties [4], large metabolic adaptability to adverse factors [5], and the ability by invasive (hyphal) form to secrete a cytolytic peptide toxin—candidalysin [6]. This toxin is critical for fungal infections and is a key driver of host cell activation, neutrophil recruitment, and Type 17 immunity [6].
In humans, C. albicans can cause vaginal, oral, or intestinal candidiasis and life-threatening systemic infections [7]. Vulvovaginal candidiasis (VVC) is the most widespread. VVC, caused by the Candida species, affects an estimated 70–75% of women at least once during their lives. In most countries, C. albicans is the major cause of VVC. C. albicans was reported to account for 85–95% of yeast strains isolated from the vagina [8]. In addition, 5–8% of women have recurrent vulvovaginal candidiasis (RVVC), which is defined by having four or more episodes every year [9]. VVC and RVVC remain a serious problem in reproductive-age women [10]. The risk factors for VVC include hormonal changes, an immunocompromised state, pregnancy, and antibiotic exposure [8].
Oral candidiasis, known as thrush or oropharyngeal candidiasis (OPC), is a common opportunistic infection of the oral cavity, mainly caused by an overgrowth of C. albicans [11,12,13,14]. OPC is common among the elderly (especially those who wear dentures), infants, and people with weakened immune systems or those receiving long-term antibiotic treatment. It may also be associated with systemic diseases, such as diabetes mellitus. In individuals with a defective immune response, the pathogen can spread to the pharynx and the esophagus [15].
Intestinal candidiasis (IC) is a common opportunistic infection of the gastrointestinal tract (GIT) [15,16]. The GIT is the largest source of candidemia in violation of the systemic or local immune functions of the mucous membrane [17,18,19]. The GIT is an important niche in the lifecycle of C. albicans because this organism does not have a significant environmental reservoir. C. albicans is almost always found associated with humans or other mammals, typically in the GIT, including the oral cavity, genitourinary tract, or skin [20]. In the GIT, C. albicans encounters and responds to varying features of the physical environment such as pH, oxygen levels, and nutrient levels [21]. C. albicans also responds to the secretions produced in the GIT, such as bile [22]. Thus, C. albicans is well adapted for growth in the GIT. Oral treatment with broad-spectrum antibiotics destroys the local intestinal microbiota, which provokes the reproduction of intestinal C. albicans and negatively affects the immunological system of patients, thereby contributing to the occurrence of viral infection [23]. Violation of the normal intestinal microbiota leads to the development of low-level inflammation, which contributes to increased fungal adhesion and colonization, with the latter leading to the subsequent development of inflammation and stimulation of the production of proinflammatory cytokines IL-23 and IL-17 in the GIT [16,24]. A vicious circle occurs in the intestine: low-level inflammation stimulates increased adhesion of C. albicans on enterocytes, whereas the increased adhesion and colonization of C. albicans stimulate inflammation [24]. According to a number of studies, inflammation destroys the intestinal barrier, which creates conditions for the penetration of C. albicans into the bloodstream [25,26,27]. This creates conditions for the development of systemic candidiasis.
Systemic or disseminated severe candidiasis develops mainly in patients with HIV/AIDS, as well as in patients with diabetes mellitus, and is characterized by high morbidity and mortality rates [28]. Deaths per annum from systemic fungal infections are greater than the global mortality due to malaria or breast cancer and are similar to deaths due to tuberculosis or HIV [29].
Four main classes of antifungal drugs are used for the treatment of candidiasis: azoles, polyenes, allylamines, and echinocandins. Antifungal agents, such as nystatin, amphotericin B, clotrimazole, and fluconazole, are first-line therapies and remain the main strategy for the prevention and treatment of infections, such as vaginal, oral, or intestinal candidiasis, and life-threatening systemic infections [30,31]. However, oral azoles exhibit numerous side effects [32] caused by drug toxicity and the emergence of drug-resistant strains [33]. Treatment with these drugs can lead to growth in Candida spp. resistance and to the development of RVVC. This disease is a common cause of significant morbidity in women in all strata of society, affecting millions of women worldwide [10,30]. Investigations presented in [34] indicated that fluconazole caused dysbiosis of both mycobiota and bacteriobiota. Among the mycobiota, Candida spp. decreased but other fungi, such as Aspergillus, Wallemia, and Epicoccum, increased. Among the bacteriobiota, Bacteroides, Allobaculum, Clostridium, Desulfovibrio, and Lactobacillus spp. decreased, while Anaerostipes, Coprococcus, and Streptococcus increased. During recent decades, the high rates of morbidity and mortality caused by fungal infections are associated with the current limited antifungal arsenal and the high toxicity of the drugs [35,36]. These facts emphasize the relevance of developing innovative therapeutic strategies. In this regard, probiotics may represent a promising alternative approach. Several studies have reported the anti-pathogenic potential of probiotic bacteria for the prevention and/or the treatment of OPC [32,37,38,39,40,41] and IC [42,43,44,45,46,47,48,49].
Healthy vaginal microbiota is typically dominated by the Lactobacillus species, such as L. crispatus, L. jensenii, and L. gasseri [50,51]. L. crispatus is prevailing over the other hydrogen peroxide-producing Lactobacillus species [52].
A type I microbiota with the dominance of the L. crispatus species is important for maintaining a healthy birth tract [50,51,52,53]. L. crispatus species is a microbial biomarker of a healthy vaginal microbiota [54,55]. L. crispatus is involved in maintaining the homeostasis of the vaginal environment, where it supports the immune barriers of the vagina without causing inflammation, while at the same time reducing pro-inflammatory cytokines [56,57] that usually increase during bacterial vaginosis, BV [58,59], and vulvovaginal candidiasis, VVC [10]. Strains of L. crispatus produce various bacteriocins. In eight genomes of vaginal L. crispatus strains isolated from women, the genetic determinants of bacteriocins were characterized by studies in silico [60,61]. Genetic determinants of bacteriocins were characterized in the genomes of seven strains of L. crispatus isolated from chicken feces [60]. L. crispatus 2029 produces the class III bacteriocin Helveticin-M [62] and secretes high levels of hydrogen peroxide—120 mg/L [56]. Comparative analysis of the genome of L. crispatus isolated from various ecological niches shows the ecological adaptation of this species to the environment of the human vagina and may still undergo adaptation to enhance its competitiveness for niche colonization [63,64].
Despite the fact that L. crispatus species shows ecological adaptation to the human reproductive system, it also effectively implements probiotic and postbiotic properties in the digestive system of humans and animals. Human strain L. crispatus 2029 secretes S-layer protein that enhances growth, differentiation, VEGF production, and barrier functions in intestinal epithelial cell line Caco-2 [65]. L. crispatus JCM5810, isolated from chicken feces, reduces Campylobacter jejuni colonization in chicken intestines [66]. L. crispatus ZJ001, isolated from pig intestines produced S-layer protein that inhibited the adhesion of S. typhimurium and E. coli O157:H7 to HeLa cells used in the experiment as an in vitro cellular biomodel [67].
It is well known that a crucial prerequisite for the C. albicans pathogenicity is the initial adhesion to host cells [68] leading to genitourinary, oropharyngeal, or intestinal candidiasis and life-threatening systemic infections [4,69,70]. Studies on the anti-adhesive effects of the culture supernatants of Lactobacillus species against Candida cells have been previously reported [71,72], and the supernatants capable of inhibiting C. albicans hyphae formation by modulating Lactobacillus gene expression were studied [71,72]. The culture supernatants of L. crispatus JCM 1185 inhibited C. albicans adhesion to HeLa cells and C. albicans biofilm formation by downregulating biofilm formation-related genes [73]. We have previously demonstrated that soluble S-layer protein 2 (Slp2) reduces the adhesion of foodborne pathogens Campylobacter jejuni, Salmonella enterica serovar Enteritidis, and Escherichia coli O157:H to the intestinal Caco-2 cells [65]. The anti-adhesive properties of Slp2 secreted into the culture medium by the LC2029 strain against C. albicans remained mostly unexplored. The goal of the present study was to elucidate the anti-adhesive effects of Slp2 and its role in the protective properties of the LC2029 strain against C. albicans infections.

2. Materials and Methods

2.1. Bacterial Strains and Growth Conditions

The L. crispatus 2029 (LC2029) Slp2-positive strain was originally isolated from a vaginal smear of a healthy woman of reproductive age [56]. Genomic sequencing of this strain revealed that the slp2 gene was responsible for Slp2 synthesis [62]. The strain was deposited at the All-Russian Collection of Microorganisms at the G. K. Skryabin Institute of Biochemistry and Physiology of Microorganisms under registration number VKM B-2727D. The L. crispatus (LC1385) Slp2-negative strain was originally isolated from a vaginal smear of a woman of reproductive age with clinical diagnosis: RVVC, bacterial vaginosis (BV) in anamnesis. LC2029 and LC1385 Slp2-negative strains were grown in Man-Rogosa–Sharpe (MRS) broth or an agar containing MRS plates (Himedia, India) at 37 °C in 5% CO2 or anaerobically for 48 h. C. albicans strains were cultured to the third generation on a Sabouraud dextrose (SD) agar (HiMedia, India) at 37 °C for 72 h and harvested. After resuspension in phosphate buffered saline (PBS), the cells were counted and adjusted to a final concentration of 1 × 105 CFU/mL. A complete list of the microorganisms used in this study and their growth conditions is provided in Table 1.

2.2. In Vitro Models of the Human Epithelial Cells

Cervical HeLa epithelial cells were cultured on Dulbecco’s Modified Eagle Medium (DMEM) with 10% heat-inactivated fetal bovine serum (FBC), 1% nonessential amino acid, and 1% glutamine at 37 °C in a 5% CO2 in humidified atmosphere. To obtain a monolayer of immature HeLa cells (formed 70–80%), the cells were cultured for 24 h at 37 °C in a 5% CO2 humidified atmosphere. To obtain a monolayer of immature HeLa cells (formed 100%), the cells were cultured for 72 h at 37 °C in a 5% CO2 humidified atmosphere.
Vaginal Vk2/E6E7 epithelial cells (ATCC #CRL-2616, VA) were cultured in a keratinocyte serum-free medium (KSFM) (Gibco, Waltham, MA, USA) supplemented with 50 mg/mL of bovine pituitary extract (BPE) and 0.1 ng/mL of epidermal growth factor (EGF) at 37 °C with 5% CO2. Experiments were carried out in KSFM without supplements.
Buccal TR146 epithelial carcinoma cell line [74], obtained from the European Collection of Authenticated Cell Cultures (ECACC), were cultured in DMEM (Sigma-Aldrich, St. Louis, MO, USA) and supplemented with 10% FBC and 1% penicillin–streptomycin. Serum-free DMEM was used to replace the normal growth medium 24 h before and during the experimentation process.
Intestinal Caco-2 epithelial cells and colon HT-29 epithelial cells were maintained in a culture medium, DMEM, with 10% FBS, 1% nonessential amino acid, and 1% glutamine. Cells were cultured at 37 °C in a 5% CO2 humidified atmosphere.
Human umbilical vein endothelial cells (HUVECs) were obtained from six human donors. Umbilical cords (15 cm) were cut soon after birth and stored in PBS immediately. The endothelial cells were obtained from filling the lumens of umbilical veins with collagen enzyme solution.
The cells were cultured at 37 °C in a 5% CO2 atmosphere in a standard endothelial cell basal medium (EBM) (PanEco, Moscow, Russia) plus endothelial cell-growth medium (EGM) supplements (CAMBREX Bios Science, Inc., Walkersville, MD, USA) and 10% heat-inactivated FBC (Invitrogen, Waltham, MA, USA). After reaching a confluence of 80%, the cells were detached using 0.25% trypsin-EDTA (PanEco, Moscow, Russia). The cells were used by withing three to four passages.
Human lung carcinoma A-549 cells were cultured in Minimum Essential Medium with 10% FBS, 2 mmol/L L-glutamine, 1 mmol/L pyruvate, 10 mmol/L HEPES, and 50 μmol/L penicillin/streptomycin and maintained at 37 °C in a sterile humidified atmosphere of 5% CO2.
All epithelial cells used tested negative for mycoplasma.

