Skip to main content

REVIEW article

Front. Microbiol., 20 September 2022
Sec. Microbial Symbioses

Interactions between Candida albicans and the resident microbiota

  • 1Department of Pharmacy, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
  • 2Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
  • 3Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China

Candida albicans is a prevalent, opportunistic human fungal pathogen. It usually dwells in the human body as a commensal, however, once in its pathogenic state, it causes diseases ranging from debilitating superficial to life-threatening systemic infections. The switch from harmless colonizer to virulent pathogen is, in most cases, due to perturbation of the fungus-host-microbiota interplay. In this review, we focused on the interactions between C. albicans and the host microbiota in the mouth, gut, blood, and vagina. We also highlighted important future research directions. We expect that the evaluation of these interplays will help better our understanding of the etiology of fungal infections and shed new light on the therapeutic approaches.

Introduction

Candida species are commensal colonizers of the human body. They dwell in several niches including the oral cavity, gastrointestinal tract, and vagina. Up to 60% of healthy individuals are colonized by Candida spp. (Kullberg and Arendrup, 2015; McCarty and Pappas, 2016). Candida spp. are also the most prevalent opportunistic human fungal pathogens. They can cause superficial and dermal infections as well as life-threatening invasive candidiasis (Pappas et al., 2018). They are the fourth most common cause of nosocomial bloodstream infections and are the major fungal pathogens isolated from medical device infections (Wisplinghoff et al., 2004; Pfaller and Diekema, 2007). Among the approximately 200 species in the genus Candida, at least 30 of them can cause human disease (Brandt and Lockhart, 2012), but most of Candida-related infections are caused by C. albicans, followed by other non-albicans Candida species, including C. glabrata, C. tropicalis, C. parapsilosis, and C. krusei (Pfaller et al., 2019).

In the host, many microorganisms exist as biofilms. Thus, C. albicans usually exists as a heterogeneous biofilm with microorganisms in the host (Diaz et al., 2014), and infections caused by C. albicans are usually (30–60%) polymicrobial diseases (Sheehan et al., 2020). Under normal conditions, the host’s innate and acquired defense mechanisms and the resident microbiota act in concert to ensure C. albicans inhabits as a commensal. Innate and acquired immune response to C. albicans infections have been extensively reviewed previously (Netea et al., 2008; Richardson and Moyes, 2015; Qin et al., 2016; Pellon et al., 2020). Trillions of microorganisms reside in and on the human body. Some microorganisms are protective to the host while some could be potential pathogens. Under normal circumstances in healthy hosts, the microbiota has a balanced composition of metabolites and energy. The homeostatic equilibrium prevents the overgrowth of potentially pathogenic microorganisms. Imbalance of the microbiota community, also known as dysbiosis, is often associated with human diseases (DeGruttola et al., 2016; El-Sayed et al., 2021; Mizutani et al., 2022). In this review we summarize in vitro and in vivo interactions of C. albicans with the resident microbiota, as well as the molecular mechanisms. We also highlighted some questions that have been previously ignored and discuss suggestions for future research.

Interactions between C. albicans and microbiota in the oral cavity

Human oral cavity provides multiple habitats for the microbiota, including teeth, tongue, cheek, gingival sulcus, attached gingiva, lip, hard, and soft palates. Over 700 different species of bacteria, together with other microorganisms such as fungi, viruses and protozoa, live in the human oral cavity (Marsh and Zaura, 2017). Candida spp. are the most frequent fungi isolated from the oral cavity (Byadarahally Raju and Rajappa, 2011). Oral candidiasis, also known as oral thrush, is a common opportunistic fungal infection of the oral cavity, especially in the elderly or hospitalized patients (Scully et al., 1994; Abu-Elteen and Abu-Alteen, 1998; Fanello et al., 2006). It is mostly caused by overgrowth of C. albicans (Millsop and Fazel, 2016; Hellstein and Marek, 2019). The annual global incidence of oral candidiasis is approximately 2 million (Dufresne et al., 2017). Among HIV-infected patients, oropharyngeal candidiasis (OPC) is the most common oral fungal infections. More than 90% of HIV-positive patients develop OPC at some stage of their disease (Flint et al., 2006).