2.3. Slp2 Production

Slp2 was extracted from the LC2029 strain by 5 M LiCl and purified by cation exchange chromatography. The purified Slp2 was examined by SDS-PAGE using 12% polyacrylamide gel and Western blotting analysis. The molecular weight of the purified soluble Slp2 was ~46 kDa [65]. Slp2 concentration in the solution was determined using a Bio-Rad Protein Assay (Bio-Rad Laboratories, Hercules, CA, USA).

2.4. Spectrophotometric Co-Aggregation Assay for the Determination of Interactions between the LC2029 Strain and C. albicans Unicellular Yeast Form

The LC2029 strain and Slp2-negative LC1385 strain were grown in MRS broth at 37 °C anaerobically for 48 h. C. albicans strains were grown at 37 °C in SD broth at 37 °C in 5% CO2 for 48 h. The cultures were harvested by centrifugation at 10,000× g for 10 min, washed twice with sterile PBS pH 7.0, and resuspended in PBS. Absorbance (A600 nm) of bacterial suspensions was adjusted to 0.60 ± 0.02 in order to standardize the concentration of bacteria (107 to 108 CFU/mL). LC2029 strain suspension or Slp2-negative LC1385 strain suspension (2 mL each) was mixed with 2 mL of C. albicans strain suspensions for at least 10 s by a vortex mixer. After 4 h incubation at 37 °C, the suspensions were measured using a spectrophotometer at OD600 (T1800; Hitachi, Tokyo, Japan).
Co-aggregation between the LC2029 strain or Slp2-negative LC1385 strain and the yeast form of C. albicans strains was determined by measuring optical density (OD) at 600 nm according to [75]. The percentage of co-aggregation was calculated by following equation:
C o - a g g r e g a t i o n ( % ) = ( ( O D t a r g e t + O D L C 2029 ) 2 · O D m i x ) ( O D t a r g e t + O D L C 2029 )
ODtarget, ODLC2029, and ODmix represent the OD measure at 600 nm of individual C. albicans strains, the LC2029 strain, and their mixture after incubation for 2 h.

2.5. Slp2 Effect on C. albicans Adhesion to Epithelial Cells of Human Biotopes

Monolayers (formed 70–80%) of immature cervical HeLa epithelial cells, vaginal Vk2/E6E7 epithelial cells, buccal TR146 epithelial cells, intestinal Caco-2 epithelial cells, and colon HT-29 epithelial, HUVECs, and lung A-549 cells were obtained by cultivation in an appropriate nutrient medium for 24–48 h at 37 °C in a 5% CO2 in humidified atmosphere.
Suspensions of C. albicans strains with 5 × 107 CFU/mL were incubated in the presence of Slp2 at concentrations of 10, 50, and 100 µg/mL in PBS for 30 min. The strains of C. albicans treated with Slp2 and the control group of C. albicans treated with PBS were then applied to the monolayers of cervical HeLa epithelial cells, vaginal Vk2/E6E7 epithelial cells, buccal TR146 epithelial cells, intestinal Caco-2 epithelial cells, and colon HT-29 epithelial, HUVECs, and lung A-549 epithelial cells. The plates were incubated for 1 h at 37 °C under 5% CO2. Cell monolayers were washed three times with sterile PBS to remove unbound C. albicans strains, fixed with methanol, stained with azure-eosin (Pan Eco, Russia), and examined under a Leica DM 4500B microscope (Leica, Richmond Hill, ON, Canada). The adhesion activity of C. albicans strains was quantified using the Leica IM modular applications system (Leica, Canada). The adhesion activity of C. albicans strains to epithelial cells was expressed as percentage of epithelial cells containing candida cells on their surface.

2.6. Quantification of C. albicans-Mediated Damage of Human Epithelial Cells

The influence of lactobacilli on C. albicans-mediated host cell damage was investigated by measuring the release of cytoplasmic human lactate dehydrogenase (LDH), as an indicator for the loss of membrane integrity and a hallmark of necrosis, according to [76,77]. LDH was quantified in the supernatant of infected epithelial call monolayers 24 h post-infection using the Cytotoxicity Detection Kit (Roche, Basel, Switzerland) according to the manufacturer’s instructions. LDH from rabbit muscle (5 mg/mL, Roche) was used to generate a standard curve for the determination of LDH concentrations. The background control level of uninfected epithelial cells was subtracted from the experimental LDH release and usually compared to 100% C. albicans single infection.

2.7. ELISA

To determine the concentration of HBD-3, the supernatants obtained from epithelial cultures stimulated with the Slp2, LC2029 strain, Slp2-negative ΔLC2029 strain (Slp2 was eliminated with 5 M lithium chloride), and Slp2-negative LC1385 strain for 12 h were collected, centrifuged to separate bacteria, and stored at 80 °C until assayed using an ELISA kit (Phoenix Pharmaceuticals, Inc., Burlingame, CA, USA) following the protocol provided by the manufacturer.

2.8. Statistical Analysis

The results were analyzed using a one-way ANOVA and represented means ± standard errors of the means (SEM). Statistically significant differences were accepted at p < 0.05.

3. Results and Discussion

3.1. Inhibitory Effects of the LC2029 Strain on the Transition of the C. albicans Yeast Form to Hyphae

Figure 1A shows the non-pathogenic yeast form of the C. albicans ATCC 10231 strain. The non-pathogenic yeast form of C. albicans is found in the normal vaginal microbiota of women [8]. Figure 1B shows a monolayer (formed by 100%) of intact immature HeLa cells used as an in vitro biomodel of vaginal dysbiosis when the content of lactobacilli in the vaginal biotope decreases until complete disappearance [10,50]. C. albicans cells in a yeast form introduced into the wells with an intact monolayer of HeLa cells quickly adhere to the surface of the intact monolayer and after 2 h of co-cultivation turn them into a pathogenic hyphal form (Figure 1C). The hyphal form of C. albicans is not removed from the surface of the HeLa cell monolayers after washing up to three times with PBS (Figure 1C).
Cells in the LC2029 strain added to wells with a monolayer of Hella cells (formed by 100%) effectively adhere to the surface of the monolayer (Figure 1D). A monolayer (formed by 100%) of intact immature HeLa cells with cells of the LC2029 strain adhered to the monolayer surface and was used by us as an in vitro biomodel of normal vaginal microbiota [50]. Figure 1E shows a monolayer of Hela cells formed by 70–80%. In the field of view, LC2029 cells adhered to the surface of HeLa cells in suspension. C. albicans cells in yeast form, introduced into the nutrient medium after 30 min, are subjected to co-aggregation with LC2029 cells in suspension (ratio of cells of LC2029 strain/C. albicans cells in yeast form was 200/1). After 4 h of joint cultivation, the co-aggregates of LC2029 cells with the yeast form of C. albicans cells in suspension are easily removed from the well by washing three times with PBS. In the field of view, only single Candida cells in yeast form are visible, associated with LC2029 cells protecting the monolayer of HeLa cells (Figure 1F). There are no C. albicans cells in the pathogenic hyphal form in the field of vision (Figure 1F). The LC2029 strain effectively inhibits the transition of the non-pathogenic yeast form of C. albicans strain 10231 into the pathogenic hyphal form (Figure 1F). The obtained results indicate that Slp2 fixed on the surface of LC2029 cells is responsible for the co-aggregation of LC2029 cells with C. albicans cells and for the ability of LC2029 cells to inhibit the transition of the non-pathogenic yeast form of C. albicans into the pathogenic hyphal form. A type I microbiota with the dominance of the L. crispatus species is important for maintaining a healthy birth tract [50]. The vaginal Slp-positive strain CTV-05 of L. crispatus successfully colonizes the cervicovaginal biotope [78]. The vaginal probiotic L. crispatus exhibits strong antifungal effects against VVC in a rat model [79].

3.2. LF2029 Co-Aggregation with Unicellular Yeast Form of Different C. albicans Strains

Comparative studies of LC2029 strain and Slp2-negative LC1385 strain сo-aggregative abilities with unicellular yeast form of different C. albicans strains have been conducted (Table 2). The LC2029 strain is a strong co-aggregator of unicellular yeast form of different C. albicans strains. LC2029 actively co-aggregated with different C. albicans strains, including clinical isolates (MD IIE Ca-CI-8, MD IIE Ca-CI-126, MD IIE Ca-CI-98, MD IIE Ca-CI-154). The percentage of co-aggregation ranged from 74.7 ± 4.3% to 84.1 ± 6.5%. The co-aggregation activity of the Slp2-negative LC1385 strain with unicellular yeast form of different C. albicans strains was very low. The co-aggregation abilities are characteristic attributes of some probiotic strains belonging to the Lactobacillaceae family. Co-aggregation is one of the mechanisms exerted by probiotics to create a competitive micro-environment around the pathogen and limit its adhesion on the host epithelial cells. Co-aggregation of C. albicans and lactobacilli may be important in the vagina, especially to reduce the adhesion of the fungus to the vaginal mucosa and to restore the vaginal microbiome by the production of favorable metabolites [80] and the modulation of cytokine expression [53]. Due to these properties of probiotic lactobacilli, the host organism can avoid colonization of the oral cavity, intestines, reproductive tract, and other organs by fungal pathogens [4].

3.3. Slp2 Effect on C. albicans Adhesion to the Epithelial Cells of the Human Cervicovaginal Biotope

C. albicans is an opportunistic pathogen with a significant ability to interact with epithelial cells of different human biotopes in terms of the adherence, invasion, and induction of cell damage [16].
Slp2 isolated from vaginal lactobacilli strain LC2029 very efficiently inhibited the adhesion of C. albicans yeast form to human cervical and vaginal epithelial cells (Table 3 and Table 4). Slp2 was equally effective in inhibiting the adhesion of C. albicans strains from the collection and clinical isolates, including the Ca-CI-8 strain isolated from a patient with recurrent candidiasis.
Necrosis and inflammation of the cervical epithelium induced by C. albicans increase the ability of the human papilloma virus (HPV) to form a malignant epithelial tumor [81]. Attachment of C. albicans to epithelial cells is a prerequisite for the colonization and pathogenic infiltration of mucosal barriers [82].
Chemical strategies to reduce physical interaction between pathogenic fungi and host cells show promising results. The synthesis of a multivalent glycoconjugate in which an inhibitor of C. albicans adhesion is chemically coupled to a linear peptoid scaffold was described.
Fungal adherence to buccal epithelial cells was reduced in vitro following treatment with glycoconjugate formulation, and investigations to elucidate the precise mechanism of action are currently ongoing [83]. These studies confirm the importance of the anti-adhesive properties of soluble Slp2 of LC2029 in relation to C. albicans and the expediency of its use for the prevention of VVC.