Bidirectional interactions between C. albicans and oral bacteria

Some oral bacteria enhance while others inhibit biofilm formation in C. albicans. In an in vitro measurement of mixed species biofilm formed on the salivary pellicle, Streptococcus spp. and Actinomyces spp. up-regulated the expression of C. albicans virulence genes and hyphal and biofilm development of C. albicans. Streptococcus oralis also enhanced hyphal and biofilm development of C. albicans (Cavalcanti et al., 2017). In another study, when co-cultured with C. albicans on denture acrylic, Porphyromonas gingivalis inhibited expression of virulence genes and production of hyphae of C. albicans (Morse et al., 2019). In contrast, a mouse intravenous chemotherapy model, C. albicans infection of 5-fluorouracil-immunosuppressed mice caused an overgrowth but decreased diversity of oral bacteria (Bertolini et al., 2019). A study measured 82 Dutch adults for the relative abundance of salivary Candida and bacteria. The study revealed a negative correlation between Candida load and the bacterial profiles of human saliva: increased Candida load was accompanied by decreased bacterial diversity (Kraneveld et al., 2012).

Interactions between C. albicans and S. gordonii

Streptococcus gordonii is one of the first colonizers of the oral cavity (Frandsen et al., 1991). In an in vitro mucosal model, S. gordonii increased the mucosal invasion of the co-cultured C. albicans (Diaz et al., 2012). In C. albicans, deletion of EFG1 and BRG1 caused defection of adhesion and hyphae formation. The co-cultured S. gordonii could restore the ability of forming biofilms in these deletion strains (Montelongo-Jauregui et al., 2019). In the mixed species biofilms formed by C. albicans and S. gordonii, the extracellular matrix produced by C. albicans conferred resistance to antifungal drugs and antibiotics (Montelongo-Jauregui et al., 2016, 2018, 2019). In an in vitro test, S. gordonii and C. albicans were co-cultured on the surface of 24-well culture plates. S. gordonii increased the production of host cytokines IL-8 and TNF-α via up-regulating the expression of the cytokine genes (Bhardwaj et al., 2020). When co-infected with S, gordonii, C. albicans could survive more in murine macrophages, and escape more from murine macrophages, and form larger size of microcolonies on murine macrophage monolayers (Salvatori et al., 2020). In the in vitro mucosal model mentioned above, C. albicans promoted biofilm formation of S. gordonii (Diaz et al., 2012).

Physical contact and chemical signals are involved in the interactions between C. albicans and S. gordonii. The physical contact is mediated by the hyphal-wall proteins Als3p, Eap1p, and to a lesser degree Hwp1p of C. albicans, and S. gordonii cell wall-anchored proteins SspA and SspB of S. gordonii. The chemical interactions involve the production of autoinducer 2 by S. gordonii, which induces hyphal formation, and suppression of farnesol (the quorum sensing molecule of C. albicans) inhibition of hyphal formation of C. albicans (Holmes et al., 1998; Bamford et al., 2009; Nobbs et al., 2010; Silverman et al., 2010). The N-terminal domain of Als3p is required for the physical contact (Bamford et al., 2015). The early O-mannosylation is a critical stage required for activation of hyphal adhesin functions, recognition by S. gordonii and mixed species biofilm formation (Dutton et al., 2014). The only S. gordonii glucosyltransferases (Gtf) enzyme, GtfG, promotes coaggregation with C. albicans, binding to C. albicans preformed biofilms, and formation of mixed biofilm (Ricker et al., 2014). Similarly, the only glucosyltransferases (Gtf) enzyme of S. oralis, GtfR, also enhances mixed species biofilm formation by S. oralis and C. albicans (Souza et al., 2020). The comCDE operon (Jack et al., 2015) and the fruRBA operon (Jesionowski et al., 2016) are required for the development of enhanced mixed species biofilm. Compared to wild type C. albicans strain, co-culturing of S. gordonii with C. albicans strains with deletions of LEU3, CAS4, CTA4, and SKO1 resulted in heavier biofilm but coculturing of S. gordonii with C. albicans strains with deletions of SFL2, BRG1, TEC1, TUP1, EFG1, and RIM101 resulted in lighter biofilms. Co-culturing of S. gordonii with C. albicans strains with deletions of SFL2, TEC1, and EFG1 also caused reduced tolerance to antibiotics (Chinnici et al., 2019).