3.4. Slp2 Effect on C. albicans the Adhesion to Epithelial Cells of the Human Oropharyngeal Biotope

Slp2 very efficiently inhibited the adhesion of C. albicans yeast form to human buccal TR146 epithelial cells (Table 5). Slp2 was equally effective in inhibiting the adhesion of C. albicans strains from the collection and clinical isolates, including the Ca-CI-126 strain isolated from a patient with OPC. C. albicans is a frequent component of oral ecology.
In immunocompromised patients, C. albicans can cause a multitude of disease manifestations ranging from mild oral disease to disseminated candidiasis. Diagnosis and treatment of disease caused by C. albicans are especially important in HIV/AIDS patients who, despite the advent of antiretroviral therapy (ART), continue to suffer significant Candida-associated morbidity [84,85].

3.5. Slp2 Effect on C. albicans Adhesion to the Epithelial Cells of the Human Intestinal Biotope

Slp2 inhibited the adhesion of C. albicans yeast form to human intestinal Caco-2 cells and HT-29 cells (Table 6 and Table 7). Slp2 was equally effective in inhibiting the adhesion of C. albicans strains from the collection and clinical isolates, including Ca-CI-98 strain isolated from a patient with IC.
In healthy individuals C. albicans is predominantly found as part of the gastrointestinal microbiome. With IC, C. albicans turns into a fungal pathobiont that is able to cause epithelial cell necrosis and is immune activation and intestinal inflammation [23,86]. Intestinal inflammation is known to destroy the intestinal barrier [87]. This creates conditions for the entry of C. albicans into the blood and the development of systemic candidiasis [2,88]. We have previously shown that the Slp2 and LC2029 strain inhibited proinflammatory cytokine Il-8 production in Caco-2 and HT-29 cells induced by MALP-2 (agonist of TLR2) and increased the production of anti-inflammatory cytokine Il-6 [57]. Slp2 inhibited the production of CXCL1 and RANTES by Caco-2 cells during differentiation and maturation [57]. Slp2 stimulated VEGF production, decreased paracellular permeability, and increased transepithelial electrical resistance, strengthening barrier function of Caco-2 cell monolayers [65]. The results of these studies indicate the prospects of using the Slp2 and LC2029 strain for the prevention of systemic or disseminated severe candidiasis.

3.6. Slp2 Effect on C. albicans Adhesion to HUVECs and Lung A-549 Epithelial Cells

During systemic or disseminated severe candidiasis in susceptible individuals, C. albicans gains access to the bloodstream and various internal organs, including the lungs [2]. The intestine–lung axis functions in the human body [89]. During systemic candidiasis, C. albicans cells can spread to the lungs, which potentially differ in the availability of nutrients compared to the digestive or reproductive systems [2]. As an in vitro biomodel of systemic candidiasis, we used HUVECs and lung epithelial A-549 cells to study the ability of Slp2 to inhibit the adhesion of various C. albicans strains to these cells. Slp2 inhibited the adhesion of C. albicans yeast form to HUVECs and A-549 cells (Table 8 and Table 9). Slp2 was equally effective in inhibiting the adhesion of C. albicans strains from the collection and clinical isolates, including the Ca-CI-154 strain clinical bloodstream isolate from a patient with systemic candidiasis.
The use of molecular typing methods to compare strains isolated from a patient’s blood with strains isolated from other body sites has shown that C. albicans blood isolates are often identical to rectal isolates [18]. These findings support the model that commensal organisms residing in the GIT can escape from this niche and reach the bloodstream. The entry of C. albicans strains into the blood occurs after the destruction of the intestinal barrier [87].
The ability of Slp2 to inhibit the adhesion of C. albicans strains to HUVECs and lung A-549 epithelial cells suggests a possible application of this soluble protein for the complex treatment of systemic candidiasis.

3.7. Reducing C. albicans-Induced Necrotizing Epithelial Damage by Colonization with the LC 2029 Strain

The necrotizing potential of producing the candidalysin C. albicans ATCC 10231 strain against different human epithelial cells was assessed by an increase in epithelial cytosolic LDH secreted into the culture medium. The results of the studies are shown in Table 10. The Slp2-negative LC1385 strain and LC2029 strain do not produce LDH and are non-toxic for HUVECs and epithelial cells from a cervicovaginal biotope, including HeLa cells and Vk2/E6E7 vaginal epithelial cells from an oropharyngeal biotope, buccal TR146 epithelial cells from an intestinal biotope, Caco-2 cells and HT-29 from a lung biotope, and A-549 cells. Colonization with the Slp2-negative LC1385 strain before C. albicans infection had no effect on Candida-induced damage (Table 10). Colonization with the LC 2029 strain before C. albicans infection reduced the C. albicans-induced necrotic cytotoxicity of epithelial cells. It is known that candidalysin is secreted by C. albicans cells that have passed into the pathogenic hyphal form and adhered to the surface of epithelial cells [90]. Secreted candidalysin is readily assembled into polymers in a solution and binds to the plasma membrane of the epithelial cell, forms membrane pores, and causes its necrotic lesion [6,91]. The plasma membrane is central for homeostatic maintenance in mammalian cells [77,92]. Necrotic cell death induces inflammatory responses and is closely associated with inflammatory diseases [93]. The results obtained indicate the ability of the LC2029 strain, but not the Slp2-negative LC1385 strain, to inhibit the secretion of candidalysin by the C. albicans ATCC 10231 strain and prevent necrotic damage to epithelial cells of various human biotopes.

3.8. Slp2 and LC2029 Strain Stimulate HBD-3 Production in Epithelial Cells of Various Biotopes

Slp2 and LC2029 bacteria, but not Slp2-negative ΔLC2029 bacteria (Slp2 was eliminated with 5 M lithium chloride) and Slp2-negative LC1385 bacteria, stimulate the production of protective antimicrobial peptide (AMP) HBD-3 in epithelial cells belonging to various human biotopes (Table 11). It is known that β-defensins induce cationic peptides produced by epithelial cells that have been proposed to be an important component of immune function on mucosal surfaces [94]. It is known that HBD-3 significantly reduces C. albicans viability and growth [53] and serves as an innate defense against Candida invasion on human mucosal surfaces.

4. Conclusions

The adhesion of C. albicans to the mucosal surfaces is the basis of its invasion into the body, which occurs already in the first minutes of the pathogen’s interaction with the epithelial cells of the vagina, oral cavity, or intestines. From this moment on, vaginal, oropharyngeal, or intestinal candidiasis develops in the macroorganism. Using cervical epithelial HeLa cells as a model for the in vitro cervical biotope, it has been shown that Slp2 fixed on the surface of LC2029 cells is responsible for the co-aggregation of LC2029 cells with yeast form of C. albicans cells and for the ability of LC2029 cells to inhibit the transition of the non-pathogenic yeast form of C. albicans into the pathogenic hyphal form. Soluble Slp2 (postbiotics) inhibits C. albicans yeast form adhesion to HeLa cells. Using human Vk2/E6E7 vaginal epithelial cells as a model for the in vitro vaginal biotope, it has been shown that soluble Slp2 inhibits various strains of C. albicans yeast form adhesion to these cells. Using human buccal TR 146 epithelial cells as a model for the in vitro oropharyngeal biotope, it has been shown that soluble Slp2 inhibits various strains of C. albicans yeast form adhesion to these cells. Using human intestinal Caco-2 and HT-29 epithelial cells as a model for the in vitro intestinal biotope, it has been shown that soluble Slp2 inhibits various strains of C. albicans yeast form adhesion to these cells. Using human HUVECs and A549 epithelial cells as a model for the in vitro systemic candidiasis, it has been shown that soluble Slp2 inhibits various strains of C. albicans yeast form adhesion to these cells. The Slp2-positive LC2029 strain, but not the Slp2-negative LC1385 strain, protects HeLa, Vk2/E6E7, TR146, Caco-2, HT-29, HUVECs, and A549 epithelial cells from C. albicans-induced necrotizing epithelial damage. Soluble Slp2 and Slp2-positive LC2029 bacteria stimulate the production of anti-Candida protective HBD-3 in epithelial cells belonging to the various biotopes. Further research will be devoted to the study of the mechanisms associated with the ability of Slp2-positive LC2029 and soluble Slp2 to inhibit the transition of the non-pathogenic yeast form of C. albicans into the pathogenic hyphal form.