Interactions between C. albicans and Streptococcus mutans

Streptococcus mutans is the major species associated with dental caries (Loesche, 1986). S. mutans is often isolated together with C. albicans in early childhood caries (Raja et al., 2010; Qiu et al., 2015a,b). In mixed biofilm formed by C. albicans and S. mutans on poly-L-lysine-coated glass slides, S. mutans secreted glucotransferase B (GtfB), which bound to the surfaces of S. mutans and C. albicans. GtfB bound to the surface of C. albicans produced more glucan than GtfB bound to the surface of S. mutans (Hwang et al., 2015). The glucans are binding sites for S. mutans (Gregoire et al., 2011) and the glucans enhance S. mutans adhesion and cohesion to the surface of teeth (Bowen and Koo, 2011). GtfB itself increases C. albicans biofilm formation, even in the bcr1Δ/Δ strain by up-regulating the expression of HWP1, ALS1, and ALS3 in wild type and bcr1Δ/Δ strains of C. albicans (Ellepola et al., 2017). In an in vivo dental plaque biofilm model using healthy female rats, the mannans provided by C. albicans in the outer surface of the cell wall acted as the binding sites of GtfB, which increased the production of the glucan-matrix and modulated the interspecies interaction in the dual species biofilm (Hwang et al., 2017). The secreted polysaccharides by C. albicans might also contribute to the dual species biofilm formation in vitro in 96-well plates and in vivo in immunosuppressed mice (Khoury et al., 2020).

In dual species biofilms formed in 96 well plates, GtfB directly enhanced antifungal tolerance of C. albicans to fluconazole via the exopolysaccharides (EPS) matrix, which were mostly glucans synthesized by GtfB and GtfC. EPS directly bound and sequestered fluconazole. GtfB indirectly enhanced antifungal tolerance of via increasing the expression of C. albicans genes involved in biofilm formation (Ellepola et al., 2017). In an in vitro biofilm model using hydroxyapatite discs coated with saliva, inhibition of GtfB activity by povidone iodine re-established C. albicans susceptibility to fluconazole (Kim et al., 2018). C. albicans also plays an active role by up-regulating genes involved in carbohydrate metabolism, cell morphogenesis and cell wall components such as mannan. GtfB can enhance C. albicans growth and acid production via breaking down sucrose into glucose and fructose, which can be readily metabolized by C. albicans. Environmental acidification is a key virulence factor associated with the onset of caries (Ellepola et al., 2019). In dual species biofilms formed on hydroxyapatite discs coated with saliva, C. albicans could augment the production of exopolysaccharides, which was a key mediator of mixed species biofilm development and induce the expression of S. mutans virulence genes, including gtfB. As a result, the dual species biofilms had more biomass and more viable S. mutans cells than single-species biofilms (Falsetta et al., 2014). In an in vivo murine dental caries model, compared to rats infected with C. albicans or S. mutans alone, co-infected rats displayed higher levels of infection and had more microbial abundance within plaque biofilms. Co-infection also caused higher virulence of plaque biofilms in rats than single-species infection, indicated by the level of colonization by S. mutans and C. albicans in the plaque biofilm, and the onset of carious lesions on teeth surface (Falsetta et al., 2014). On the other hand, when co-cultured in 96-well plates, S. mutans secreted some products which were capable of inhibiting C. albicans morphogenesis, biofilm formation, and pathogenicity (Barbosa et al., 2016; Dos Santos et al., 2020). Co-culturing of C. albicans and S. mutans on glass microscope slides, farnesol inhibited formation of single and mixed species biofilms (Koo et al., 2003; Fernandes et al., 2016). However, in another study, using an in vitro model of biofilms formed on hydroxyapatite discs coated with saliva, low concentrations of farnesol enhanced S. mutans biofilm formation and GtfB activity (Kim et al., 2017).

In addition to streptococci, the periodontal pathogens Aggregatibacter actinomycetemcomitans and Fusobacterium nucleatum also interact with C. albicans. In vivo, they could excrete the quorum sensing molecule AI-2, which inhibits germination of C. albicans (Bachtiar et al., 2014; Bor et al., 2016).

Interactions of C. albicans and microbiota in the gut

As a commensal dweller of the human gastrointestinal (GI) tract, upon disruption to the host immune system or imbalance of the microbiota, C. albicans can disseminate from the GI tract and cause systemic infections with high mortality (d’Enfert et al., 2021). Within the gut, multiple factors can influence the diversity and density of the microbiota, including environmental parameters such as abundance of oxygen and nutrients, and pH (Donaldson et al., 2016). Abundance of C. albicans is also regulated by the gut microbiota and vice versa. For example, Firmicutes and Bacteroides restrict colonization of C. albicans in the mouse gut by activating transcription factor HIF-1α in intestinal cells, which causes an increase in the antimicrobial peptide LL-37 (Fan et al., 2015). C. albicans and lactic acid bacteria (LAB) have the same metabolic niches throughout the GI tract. Dysbiosis of C. albicans causes altered levels of LAB, especially Lactobacillus spp. and Enterococcus spp. Lactobacilli inhibit C. albicans, while Enterococcus faecalis and C. albicans are mutualistic (Zeise et al., 2021). In an in vitro gut model, Lactobacillus rhamnosus reduced C. albicans hyphal elongation, drove the shedding of hyphae from the surface of epithelial cells and protected against C. albicans induced epithelial damage (Graf et al., 2019).

Effect of broad-spectrum antibiotics on C. albicans

Abuse of broad-spectrum antibiotic treatment depletes gut bacteria and facilitates fungal overgrowth through fungal dysbiosis and increases the risk of disseminated candidiasis (Seelig, 1966a,b; Mason et al., 2012; Dollive et al., 2013). In immunocompromised patients, broad-spectrum antibiotics induced imbalance in the normal bacterial flora is a prerequisite for colonization of C. albicans (Högenauer et al., 1998). Broad-spectrum antibiotics cause overgrowth of C. albicans directly and indirectly. The direct effect is due to the killing of a large proportion of the microbiota, and the indirect effect is due to decreased colonization resistance within the GI-tract (van Ogtrop et al., 1991). Colonization resistance is a term used to describe the capacity of the healthy microbiota to limit the introduction and/or expansion of potential pathogens (Khan et al., 2021). Altered microbiota caused by antibiotic treatment can persist for months, thereby causing long-term decreases in beneficial anaerobic organisms (Sjövall et al., 1986; Lidbeck and Nord, 1993; Orrhage and Nord, 2000), and increases in potential pathogens (Guggenbichler et al., 1985; van der Waaij, 1989; Samonis et al., 1993; Payne et al., 2003). One of the most common side effects of broad-spectrum antibiotic treatment is yeast infections at mucosal sites, including those caused by C. albicans (Giuliano et al., 1987; Samonis et al., 1993; Sullivan et al., 2001).

In addition to directly promoting the overgrowth of C. albicans, antibiotics can also directly alter fungal metabolism. The β-lactam antibiotics, such as cefepime, amoxicillin, and vancomycin, can stimulate the in vitro planktonic growth of C. albicans in 96-well plates and in vivo virulence tested with Caenorhabditis elegans infection model (Aguiar Cordeiro et al., 2018). Cefepime and amoxicillin also directly stimulate C. albicans metabolism and biofilm formation in 96-well plates, thereby enhancing antifungal tolerance (Cordeiro et al., 2019). Gut bacteria treated with β-lactam antibiotics released significant amounts of peptidoglycan fragments, a procedure so called “a peptidoglycan storm” (Tan et al., 2021), which strongly stimulated hyphal formation in C. albicans in vitro and in vivo (Xu et al., 2008; Tan et al., 2021). Hyphal growth is a key virulence factor required for penetration of mucosal barriers (Lo et al., 1997). The altered gut metabolites caused by antibiotic treatment include primary and secondary bile acids. The primary bile acid taurocholic acid promoted growth and induces the expression of hyphae-specific genes ECE1, UME6, HWP1, and HCG1, thereby enhancing hyphal formation of C. albicans in vitro (Hsieh et al., 2017; Guinan and Thangamani, 2018). In contrast, the secondary bile acids (lithocholic acid and deoxycholic acid) inhibited C. albicans growth, adherence, hyphae and biofilm formation in vitro in 96-well plates (Guinan et al., 2018). Cefoperazone treatment caused an increased level of the primary bile acid taurocholic acid in mice gut. In addition, cefoperazone treatment changed the microbiome at both phyla-level and family level in mice gut. It caused a higher abundance of Firmicutes and lower abundance of Bacteroidetes and Verrucomicrobia, a higher abundance of Panibacillaceae and lower abundance of Lactobacillaceae, Turicibacteraceae, Clostridiales, and Clostridiaceae. Cefoperazone treatment also altered the ability of C. albicans to modify the mice gut bacterial microbiota (Gutierrez et al., 2020).