Author Contributions

Conceptualization, V.M.A., A.V.M., T.V.P., G.T.S., A.N.P. and A.V.K.; methodology, V.M.A.; validation, I.V.K., A.V.M., I.O.C., E.I.D., T.N.A., V.G.M., N.E.S., I.N.N., V.S.K., V.K.S., V.A.S., R.N.V., A.B.G., A.A.A., V.N.U. and A.V.K.; investigation, I.V.K., A.V.M., I.O.C., E.I.D., T.N.A., V.G.M., N.E.S., I.N.N., A.A.A., V.S.K., V.K.S., R.N.V., V.N.U. and V.A.S.; data curation, V.M.A. and I.V.K.; writing—original draft preparation, V.M.A. and I.V.K.; writing—review and editing, A.V.M., E.I.D., T.V.P., V.N.U. and A.V.K.; supervision, V.M.A., V.N.U. and A.V.K.; project administration, V.M.A. and A.V.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Government of the Russian Federation (Agreement No. 075-15-2022-1124; dated 1 July 2022).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Richardson, J.P. Candida albicans: A Major Fungal Pathogen of Humans. Pathogens 2022, 11, 459. [Google Scholar] [CrossRef] [PubMed]
  2. Mayer, F.L.; Wilson, D.; Hube, B. Candida albicans Pathogenicity Mechanisms. Virulence 2013, 4, 119–128. [Google Scholar] [CrossRef] [PubMed]
  3. Calderone, R.A.; Fonzi, W.A. Virulence Factors of Candida albicans. Trends Microbiol. 2001, 9, 327–335. [Google Scholar] [CrossRef] [PubMed]
  4. de Groot, P.W.J.; Bader, O.; de Boer, A.D.; Weig, M.; Chauhan, N. Adhesins in Human Fungal Pathogens: Glue with Plenty of Stick. Eukaryot. Cell 2013, 12, 470–481. [Google Scholar] [CrossRef] [PubMed]
  5. Lorenz, M.C.; Bender, J.A.; Fink, G.R. Transcriptional Response of Candida Albicans upon Internalization by Macrophages. Eukaryot. Cell 2004, 3, 1076–1087. [Google Scholar] [CrossRef] [PubMed]
  6. Naglik, J.R.; Gaffen, S.L.; Hube, B. Candidalysin: Discovery and Function in Candida albicans Infections. Curr. Opin. Microbiol. 2019, 52, 100–109. [Google Scholar] [CrossRef]
  7. Calderone, R.A.; Clancy, C.J. Candida and Candidiasis, 2nd ed.; ASM Press: Washington, DC, USA, 2012; ISBN 9781555815394. [Google Scholar]
  8. Sobel, J.D. Vulvovaginal Candidosis. Lancet 2007, 369, 1961–1971. [Google Scholar] [CrossRef]
  9. Sobel, J.D. Pathogenesis of Candida Vulvovaginitis. Curr. Top. Med. Mycol. 1989, 3, 86–108. [Google Scholar] [CrossRef]
  10. Sobel, J.D. Recurrent Vulvovaginal Candidiasis. Am. J. Obstet. Gynecol. 2016, 214, 15–21. [Google Scholar] [CrossRef]
  11. Akpan, A.; Morgan, R. Oral Candidiasis. Postgrad. Med. J. 2002, 78, 455–459. [Google Scholar] [CrossRef]
  12. Naglik, J.R.; Moyes, D. Epithelial Cell Innate Response to Candida albicans. Adv. Dent. Res. 2011, 23, 50–55. [Google Scholar] [CrossRef] [PubMed]
  13. Melkoumov, A.; Goupil, M.; Louhichi, F.; Raymond, M.; de Repentigny, L.; Leclair, G. Nystatin Nanosizing Enhances in Vitro and in Vivo Antifungal Activity against Candida albicans. J. Antimicrob. Chemother. 2013, 68, 2099–2105. [Google Scholar] [CrossRef] [PubMed]
  14. Kenno, S.; Perito, S.; Mosci, P.; Vecchiarelli, A.; Monari, C. Autophagy and Reactive Oxygen Species Are Involved in Neutrophil Extracellular Traps Release Induced by C. Albicans Morphotypes. Front. Microbiol. 2016, 7, 879. [Google Scholar] [CrossRef] [PubMed]
  15. Kumamoto, C.A. Inflammation and Gastrointestinal Candida Colonization. Curr. Opin. Microbiol. 2011, 14, 386–391. [Google Scholar] [CrossRef] [PubMed]
  16. Kumamoto, C.A.; Gresnigt, M.S.; Hube, B. The Gut, the Bad and the Harmless: Candida albicans as a Commensal and Opportunistic Pathogen in the Intestine. Curr. Opin. Microbiol. 2020, 56, 7–15. [Google Scholar] [CrossRef] [PubMed]
  17. Li, X.; Kolltveit, K.M.; Tronstad, L.; Olsen, I. Systemic Diseases Caused by Oral Infection. Clin. Microbiol. Rev. 2000, 13, 547–558. [Google Scholar] [CrossRef]
  18. Miranda, L.N.; van der Heijden, I.M.; Costa, S.F.; Sousa, A.P.I.; Sienra, R.A.; Gobara, S.; Santos, C.R.; Lobo, R.D.; Pessoa, V.P.; Levin, A.S. Candida Colonisation as a Source for Candidaemia. J. Hosp. Infect. 2009, 72, 9–16. [Google Scholar] [CrossRef] [PubMed]
  19. Schulze, J.; Sonnenborn, U. Yeasts in the Gut: From Commensals to Infectious Agents. Dtsch. Arztebl. Int. 2009, 106, 837–842. [Google Scholar] [CrossRef]
  20. Soll, D.R.; Galask, R.; Schmid, J.; Hanna, C.; Mac, K.; Morrow, B. Genetic Dissimilarity of Commensal Strains of Candida Spp. Carried in Different Anatomical Locations of the Same Healthy Women. J. Clin. Microbiol. 1991, 29, 1702–1710. [Google Scholar] [CrossRef]
  21. Rosenbach, A.; Dignard, D.; Pierce, J.V.; Whiteway, M.; Kumamoto, C.A. Adaptations of Candida albicans for Growth in the Mammalian Intestinal Tract. Eukaryot. Cell 2010, 9, 1075–1086. [Google Scholar] [CrossRef]
  22. Vu, B.; Essmann, M.; Larsen, B. Sodium Choleate (NaCho) Effects on Candida albicans: Implications for Its Role as a Gastrointestinal Tract Inhabitant. Mycopathologia 2010, 169, 183–191. [Google Scholar] [CrossRef] [PubMed]
  23. Ruiz-Sánchez, D.; Calderón-Romero, L.; Sánchez-Vega, J.T.; Tay, J. Intestinal Candidiasis. A Clinical Report and Comments about This Opportunistic Pathology. Mycopathologia 2002, 156, 9–11. [Google Scholar] [CrossRef] [PubMed]
  24. Allert, S.; Förster, T.M.; Svensson, C.-M.; Richardson, J.P.; Pawlik, T.; Hebecker, B.; Rudolphi, S.; Juraschitz, M.; Schaller, M.; Blagojevic, M.; et al. Candida albicans-Induced Epithelial Damage Mediates Translocation through Intestinal Barriers. mBio 2018, 9, e00915-18. [Google Scholar] [CrossRef] [PubMed]
  25. Kadosh, D.; Najvar, L.K.; Bocanegra, R.; Olivo, M.; Kirkpatrick, W.R.; Wiederhold, N.P.; Patterson, T.F. Effect of Antifungal Treatment in a Diet-Based Murine Model of Disseminated Candidiasis Acquired via the Gastrointestinal Tract. Antimicrob. Agents Chemother. 2016, 60, 6703–6708. [Google Scholar] [CrossRef] [PubMed]
  26. Koh, A.Y.; Köhler, J.R.; Coggshall, K.T.; Van Rooijen, N.; Pier, G.B. Mucosal Damage and Neutropenia Are Required for Candida albicans Dissemination. PLoS Pathog. 2008, 4, e35. [Google Scholar] [CrossRef] [PubMed]
  27. Vautier, S.; Drummond, R.A.; Redelinghuys, P.; Murray, G.I.; MacCallum, D.M.; Brown, G.D. Dectin-1 Is Not Required for Controlling Candida albicans Colonization of the Gastrointestinal Tract. Infect. Immun. 2012, 80, 4216–4222. [Google Scholar] [CrossRef]
  28. Gudlaugsson, O.; Gillespie, S.; Lee, K.; Vande Berg, J.; Hu, J.; Messer, S.; Herwaldt, L.; Pfaller, M.; Diekema, D. Attributable Mortality of Nosocomial Candidemia, Revisited. Clin. Infect. Dis. 2003, 37, 1172–1177. [Google Scholar] [CrossRef]
  29. Brown, G.D.; Denning, D.W.; Levitz, S.M. Tackling Human Fungal Infections. Science 2012, 336, 647. [Google Scholar] [CrossRef]
  30. Sobel, J.D. Factors Involved in Patient Choice of Oral or Vaginal Treatment for Vulvovaginal Candidiasis. Patient Prefer. Adherence 2013, 8, 31–34. [Google Scholar] [CrossRef]
  31. Garcia-Cuesta, C.; Sarrion-Pérez, M.-G.; Bagán, J.V. Current Treatment of Oral Candidiasis: A Literature Review. J. Clin. Exp. Dent. 2014, 6, e576–e582. [Google Scholar] [CrossRef]
  32. Ohshima, T.; Kojima, Y.; Seneviratne, C.J.; Maeda, N. Therapeutic Application of Synbiotics, a Fusion of Probiotics and Prebiotics, and Biogenics as a New Concept for Oral Candida Infections: A Mini Review. Front. Microbiol. 2016, 7, 10. [Google Scholar] [CrossRef] [PubMed]
  33. Sardi, J.C.O.; Scorzoni, L.; Bernardi, T.; Fusco-Almeida, A.M.; Mendes Giannini, M.J.S. Candida Species: Current Epidemiology, Pathogenicity, Biofilm Formation, Natural Antifungal Products and New Therapeutic Options. J. Med. Microbiol. 2013, 62, 10–24. [Google Scholar] [CrossRef] [PubMed]
  34. Wheeler, M.L.; Limon, J.J.; Bar, A.S.; Leal, C.A.; Gargus, M.; Tang, J.; Brown, J.; Funari, V.A.; Wang, H.L.; Crother, T.R.; et al. Immunological Consequences of Intestinal Fungal Dysbiosis. Cell Host Microbe 2016, 19, 865–873. [Google Scholar] [CrossRef] [PubMed]
  35. Scorzoni, L.; de Paula e Silva, A.C.A.; Marcos, C.M.; Assato, P.A.; de Melo, W.C.M.A.; de Oliveira, H.C.; Costa-Orlandi, C.B.; Mendes-Giannini, M.J.S.; Fusco-Almeida, A.M. Antifungal Therapy: New Advances in the Understanding and Treatment of Mycosis. Front. Microbiol. 2017, 8, 36. [Google Scholar] [CrossRef] [PubMed]
  36. Lockhart, S.R.; Chowdhary, A.; Gold, J.A.W. The Rapid Emergence of Antifungal-Resistant Human-Pathogenic Fungi. Nat. Rev. Microbiol. 2023, 21, 818–832. [Google Scholar] [CrossRef]
  37. Matsubara, V.H.; Bandara, H.M.H.N.; Mayer, M.P.A.; Samaranayake, L.P. Probiotics as Antifungals in Mucosal Candidiasis. Clin. Infect. Dis. 2016, 62, 1143–1153. [Google Scholar] [CrossRef] [PubMed]