Relationship between overgrowth of C. albicans and diseases

Overgrowth of C. albicans in the gut may lead to health problems, such as digestive problems (gas and diarrhea), and psychological disorders (anxiety, memory loss, depression) (Severance et al., 2016; Markey et al., 2020). In high-risk patients, C. albicans might translocate from the GI tract to extra-intestinal organs, especially the liver, the spleen, and the bloodstream. The strain causing systemic candidiasis and the strain identified from the same patient’s rectum sample are always the same, although in some cases, catheters are another source of infection (Miranda et al., 2009). C. albicans in the GI tract may also be an immunogen which triggers or promotes a variety of hypersensitivity diseases (Goldman and Huffnagle, 2009). However, the direct evidence of C. albicans associated diseases is still lacking (Li et al., 2018b,a; Kapitan et al., 2019). Reduced growth of C. albicans can also cause diseases. In mice, reduced growth of C. albicans from treatment with the antifungal drug fluconazole increased the immune response and severity of experimentally induced colitis and induced allergic airway disease (Wheeler et al., 2016; Li et al., 2018b).

Interaction of C. albicans with bacteria in the blood

Invasive candidiasis is a global health threat with high morbidity and mortality, as well as costly and lengthy hospital stays (Morgan et al., 2005; Pfaller and Diekema, 2007; Horn et al., 2009; Wu et al., 2017). Candidemia is the most common form of invasive candidiasis (Magill et al., 2014, 2018; Ricotta et al., 2020). C. albicans is the most frequent fungal pathogen and Staphylococcus spp. are the most frequent bacterial pathogens isolated from blood stream infections worldwide. In approximately 20% of all C. albicans bloodstream infections, C. albicans was co-isolated with Staphylococcus epidermidis or Staphylococcus aureus (Klotz et al., 2007; O’Donnell et al., 2015; Reno et al., 2015).

Staphylococcus epidermidis co-cultured in polystyrene tubes with C. albicans enhanced adhesion and biofilm formation of C. albicans (El-Azizi et al., 2004). Formed on optical microwell Petri dishes, the dual species biofilms of C. albicans and S. epidermidis were sicker than single species biofilms and had more dissemination of S. epidermidis in vivo in mouse model of subcutaneous catheter biofilm infection (Pammi et al., 2013).

Physical interactions of C. albicans with S. aureus and S. epidermidis involve C. albicans hyphae protein Als1p, Als3p, and cell surface O-linked mannosylations. Als3p, which is expressed exclusively in the hyphae, is the receptor for binding of S. aureus and S. epidermidis (Peters et al., 2010, 2012; Beaussart et al., 2013). Als1p and cell surface O-linked mannosylations can bind specific peptide ligands on surface of S. epidermidis (Beaussart et al., 2013). The physical interaction is required for bacterial invasion. The association of S. aureus with ALS3—expressing C. albicans in the oral cavity resulted in S. aureus bacteremia and the isolation of S. aureus bacteria from kidney tissue (Schlecht et al., 2015). C. albicans hyphae is highly immunogenic (Ballou et al., 2016). It attracts phagocytic cells, which rapidly engulf the adherent S. aureus, and subsequently migrate to cervical lymph nodes, from where S. aureus disseminate into the internal organs and cause systemic infections (Allison et al., 2019). Both organisms benefit from this interaction, which modulates the host immune response differently than monospecies infections (Peters and Noverr, 2013). In a Galleria mellonella larvae disseminated infection model, co-infection with C. albicans and S. aureus resulted in a reduced survival rate compared to a monospecies infection (Sheehan et al., 2020). In the murine model of intra-abdominal infection, C. albicans raised the pH during polymicrobial growth. The elevated pH drove the expression of the accessory gene regulator quorum sensing system, thereby increased alpha-toxin production. Thus, coinfection of C. albicans and S. aureus enhanced the virulence of S. aureus (Todd et al., 2019a,b).

Farnesol prevents biofilm formation of C. albicans on the surface of microtiter plates, and it prevents biofilm formation of S. aureus in polystyrene plates (Ramage et al., 2002; Jabra-Rizk et al., 2006). But the effect of farnesol on biofilm formation of S. aureus is dose-dependent: at low concentration, farnesol increases biofilm formation, but at high concentration, it inhibits biofilm formation. Whereas tyrosol, another quorum sensing molecule of C. albicans, has no effect on S. aureus biofilm formation in 12-well plates (Krause et al., 2015). However, prostaglandin E2, produced by C. albicans from external arachidonic acid, stimulated S. aureus biofilm formation in the mixed biofilm in 12-well plates (Krause et al., 2015). In addition, in the mixed biofilm formed in a mouse biofilm infection model using subcutaneous catheter, the bacterial repressor of autolysis (the lrg operon) was down-regulated, thereby the bacterial autolysis was enhanced. Bacterial autolysis is a procedure of self-digestion of the cell wall mediated by the peptidoglycan hydrolase autolysins (Lewis, 2000). The down-regulation might be the cause of enhanced extracellular DNA (eDNA) levels in this condition (Pammi et al., 2013). Autolysis of S. epidermidis results in release of eDNA into the extracellular matrix. In addition, C. albicans can actively secrete eDNA. eDNA could enhance adhesion and biofilm formation (Pammi et al., 2013). Furthermore, in the mixed biofilm formed in 96-well flat-bottom plates, C. albicans could enhance the pathogenicity of S. aureus through increasing tolerance to antimicrobials. Farnesol induced bursts of reactive oxygen species, which triggered the expression of efflux pumps in S. aureus (Kong et al., 2017; Vila et al., 2019). In addition, in the mixed biofilm matric, C. albicans could secrete β-1,3-glucan, which prevented penetration of drugs and enhances tolerance to antimicrobial drugs (Kong et al., 2016).

Interactions between C. albicans and microbiota in the vagina

Many females suffer from vulvovaginal infections (VVI). Approximately 75% of women develop VVC at least once in their lifetime (Sobel, 2007). Mixed vaginal infections represent >20% of women with VVI (Deidda et al., 2016). Bacterial Vaginosis (BV), Vulvovaginal Candidiasis (VVC) and Trichomoniasis (TV) are the three most prevalent VVI (Kalia et al., 2017, 2018, 2019). Recurrent vulvovaginal candidiasis (RVVC) afflicts about 8% of women globally (Denning et al., 2018). VVC is usually caused by the overgrowth of the Candida spp. (De Gregorio et al., 2020). Until recently, C. albicans was the species most detected in VVC. However, during the last two decades, non-albicans Candida (NAC) species have emerged increasingly as causative species of VVC (Rodríguez-Cerdeira et al., 2019).

Candida is a commensal dweller of the genitourinary tract with colonization rates of 11.6–17% (Andrioli et al., 2009). The composition of the vaginal microbiota is dynamic and is subject to regulation by physical conditions such as menstruation, pregnancy, and health status (Greenbaum et al., 2019). However, multiple factors can cause fungal overgrowth in the vagina. These factors include wide-spectrum antibiotic use, immunosuppressive therapy, and alterations of the vaginal conditions due to, for example, changes of nutrients or fluctuations of pH (Nobile and Johnson, 2015; Romo and Kumamoto, 2020). As an opportunistic pathogen, C. albicans can cause cutaneous and mucocutaneous candidiasis inside the vaginal. Once disseminated into inner organs or the bloodstream, C. albicans can also cause life-threatening invasive infections (Hube, 2004; Hall and Noverr, 2017).