  38. Hatakka, K.; Ahola, A.J.; Yli-Knuuttila, H.; Richardson, M.; Poussa, T.; Meurman, J.H.; Korpela, R. Probiotics Reduce the Prevalence of Oral Candida in the Elderly—A Randomized Controlled Trial. J. Dent. Res. 2007, 86, 125–130. [Google Scholar] [CrossRef] [PubMed]
  39. Ishijima, S.A.; Hayama, K.; Burton, J.P.; Reid, G.; Okada, M.; Matsushita, Y.; Abe, S. Effect of Streptococcus Salivarius K12 on the in Vitro Growth of Candida albicans and Its Protective Effect in an Oral Candidiasis Model. Appl. Environ. Microbiol. 2012, 78, 2190–2199. [Google Scholar] [CrossRef]
  40. Ishikawa, K.H.; Mayer, M.P.A.; Miyazima, T.Y.; Matsubara, V.H.; Silva, E.G.; Paula, C.R.; Campos, T.T.; Nakamae, A.E.M. A Multispecies Probiotic Reduces Oral Candida Colonization in Denture Wearers. J. Prosthodont. 2015, 24, 194–199. [Google Scholar] [CrossRef]
  41. Kraft-Bodi, E.; Jørgensen, M.R.; Keller, M.K.; Kragelund, C.; Twetman, S. Effect of Probiotic Bacteria on Oral Candida in Frail Elderly. J. Dent. Res. 2015, 94, 181S–186S. [Google Scholar] [CrossRef]
  42. Wagner, R.D.; Pierson, C.; Warner, T.; Dohnalek, M.; Farmer, J.; Roberts, L.; Hilty, M.; Balish, E. Biotherapeutic Effects of Probiotic Bacteria on Candidiasis in Immunodeficient Mice. Infect. Immun. 1997, 65, 4165–4172. [Google Scholar] [CrossRef] [PubMed]
  43. Cotter, G.; Doyle, S.; Kavanagh, K. Development of an Insect Model for the in Vivo Pathogenicity Testing of Yeasts. FEMS Immunol. Med. Microbiol. 2000, 27, 163–169. [Google Scholar] [CrossRef]
  44. Fuchs, B.B.; O’Brien, E.; El Khoury, J.B.; Mylonakis, E. Methods for Using Galleria Mellonella as a Model Host to Study Fungal Pathogenesis. Virulence 2010, 1, 475–482. [Google Scholar] [CrossRef] [PubMed]
  45. Junqueira, J.C.; Fuchs, B.B.; Muhammed, M.; Coleman, J.J.; Suleiman, J.M.A.H.; Vilela, S.F.G.; Costa, A.C.B.P.; Rasteiro, V.M.C.; Jorge, A.O.C.; Mylonakis, E. Oral Candida albicans Isolates from HIV-Positive Individuals Have Similar in Vitro Biofilm-Forming Ability and Pathogenicity as Invasive Candida Isolates. BMC Microbiol. 2011, 11, 247. [Google Scholar] [CrossRef] [PubMed]
  46. Rossoni, R.D.; Barbosa, J.O.; Vilela, S.F.G.; dos Santos, J.D.; de Barros, P.P.; Prata, M.C.D.A.; Anbinder, A.L.; Fuchs, B.B.; Jorge, A.O.C.; Mylonakis, E.; et al. Competitive Interactions between C. Albicans, C. Glabrata and C. Krusei during Biofilm Formation and Development of Experimental Candidiasis. PLoS ONE 2015, 10, e0131700. [Google Scholar] [CrossRef] [PubMed]
  47. Deng, K.; Chen, T.; Wu, Q.; Xin, H.; Wei, Q.; Hu, P.; Wang, X.; Wang, X.; Wei, H.; Shah, N.P. In Vitro and in Vivo Examination of Anticolonization of Pathogens by Lactobacillus Paracasei FJ861111.1. J. Dairy Sci. 2015, 98, 6759–6766. [Google Scholar] [CrossRef] [PubMed]
  48. Milner, E.; Stevens, B.; An, M.; Lam, V.; Ainsworth, M.; Dihle, P.; Stearns, J.; Dombrowski, A.; Rego, D.; Segars, K. Utilizing Probiotics for the Prevention and Treatment of Gastrointestinal Diseases. Front. Microbiol. 2021, 12, 689958. [Google Scholar] [CrossRef] [PubMed]
  49. Leão, M.V.P.; Tavares, T.A.A.; Gonçalves E Silva, C.R.; Dos Santos, S.S.F.; Junqueira, J.C.; de Oliveira, L.D.; Jorge, A.O.C. Lactobacillus Rhamnosus Intake Can Prevent the Development of Candidiasis. Clin. Oral Investig. 2018, 22, 2511–2518. [Google Scholar] [CrossRef]
  50. Ravel, J.; Gajer, P.; Abdo, Z.; Schneider, G.M.; Koenig, S.S.K.; McCulle, S.L.; Karlebach, S.; Gorle, R.; Russell, J.; Tacket, C.O.; et al. Vaginal Microbiome of Reproductive-Age Women. Proc. Natl. Acad. Sci. USA 2011, 108, 4680–4687. [Google Scholar] [CrossRef]
  51. Pavlova, S.I.; Kilic, A.O.; Kilic, S.S.; So, J.-S.; Nader-Macias, M.E.; Simoes, J.A.; Tao, L. Genetic Diversity of Vaginal Lactobacilli from Women in Different Countries Based on 16S RRNA Gene Sequences. J. Appl. Microbiol. 2002, 92, 451–459. [Google Scholar] [CrossRef]
  52. Antonio, M.A.; Hawes, S.E.; Hillier, S.L. The Identification of Vaginal Lactobacillus Species and the Demographic and Microbiologic Characteristics of Women Colonized by These Species. J. Infect. Dis. 1999, 180, 1950–1956. [Google Scholar] [CrossRef] [PubMed]
  53. Rizzo, A.; Losacco, A.; Carratelli, C.R. Lactobacillus Crispatus Modulates Epithelial Cell Defense against Candida albicans through Toll-like Receptors 2 and 4, Interleukin 8 and Human β-Defensins 2 and 3. Immunol. Lett. 2013, 156, 102–109. [Google Scholar] [CrossRef] [PubMed]
  54. Lepargneur, J.-P. Lactobacillus Crispatus as Biomarker of the Healthy Vaginal Tract. Ann. Biol. Clin. 2016, 74, 421–427. [Google Scholar] [CrossRef] [PubMed]
  55. DiGiulio, D.B.; Callahan, B.J.; McMurdie, P.J.; Costello, E.K.; Lyell, D.J.; Robaczewska, A.; Sun, C.L.; Goltsman, D.S.A.; Wong, R.J.; Shaw, G.; et al. Temporal and Spatial Variation of the Human Microbiota during Pregnancy. Proc. Natl. Acad. Sci. USA 2015, 112, 11060–11065. [Google Scholar] [CrossRef] [PubMed]
  56. Abramov, V.; Khlebnikov, V.; Kosarev, I.; Bairamova, G.; Vasilenko, R.; Suzina, N.; Machulin, A.; Sakulin, V.; Kulikova, N.; Vasilenko, N.; et al. Probiotic Properties of Lactobacillus Crispatus 2,029: Homeostatic Interaction with Cervicovaginal Epithelial Cells and Antagonistic Activity to Genitourinary Pathogens. Probiotics Antimicrob. Proteins 2014, 6, 165–176. [Google Scholar] [CrossRef] [PubMed]
  57. Abramov, V.M.; Kosarev, I.V.; Priputnevich, T.V.; Machulin, A.V.; Khlebnikov, V.S.; Pchelintsev, S.Y.; Vasilenko, R.N.; Sakulin, V.K.; Suzina, N.E.; Chikileva, I.O.; et al. S-Layer Protein 2 of Lactobacillus Crispatus 2029, Its Structural and Immunomodulatory Characteristics and Roles in Protective Potential of the Whole Bacteria against Foodborne Pathogens. Int. J. Biol. Macromol. 2020, 150, 400–412. [Google Scholar] [CrossRef] [PubMed]
  58. Rose, W.A.; McGowin, C.L.; Spagnuolo, R.A.; Eaves-Pyles, T.D.; Popov, V.L.; Pyles, R.B. Commensal Bacteria Modulate Innate Immune Responses of Vaginal Epithelial Cell Multilayer Cultures. PLoS ONE 2012, 7, e32728. [Google Scholar] [CrossRef] [PubMed]
  59. Doerflinger, S.Y.; Throop, A.L.; Herbst-Kralovetz, M.M. Bacteria in the Vaginal Microbiome Alter the Innate Immune Response and Barrier Properties of the Human Vaginal Epithelia in a Species-Specific Manner. J. Infect. Dis. 2014, 209, 1989–1999. [Google Scholar] [CrossRef]
  60. Fontana, F.; Alessandri, G.; Lugli, G.A.; Mancabelli, L.; Longhi, G.; Anzalone, R.; Viappiani, A.; Ventura, M.; Turroni, F.; Milani, C. Probiogenomics Analysis of 97 Lactobacillus Crispatus Strains as a Tool for the Identification of Promising Next-Generation Probiotics. Microorganisms 2020, 9, 73. [Google Scholar] [CrossRef]
  61. Stoyancheva, G.; Marzotto, M.; Dellaglio, F.; Torriani, S. Bacteriocin Production and Gene Sequencing Analysis from Vaginal Lactobacillus Strains. Arch. Microbiol. 2014, 196, 645–653. [Google Scholar] [CrossRef]
  62. Karlyshev, A.V.; Melnikov, V.G.; Khlebnikov, V.C.; Abramov, V.M. Draft Genome Sequence of Lactobacillus Crispatus 2029. Genome Announc. 2014, 2, e01221-13. [Google Scholar] [CrossRef] [PubMed]
  63. Mancabelli, L.; Mancino, W.; Lugli, G.A.; Milani, C.; Viappiani, A.; Anzalone, R.; Longhi, G.; van Sinderen, D.; Ventura, M.; Turroni, F. Comparative Genome Analyses of Lactobacillus Crispatus Isolates from Different Ecological Niches Reveal an Adaptation of This Species to the Human Vaginal Environment. Appl. Environ. Microbiol. 2021, 87, e02899-20. [Google Scholar] [CrossRef] [PubMed]
  64. Argentini, C.; Fontana, F.; Alessandri, G.; Lugli, G.A.; Mancabelli, L.; Ossiprandi, M.C.; van Sinderen, D.; Ventura, M.; Milani, C.; Turroni, F. Evaluation of Modulatory Activities of Lactobacillus Crispatus Strains in the Context of the Vaginal Microbiota. Microbiol. Spectr. 2022, 10, e0273321. [Google Scholar] [CrossRef] [PubMed]
  65. Abramov, V.M.; Kosarev, I.V.; Priputnevich, T.V.; Machulin, A.V.; Abashina, T.N.; Chikileva, I.O.; Donetskova, A.D.; Takada, K.; Melnikov, V.G.; Vasilenko, R.N.; et al. S-Layer Protein 2 of Vaginal Lactobacillus Crispatus 2029 Enhances Growth, Differentiation, VEGF Production and Barrier Functions in Intestinal Epithelial Cell Line Caco-2. Int. J. Biol. Macromol. 2021, 189, 410–419. [Google Scholar] [CrossRef] [PubMed]
  66. Wooten, J.M. The Coaggregative Abilities of Lactobacilli and Campylobacter Jejuni. Master’s Thesis, University of Illinois, Urbana-Champaign, IL, USA, 2015. [Google Scholar]
  67. Chen, X.; Xu, J.; Shuai, J.; Chen, J.; Zhang, Z.; Fang, W. The S-Layer Proteins of Lactobacillus Crispatus Strain ZJ001 Is Responsible for Competitive Exclusion against Escherichia Coli O157:H7 and Salmonella Typhimurium. Int. J. Food Microbiol. 2007, 115, 307–312. [Google Scholar] [CrossRef]
  68. Graf, K.; Last, A.; Gratz, R.; Allert, S.; Linde, S.; Westermann, M.; Gröger, M.; Mosig, A.S.; Gresnigt, M.S.; Hube, B. Keeping Candida Commensal: How Lactobacilli Antagonize Pathogenicity of Candida albicans in an in Vitro Gut Model. Dis. Model. Mech. 2019, 12, dmm039719. [Google Scholar] [CrossRef] [PubMed]
  69. Behzadi, P.; Behzadi, E.; Ranjbar, R. Urinary Tract Infections and Candida albicans. Cent. Eur. J. Urol. 2015, 68, 96–101. [Google Scholar] [CrossRef]
  70. Behzadi, P.; Behzadi, E.; Pawlak-Adamska, E.A. Urinary Tract Infections (UTIs) or Genital Tract Infections (GTIs)? It’s the Diagnostics That Count. GMS Hyg. Infect. Control 2019, 14, Doc14. [Google Scholar] [CrossRef]
  71. Wang, S.; Wang, Q.; Yang, E.; Yan, L.; Li, T.; Zhuang, H. Antimicrobial Compounds Produced by Vaginal Lactobacillus Crispatus Are Able to Strongly Inhibit Candida albicans Growth, Hyphal Formation and Regulate Virulence-Related Gene Expressions. Front. Microbiol. 2017, 8, 564. [Google Scholar] [CrossRef]
  72. James, K.M.; MacDonald, K.W.; Chanyi, R.M.; Cadieux, P.A.; Burton, J.P. Inhibition of Candida albicans Biofilm Formation and Modulation of Gene Expression by Probiotic Cells and Supernatant. J. Med. Microbiol. 2016, 65, 328–336. [Google Scholar] [CrossRef]
  73. Matsuda, Y.; Cho, O.; Sugita, T.; Ogishima, D.; Takeda, S. Culture Supernatants of Lactobacillus gasseri and L. crispatus Inhibit Candida albicans Biofilm Formation and Adhesion to HeLa Cells. Mycopathologia 2018, 183, 691–700. [Google Scholar] [CrossRef] [PubMed]
  74. Rupniak, H.T.; Rowlatt, C.; Lane, E.B.; Steele, J.G.; Trejdosiewicz, L.K.; Laskiewicz, B.; Povey, S.; Hill, B.T. Characteristics of Four New Human Cell Lines Derived from Squamous Cell Carcinomas of the Head and Neck. J. Natl. Cancer Inst. 1985, 75, 621–635. [Google Scholar] [PubMed]
  75. Handley, P.S.; Harty, D.W.; Wyatt, J.E.; Brown, C.R.; Doran, J.P.; Gibbs, A.C. A Comparison of the Adhesion, Coaggregation and Cell-Surface Hydrophobicity Properties of Fibrillar and Fimbriate Strains of Streptococcus Salivarius. J. Gen. Microbiol. 1987, 133, 3207–3217. [Google Scholar] [CrossRef] [PubMed]
  76. Chan, F.K.-M.; Moriwaki, K.; De Rosa, M.J. Detection of Necrosis by Release of Lactate Dehydrogenase Activity. Methods Mol. Biol. 2013, 979, 65–70. [Google Scholar] [CrossRef]
  77. Zhang, Y.; Chen, X.; Gueydan, C.; Han, J. Plasma Membrane Changes during Programmed Cell Deaths. Cell Res. 2018, 28, 9–21. [Google Scholar] [CrossRef]
  78. Antonio, M.A.D.; Hillier, S.L. DNA Fingerprinting of Lactobacillus crispatus Strain CTV-05 by Repetitive Element Sequence-Based PCR Analysis in a Pilot Study of Vaginal Colonization. J. Clin. Microbiol. 2003, 41, 1881–1887. [Google Scholar] [CrossRef]
  79. Li, T.; Liu, Z.; Zhang, X.; Chen, X.; Wang, S. Local Probiotic Lactobacillus crispatus and Lactobacillus delbrueckii Exhibit Strong Antifungal Effects against Vulvovaginal Candidiasis in a Rat Model. Front. Microbiol. 2019, 10, 1033. [Google Scholar] [CrossRef]
  80. Donnarumma, G.; Molinaro, A.; Cimini, D.; De Castro, C.; Valli, V.; De Gregorio, V.; De Rosa, M.; Schiraldi, C. Lactobacillus Crispatus L1: High Cell Density Cultivation and Exopolysaccharide Structure Characterization to Highlight Potentially Beneficial Effects against Vaginal Pathogens. BMC Microbiol. 2014, 14, 137. [Google Scholar] [CrossRef]
  81. Koh, W.-J.; Abu-Rustum, N.R.; Bean, S.; Bradley, K.; Campos, S.M.; Cho, K.R.; Chon, H.S.; Chu, C.; Clark, R.; Cohn, D.; et al. Cervical Cancer, Version 3.2019, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2019, 17, 64–84. [Google Scholar] [CrossRef]
  82. Nikou, S.-A.; Kichik, N.; Brown, R.; Ponde, N.O.; Ho, J.; Naglik, J.R.; Richardson, J.P. Candida albicans Interactions with Mucosal Surfaces during Health and Disease. Pathogens 2019, 8, 53. [Google Scholar] [CrossRef]
  83. Martin, H.; Masterson, H.; Kavanagh, K.; Velasco-Torrijos, T. The Synthesis and Evaluation of Multivalent Glycopeptoids as Inhibitors of the Adhesion of Candida albicans. Pathogens 2021, 10, 572. [Google Scholar] [CrossRef] [PubMed]
  84. Delgado, A.C.D.; de Jesus Pedro, R.; Aoki, F.H.; Resende, M.R.; Trabasso, P.; Colombo, A.L.; de Oliveira, M.S.M.; Mikami, Y.; Moretti, M.L. Clinical and Microbiological Assessment of Patients with a Long-Term Diagnosis of Human Immunodeficiency Virus Infection and Candida Oral Colonization. Clin. Microbiol. Infect. 2009, 15, 364–371. [Google Scholar] [CrossRef] [PubMed]
  85. Thompson, G.R.; Patel, P.K.; Kirkpatrick, W.R.; Westbrook, S.D.; Berg, D.; Erlandsen, J.; Redding, S.W.; Patterson, T.F. Oropharyngeal Candidiasis in the Era of Antiretroviral Therapy. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endodontol. 2010, 109, 488–495. [Google Scholar] [CrossRef] [PubMed]
  86. Crandall, M. The Pathogenetic Significance of Intestinal Candida Colonization. Int. J. Hyg. Environ. Health 2004, 207, 79–81. [Google Scholar] [CrossRef] [PubMed]
  87. Lechuga, S.; Ivanov, A.I. Disruption of the Epithelial Barrier during Intestinal Inflammation: Quest for New Molecules and Mechanisms. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 1183–1194. [Google Scholar] [CrossRef]
  88. Talapko, J.; Juzbašić, M.; Matijević, T.; Pustijanac, E.; Bekić, S.; Kotris, I.; Škrlec, I. Candida albicans—The Virulence Factors and Clinical Manifestations of Infection. J. Fungi 2021, 7, 79. [Google Scholar] [CrossRef]
  89. Dang, A.T.; Marsland, B.J. Microbes, Metabolites, and the Gut-Lung Axis. Mucosal Immunol. 2019, 12, 843–850. [Google Scholar] [CrossRef]
  90. Richardson, J.P.; Mogavero, S.; Moyes, D.L.; Blagojevic, M.; Krüger, T.; Verma, A.H.; Coleman, B.M.; De La Cruz Diaz, J.; Schulz, D.; Ponde, N.O.; et al. Processing of Candida albicans Ece1p Is Critical for Candidalysin Maturation and Fungal Virulence. mBio 2018, 9, e02178-17. [Google Scholar] [CrossRef]
  91. Russell, C.M.; Schaefer, K.G.; Dixson, A.; Gray, A.L.H.; Pyron, R.J.; Alves, D.S.; Moore, N.; Conley, E.A.; Schuck, R.J.; White, T.A.; et al. The Candida albicans Virulence Factor Candidalysin Polymerizes in Solution to Form Membrane Pores and Damage Epithelial Cells. eLife 2022, 11, e75490. [Google Scholar] [CrossRef]
  92. Nagata, S.; Tanaka, M. Programmed Cell Death and the Immune System. Nat. Rev. Immunol. 2017, 17, 333–340. [Google Scholar] [CrossRef]
  93. Kono, H.; Rock, K.L. How Dying Cells Alert the Immune System to Danger. Nat. Rev. Immunol. 2008, 8, 279–289. [Google Scholar] [CrossRef] [PubMed]
  94. Burd, R.S.; Furrer, J.L.; Sullivan, J.; Smith, A.L. Murine Beta-Defensin-3 Is an Inducible Peptide with Limited Tissue Expression and Broad-Spectrum Antimicrobial Activity. Shock 2002, 18, 461–464. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Inhibitory effects of the LC2029 strain on the transition of C. albicans yeast form to hyphal. Optical microscopy (bar—10 µm). (A) Yeast form of C. albicans strain ATCC 10231. (B) Monolayer (formed by 100%) of intact immature HeLa cells. (C) Intensive transition of C. albicans yeast form to hyphal on intact monolayer of HeLa cells. All cells of the C. albicans yeast form, after being added to the intact HeLa monolayer, turned into hyphal form after 2 h of joint cultivation. (D) The monolayer (formed by100%) of immature HeLa cells is protected by cells of strain LC2029 in the adhered state. (E) The monolayer (formed by 70–80%) of immature HeLa cells is protected by cells of the LC2029 strain in the adhered state. Cells of the LC2029 strain are also visible in the nutrient medium in a suspension state. Thirty minutes after the introduction of the C. albicans in yeast form into the wells (the ratio of LC2029 strain/C. albicans yeast form is 100/1 CFU/mL), the cells of the LC2029 strain in the nutrient medium in suspension form co-aggregates with the C. albicans yeast form. (F) Co-aggregates of strain LC2029 cells with C. albicans cells in yeast form are removed when the culture medium is replaced or washed once by PBS. Only single C. albicans cells in yeast form are visible in the field of view HeLa cells.
Figure 1. Inhibitory effects of the LC2029 strain on the transition of C. albicans yeast form to hyphal. Optical microscopy (bar—10 µm). (A) Yeast form of C. albicans strain ATCC 10231. (B) Monolayer (formed by 100%) of intact immature HeLa cells. (C) Intensive transition of C. albicans yeast form to hyphal on intact monolayer of HeLa cells. All cells of the C. albicans yeast form, after being added to the intact HeLa monolayer, turned into hyphal form after 2 h of joint cultivation. (D) The monolayer (formed by100%) of immature HeLa cells is protected by cells of strain LC2029 in the adhered state. (E) The monolayer (formed by 70–80%) of immature HeLa cells is protected by cells of the LC2029 strain in the adhered state. Cells of the LC2029 strain are also visible in the nutrient medium in a suspension state. Thirty minutes after the introduction of the C. albicans in yeast form into the wells (the ratio of LC2029 strain/C. albicans yeast form is 100/1 CFU/mL), the cells of the LC2029 strain in the nutrient medium in suspension form co-aggregates with the C. albicans yeast form. (F) Co-aggregates of strain LC2029 cells with C. albicans cells in yeast form are removed when the culture medium is replaced or washed once by PBS. Only single C. albicans cells in yeast form are visible in the field of view HeLa cells.
Biomolecules 13 01740 g001
Table 1. The microorganisms used in this study.
Table 1. The microorganisms used in this study.
MicroorganismStrainGrowth Conditions
L. crispatusLC2029 1MRS a, 37 °C in a CO2 incubator, 10% CO2 or anaerobically 48 h
L. crispatusLC1385 2The same
C. albicansATCC 3 10231SD b 37 °C in 5% CO2 24–48 h
C. albicansATCC 11006The same
C. albicansATCC 2091The same
C. albicansATCC 64548The same
C. albicansMD IIE Ca-CI-8 4The same
C. albicansMD IIE Ca-CI-126 5The same
C. albicansMD IIE Ca-CI-98 6The same
C. albicansMD IIE Ca-CI-154 7The same
1 Russian Collection of Microorganisms at the Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research, RAS” Pushchino, Moscow Region, Russia. 2 Collection of Microorganisms at the Institute of Immunological Engineering, Department of Biochemistry of Immunity and Biodefence, Lyubuchany, Moscow Region, Russia. 3 American Type Culture Collection, Manassas, VA, USA. 4 Clinical isolates (recurrent VVC). 5 Clinical isolates (OPC). 6 Clinical isolates (IC). 7 Clinical bloodstream isolates. a MRS broth or an agar containing plates (HiMedia, India). b SD broth or an agar containing plates (HiMedia, India).
Table 2. Co-aggregative abilities of the LC2029 strain and Slp2-negative LC1385 strain with unicellular yeast form of C. albicans strains.
Table 2. Co-aggregative abilities of the LC2029 strain and Slp2-negative LC1385 strain with unicellular yeast form of C. albicans strains.
StrainLC2029LC1385
C. albicans ATCC 1023176.1 ± 4.6 *** 3.0 ± 0.1
C. albicans ATCC 1100682.8 ± 5.4 ***2.1 ± 0.5
C. albicans ATCC 209179.3 ± 4.8 *** 4.2 ± 0.7
C. albicans ATCC 6454874.7 ± 4.3 *** 2.9 ± 0.5
C. albicans MD IIE Ca-CI-882.0 ± 5.5 *** 3.0 ± 0.2
C. albicans MD IIE Ca-CI-12678.2 ± 5.2 *** 2.2 ± 0.4
C. albicans MD IIE Ca-CI-9884.1 ± 6.5 *** 3.4 ± 0.7
C. albicans MD IIE Ca-CI-15478.6 ± 4.8 ***2.1 ± 0.5
*** p < 0.001—сo-aggregation of the LC2029 strain with C. albicans strains vs. сo-aggregation of the LC1385 strain with C. albicans strains. All data are representative of six independent experiments tested in triplicate.
Table 3. Slp2 reduction in the C. albicans strains in the process of adhesion to HeLa cells.
Table 3. Slp2 reduction in the C. albicans strains in the process of adhesion to HeLa cells.
StrainHeLa Cells Containing Adhered Bacteria, %
Concentration of Slp2 in a Medium, µg/mL
01050100
C. albicans ATCC 1023167.0 ± 4.635.1 ± 3.8 *12.0 ± 1.3 **2.1 ± 0.4 ***
C. albicans ATCC 1100665.4 ± 3.839.0 ± 3.6 *14.2 ± 1.5 **1.9 ± 0.6 ***
C. albicans ATCC 209175.5 ± 4.542.2 ± 3.7 *15.1 ± 1.7 **3.2 ± 0.4 ***
C. albicans ATCC 6454869.1 ± 5.230.0 ± 3.5 *12.9 ± 1.2 **1.7 ± 0.3 ***
C. albicans MD IIE Ca-CI-887.0 ± 2.738.4 ± 3.2 *15.5 ± 2.0 **3.0 ± 0.5 ***
C. albicans MD IIE Ca-CI-12675.2 ± 3.536.7 ± 4.5 *13.1 ± 1.9 **3.1 ± 0.6 ***
C. albicans MD IIE Ca-CI-9863.4 ± 2.636.2 ± 4.2 *15.0 ± 3.7 **3.2 ± 0.5 ***
C. albicans MD IIE Ca-CI-15464.7 ± 3.429.1 ± 3.7 *17.4 ± 4.2 **1.9 ± 0.4 ***
* p < 0.05 adhesion of strains (C. albicans ATCC 10231, C. albicans ATCC 11006, C. albicans ATCC 2091, C. albicans ATCC 64548, C. albicans MD IIE Ca-CI-8, C. albicans MD IIE Ca-CI-126, C. albicans MD IIE Ca-CI-98, C. albicans MD IIE Ca-CI-154) to HeLa cells alone vs. adhesion to HeLa cells + Slp2 (10 µg/mL); ** p < 0.01 adhesion of strains to HeLa cells alone vs. adhesion to HeLa cells + Slp2 (50 µg/mL); *** p < 0.001 adhesion of strains to HeLa cells alone vs. adhesion to HeLa cells + Slp2 (100 µg/mL). Data represent the mean and SEM of six independent experiments tested in triplicate.
Table 4. Slp2 reduction in the C. albicans strains in the process of adhesion to human Vk2/E6E7 vaginal epithelial cells.
Table 4. Slp2 reduction in the C. albicans strains in the process of adhesion to human Vk2/E6E7 vaginal epithelial cells.
StrainVk2/E6E7 Cells Containing Adhered Bacteria, %
Concentration of Slp2 in a Medium, µg/mL
01050100
C. albicans ATCC 1023164.1 ± 5.529.1 ± 3.6 *10.2 ± 1.4 **3.0 ± 0.7 ***
C. albicans ATCC 1100673.4 ± 4.642.4 ± 4.1 *13.2 ± 1.7 **2.1 ± 0.4 ***
C. albicans ATCC 209168.5 ± 3.728.1 ± 2.5 *12.3 ± 1.5 **2.2 ± 0.3 ***
C. albicans ATCC 6454865.0 ± 6.226.8 ± 3.6 *13.8 ± 1.3 **2.8 ± 0.6 ***
C. albicans MD IIE Ca-CI-854.2 ± 3.925.0 ± 2.8 *11.1 ± 1.5 **2.2 ± 0.4 ***
C. albicans MD IIE Ca-CI-12663.2 ± 4.936.1 ± 4.5 *9.0 ± 1.2 **3.9 ± 0.5 ***
C. albicans MD IIE Ca-CI-9856.1 ± 2.533.6 ± 3.7 *12.4 ± 1.0 **4.3 ± 0.8 ***
C. albicans MD IIE Ca-CI-15466.8 ± 5.338.0 ± 4.6 *12.6 ± 1.5 **1.8 ± 0.3 ***
* p < 0.05 adhesion of strains (C. albicans ATCC 10231, C. albicans ATCC 11006, C. albicans ATCC 2091, C. albicans ATCC 64548, C. albicans MD IIE Ca-CI-8, C. albicans MD IIE Ca-CI-126, C. albicans MD IIE Ca-CI-98, C. albicans MD IIE Ca-CI-154) to Vk2/E6E7 cells alone vs. adhesion to Vk2/E6E7 cells + Slp2 (10 µg/mL); ** p < 0.01 adhesion of strains to Vk2/E6E7 cells alone vs. adhesion to Vk2/E6E7 cells + Slp2 (50 µg/mL); *** p < 0.001 adhesion of strains to Vk2/E6E7 cells alone vs. adhesion to Vk2/E6E7 cells + Slp2 (100 µg/mL). Data represent the mean and SEM of six independent experiments tested in triplicate.
Table 5. Slp2 reduction in the C. albicans strains in the process of adhesion to buccal TR146 epithelial cells.
Table 5. Slp2 reduction in the C. albicans strains in the process of adhesion to buccal TR146 epithelial cells.
StrainTR146 Cells Containing Adhered Bacteria, %
Concentration of Slp2 in a Medium, µg/mL
01050100
C. albicans ATCC 1023154.2 ± 5.528.1 ± 4.3 *9.1 ± 1.6 **2.1 ± 0.5 ***
C. albicans ATCC 1100657.5 ± 5.328.8 ± 3.6 *10.0 ± 1.8 **3.0 ± 0.4 ***
C. albicans ATCC 209174.9 ± 4.633.8 ± 2.5 *10.0 ± 1.3 **3.2 ± 0.2 ***
C. albicans ATCC 6454855.8 ± 5.338.0 ± 4.2 *11.8 ± 1.6 **3.9 ± 0.5 ***
C. albicans MD IIE Ca-CI-877.8 ± 5.641.2 ± 3.8 *10.1 ± 1.5 **3.1 ± 0.3 ***
C. albicans MD IIE Ca-CI-12659.1 ± 4.335.2 ± 2.6 *13.9 ± 2.7 **2.0 ± 0.4 ***
C. albicans MD IIE Ca-CI-9868.1 ± 4.639.0 ± 3.5 *11.0 ± 1.8 **2.8 ± 0.5 ***
C. albicans MD IIE Ca-CI-15452.2 ± 4.734.3 ± 2.8 *11.8 ± 1.5 **4.0 ± 0.6 ***
* p < 0.05 adhesion of strains (C. albicans ATCC 10231, C. albicans ATCC 11006, C. albicans ATCC 2091, C. albicans ATCC 64548, C. albicans MD IIE Ca-CI-8, C. albicans MD IIE Ca-CI-126, C. albicans MD IIE Ca-CI-98, C. albicans MD IIE Ca-CI-154) to TR146 cells alone vs. adhesion to TR146 cells + Slp2 (10 µg/mL); ** p < 0.01 adhesion of strains to TR146 cells alone vs. adhesion to TR146 cells + Slp2 (50 µg/mL); *** p < 0.001 adhesion of strains to TR146 cells alone vs. adhesion to TR146 cells + Slp2 (100 µg/mL). Data represent the mean and SEM of six independent experiments tested in triplicate.
Table 6. Slp2 reduction in the C. albicans strains in the process of adhesion to intestinal Caco-2 cells.
Table 6. Slp2 reduction in the C. albicans strains in the process of adhesion to intestinal Caco-2 cells.
StrainCaco-2 Cells Containing Adhered Bacteria, %
Concentration of Slp2 in a Medium, µg/mL
01050100
C. albicans ATCC 1023178.4 ± 3.529.1 ± 2.6 *11.1 ± 2.1 **4.1 ± 0.3 ***
C. albicans ATCC 1100663.0 ± 3.031.5 ± 2.5 *13.2 ± 2.6 **3.2 ± 0.2 ***
C. albicans ATCC 209175.1 ± 4.540.7 ± 3.4 *15.0 ± 3.2 **2.0 ± 0.3 ***
C. albicans ATCC 6454856.2 ± 2.340.0 ± 2.6 *14.0 ± 2.8 **3.9 ± 0.5 ***
C. albicans MD IIE Ca-CI-873.1 ± 3.831.9 ± 3.7 *12.4 ± 2.5 **2.9 ± 0.4 ***
C. albicans MD IIE Ca-CI-12663.8 ± 2.641.2 ± 3.8 *10.9 ± 2.3 **2.0 ± 0.3 ***
C. albicans MD IIE Ca-CI-9864.7 ± 4.027.4 ± 4.3 *15.9 ± 3.1 **3.1 ± 0.5 ***
C. albicans MD IIE Ca-CI-15472.7 ± 3.830.6 ± 3.6 *11.8 ± 2.4 **3.3 ± 0.4 ***
* p < 0.05 adhesion of strains (C. albicans ATCC 10231, C. albicans ATCC 11006, C. albicans ATCC 2091, C. albicans ATCC 64548, C. albicans MD IIE Ca-CI-8, C. albicans MD IIE Ca-CI-126, C. albicans MD IIE Ca-CI-98, C. albicans MD IIE Ca-CI-154) to Caco-2 alone vs. adhesion to Caco-2 + Slp2 (10 µg/mL); ** p < 0.01 adhesion of strains to Caco-2 alone vs. adhesion to Caco-2 + Slp2 (50 µg/mL); *** p < 0.001 adhesion of strains to Caco-2 alone vs. adhesion to Caco-2 + Slp2 (100 µg/mL). Data represent the mean and SEM of six independent experiments tested in triplicate.
Table 7. Slp2 reduction in the C. albicans strains in the process of adhesion to intestinal HT-29 cells.
Table 7. Slp2 reduction in the C. albicans strains in the process of adhesion to intestinal HT-29 cells.
StrainHT-29 Cells Containing Adhered Bacteria, %
Concentration of Slp2 in a Medium, µg/mL
01050100
C. albicans ATCC 1023159.2 ± 3.536.1 ± 2.8 *9.0 ± 1.4 **2.9 ± 0.2 ***
C. albicans ATCC 1100666.5 ± 3.228.8 ± 2.5 *12.1 ± 2.1 **2.1 ± 0.1 ***
C. albicans ATCC 209171.0 ± 3.632.2 ± 3.2 *13.8 ± 2.5 **3.8 ± 0.5 ***
C. albicans ATCC 6454864.8 ± 4.836.7 ± 2.7 *11.2 ± 2.0 **2.0 ± 0.1 ***
C. albicans MD IIE Ca-CI-858.0 ± 4.241.0 ± 3.1 *9.5 ± 1.2 **2.0 ± 0.1 ***
C. albicans MD IIE Ca-CI-12671.1 ± 3.835.1 ± 2.6 *10.8 ± 1.8 **3.1 ± 0.2 ***
C. albicans MD IIE Ca-CI-9867.8 ± 3.742.9 ± 2.8 *12.6 ± 1.9 **2.0 ± 0.4 ***
C. albicans MD IIE Ca-CI-15473.0 ± 4.036.0 ± 3.5 *12.0 ± 2.1 **2.9 ± 0.3 ***
* p < 0.05 adhesion of strains (C. albicans ATCC 10231, C. albicans ATCC 11006, C. albicans ATCC 2091, C. albicans ATCC 64548, C. albicans MD IIE Ca-CI-8, C. albicans MD IIE Ca-CI-126, C. albicans MD IIE Ca-CI-98, C. albicans MD IIE Ca-CI-154) to HT-29 alone vs. adhesion to HT-29 + Slp2 (10 µg/mL); ** p < 0.01 adhesion of strains to HT-29 alone vs. adhesion to HT-29+ Slp2 (50 µg/mL); *** p < 0.001 adhesion of strains to HT-29 alone vs. adhesion to HT-29 + Slp2 (100 µg/mL). Data represent the mean and SEM of six independent experiments tested in triplicate.
Table 8. Slp2 reduction in the C. albicans strains in the process of adhesion to HUVECs.
Table 8. Slp2 reduction in the C. albicans strains in the process of adhesion to HUVECs.
StrainHUVECs Containing Adhered Bacteria, %
Concentration of Slp2 in a Medium, µg/mL
01050100
C. albicans ATCC 1023172.3 ± 4.335.6 ± 2.1 *14.0 ± 1.0 **2.1 ± 0.1 ***
C. albicans ATCC 1100669.1 ± 3.634.1 ± 2.5 *11.2 ± 0.9 **1.8 ± 0.3 ***
C. albicans ATCC 209172.7 ± 4.140.0 ± 2.3 *10.0 ± 0.8 **2.0 ± 0.1 ***
C. albicans ATCC 6454864.5 ± 3.432.3 ± 2.4 *11.7 ± 1.2 **3.1 ± 0.2 ***
C. albicans MD IIE Ca-CI-857.7 ± 3.235.1 ± 2.1 *11.0 ± 1.0 **2.0 ± 0.1 ***
C. albicans MD IIE Ca-CI-12671.0 ± 3.538.0 ± 2.2 *12.9 ± 1.1 **2.8 ± 0.5 ***
C. albicans MD IIE Ca-CI-9869.1 ± 4.129.7 ± 2.7 *11.1 ± 0.9 **2.0 ± 0.1 ***
C. albicans MD IIE Ca-CI-15467.0 ± 3.839.1 ± 2.5 *15.0 ± 1.4 **3.8 ± 0.6 ***
* p < 0.05 adhesion of strains (C. albicans ATCC 10231, C. albicans ATCC 11006, C. albicans ATCC 2091, C. albicans ATCC 64548, C. albicans MD IIE Ca-CI-8, C. albicans MD IIE Ca-CI-126, C. albicans MD IIE Ca-CI-98, C. albicans MD IIE Ca-CI-154) to HUVECs alone vs. adhesion to HUVECs + Slp2 (10 µg/mL); ** p < 0.01 adhesion of strains to HUVECs alone vs. adhesion to HUVECs + Slp2 (50 µg/mL); *** p < 0.001 adhesion of strains to HUVECs alone vs. adhesion to HUVECs + Slp2 (100 µg/mL). Data represent the mean and SEM of six independent experiments tested in triplicate.
Table 9. Slp2 reduction in the C. albicans strains in the process of adhesion to lung A-549 epithelial cells.
Table 9. Slp2 reduction in the C. albicans strains in the process of adhesion to lung A-549 epithelial cells.
StrainA-549 Cells Containing Adhered Bacteria, %
Concentration of Slp2 in a Medium, µg/mL
01050100
C. albicans ATCC 1023179.6 ± 4.637.1 ± 2.9 *16.0 ± 2.8 **3.1 ± 0.5 ***
C. albicans ATCC 1100676.1 ± 3.935.0 ± 3.1 *16.8 ± 3.1 **2.0 ± 0.1 ***
C. albicans ATCC 209182.1 ± 4.338.0 ± 3.5 *15.2 ± 2.4 **2.0 ± 0.4 ***
C. albicans ATCC 6454878.9 ± 5.442.7 ± 3.6 *13.5 ± 2.2 **2.9 ± 0.2 ***
C. albicans MD IIE Ca-CI-887.2 ± 4.239.4 ± 4.8 *18.8 ± 2.5 **3.2 ± 0.4 ***
C. albicans MD IIE Ca-CI-12684.3 ± 3.942.0 ± 3.5 *15.0 ± 3.4 **2.1 ± 0.2 ***
C. albicans MD IIE Ca-CI-9883.2 ± 4.735.1 ± 4.2 *18.1 ± 2.3 **1.1 ± 0.2 ***
C. albicans MD IIE Ca-CI-15488.7 ± 4.541.1 ± 3.7 *17.4 ± 2.6 **2.0 ± 0.3 ***
* p < 0.05 adhesion of strains (C. albicans ATCC 10231, C. albicans ATCC 11006, C. albicans ATCC 2091, C. albicans ATCC 64548, C. albicans MD IIE Ca-CI-8, C. albicans MD IIE Ca-CI-126, C. albicans MD IIE Ca-CI-98, C. albicans MD IIE Ca-CI-154) to A-549 alone vs. adhesion to A-549 + Slp2 (10 µg/mL); ** p < 0.01 adhesion of strains to A-549 alone vs. adhesion to A-549 + Slp2 (50 µg/mL); *** p < 0.001 adhesion of strains to A-549 alone vs. adhesion to A-549 + Slp2 (100 µg/mL). Data represent the mean and SEM of six independent experiments tested in triplicate.
Table 10. Colonization of epithelial cells with the LC2029 strain reduces C. albicans-induced necrosis.
Table 10. Colonization of epithelial cells with the LC2029 strain reduces C. albicans-induced necrosis.
Epithelial Cells LDH (ng/mL)
MediumLC1385LC2029C. albicansC. albicans + LC1385C. albicans + LC2029
HeLa40 ± 441 ± 441 ± 4118 ± 5 **123 ± 5 **43 ± 4
Vk2/E6EF50 ± 453 ± 453 ± 3123 ± 7 **114 ± 5 **53 ± 3
TR 14635 ± 438 ± 438 ± 3138 ± 5 **128 ± 6 **40 ± 4
Caco-259 ± 560 ± 360 ± 4145 ± 8 **133 ± 8 **63 ± 4
HT-2953 ± 453 ± 353 ± 4147 ± 5 **138 ± 6 **50 ± 5
A-54938 ± 538 ± 538 ± 6135 ± 6 **126 ± 6 **43 ± 4
HUVECs38 ± 340 ± 440 ± 5116 ± 7 **115 ± 5 **41 ± 3
** p < 0.01—LDH production in Hela cells, Vk2/E6E7 cells, TR 146 cells, Caco-2 cells, HT-29 cells, A-549 cells, HUVECs induced by C. albicans ATCC 10231 and C. albicans + LC1385 vs. the medium, Slp2-negative LC1385 strain, Slp2-negative ΔLC2029 strain. Data represent the mean and SEM of six independent experiments tested in triplicate.
Table 11. Production of HBD-3 in epithelial cells from various human biotopes.
Table 11. Production of HBD-3 in epithelial cells from various human biotopes.
Epithelial Cells HBD-3 (pg/mL)
MediumLC1385ΔLC2029LC2029Slp2
HeLa3 ± 13 ± 217 ± 3157 ± 16 ***174 ±19 ***
Vk2/E6E73 ± 23 ± 213 ± 3165 ± 14 ***168 ± 17 ***
TR 1467 ± 37 ± 317 ± 5153 ±15 ***164 ± 15 ***
Caco-27 ± 37 ± 313 ± 5169 ± 18 ***175 ±16 ***
HT-295 ± 35 ± 317 ± 6172 ±16 ***183 ± 14 ***
A-5493 ± 23 ± 210 ± 597 ± 8 **94 ± 7 **
HUVECs3 ± 13 ± 113 ± 383 ± 5 **80 ± 6 **
** p < 0.01—production of HBD-3 in A-549 cells and HUVECs induced by the Slp2 and LC2029 strain vs. the medium, Slp2-negative LC1385 strain, Slp2-negative ΔLC2029 strain; *** p < 0.001—production of HBD-3 in HeLa cells, Vk2/E6E7 cells, TR 146 cells, Caco-2 cells, HT-29 cells induced by the Slp2 and LC2029 strain vs. the medium, Slp2-negative LC1385 strain, Slp2-negative ΔLC2029 strain. Data represent the mean and SEM of six independent experiments tested in triplicate.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Abramov, V.M.; Kosarev, I.V.; Machulin, A.V.; Priputnevich, T.V.; Deryusheva, E.I.; Panin, A.N.; Chikileva, I.O.; Abashina, T.N.; Melnikov, V.G.; Suzina, N.E.; et al. Protective Properties of S-layer Protein 2 from Lactobacillus crispatus 2029 against Candida albicans Infections. Biomolecules 2023, 13, 1740. https://doi.org/10.3390/biom13121740

AMA Style

Abramov VM, Kosarev IV, Machulin AV, Priputnevich TV, Deryusheva EI, Panin AN, Chikileva IO, Abashina TN, Melnikov VG, Suzina NE, et al. Protective Properties of S-layer Protein 2 from Lactobacillus crispatus 2029 against Candida albicans Infections. Biomolecules. 2023; 13(12):1740. https://doi.org/10.3390/biom13121740

Chicago/Turabian Style

Abramov, Vyacheslav M., Igor V. Kosarev, Andrey V. Machulin, Tatiana V. Priputnevich, Evgenia I. Deryusheva, Alexander N. Panin, Irina O. Chikileva, Tatiana N. Abashina, Vyacheslav G. Melnikov, Nataliya E. Suzina, and et al. 2023. "Protective Properties of S-layer Protein 2 from Lactobacillus crispatus 2029 against Candida albicans Infections" Biomolecules 13, no. 12: 1740. https://doi.org/10.3390/biom13121740

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