Interactions between C. albicans and group B Streptococcus

Group B Streptococcus (GBS) is usually found in human rectum and vagina. It is a leading cause of neonatal sepsis, pneumonia, and meningitis worldwide and is the leading cause of death among newborns in developed countries. GBS can be vertically transmitted from mother to neonate. Therefore, maternal vaginal colonization is a key risk factor for neonatal disease caused by GBS (Chen et al., 2018). In vitro data indicated that, the GBS antigen I/II family adhesins directly bind to the vaginal epithelium (Rego et al., 2016), and directly bind to Als3p of C. albicans, thereby enhancing association of C. albicans with vaginal epithelium (Pidwill et al., 2018).

Interactions between C. albicans and lactobacilli

Lactobacillus spp. are the major bacteria in the vaginal microbiota of healthy women. Lactobacillus spp. prevents the vagina from urogenital and sexually transmitted infections via secreting metabolites such as organic acids, hydrogen peroxide (H2O2), bacteriocins, and biosurfactants (Nader-Macías and Juárez Tomás, 2015; Mendling, 2016; Wang et al., 2017; Fuochi et al., 2019; Linhares et al., 2019). Organic acids secreted by Lactobacillus spp. are the major cause of acidic pH (4–4.5) in the vaginal environment, and are the inhibitive factor of pathogen growth (Miller et al., 2016). H2O2-generating Lactobacillus spp. are in the vagina of approximately 96% of healthy women, while their populations are lower in women with VVI (Boris and Barbés, 2000). Some bacteria, such as L. fermentum (Pascual et al., 2008), Streptococcus sanguinis (Ma et al., 2015), E. faecalis (Graham et al., 2017; De Cesare et al., 2021) are capable of producing bacteriocins that inhibit growth, yeast-to-hyphae switch, biofilm formation, and virulence of C. albicans. Bacteriocins are antimicrobial peptides or proteins produced by bacteria (Benítez-Chao et al., 2021). Biosurfactants are surface-active biomolecules produced by microorganisms (Healy et al., 1996). Biosurfactants are important mediators of cell adherence and desorption from surfaces (Desai and Banat, 1997). Lactobacillus-derived biosurfactants can alter the hydrophobicity and electrical properties of interfaces, thereby inhibiting fungal adhesion and biofilm formation (De Gregorio et al., 2020; Zeise et al., 2021).

Lactobacillus crispatus is the dominant Lactobacillus spp. in healthy vagina (Ceccarani et al., 2019). In vitro tests indicated the organic acids and H2O2 produced by L. crispatus can strongly inhibit C. albicans growth and hyphal formation. In addition to lactate, in vitro tests indicated L. crispatus also produced other anti-Candida small molecules, which inhibited growth and hyphal morphogenesis of C. albicans. L. crispatus also down-regulated the expression of ALS3, HWP1, and ECE1, thereby limiting the virulence of C. albicans (Wang et al., 2017). Resistance to VVC increased when L. crispatus was the dominant bacteria community in the vaginal environment (Jang et al., 2019). L. casei also has strong anti-Candida effect. L. casei co-cultured in 96-well flat-bottom polystyrene plates with C. albicans or C. tropicalis reduced the formation of fungal hyphae and early biofilms (Sobel, 2016; Paniágua et al., 2021). In contrast, in vitro, L. iners up-regulated expression of HWP1 and ECE1, induced hyphal growth and increased both the biofilm biomass and metabolic activity of C. albicans. Furthermore, L. iners can increase C. albicans virulence via transforming C. albicans isolates with moderate or weak biofilm-forming ability to strong biofilm producers (Sabbatini et al., 2021).

Lactobacillus spp. also inhibits C. albicans by competing for adhesion sites in vitro (Allonsius et al., 2017). C. albicans invades vaginal epithelial cells mainly through endocytosis and active penetration. Lactobacillus spp. such as L. crispatus and L. plantarum can specifically adhere to the vaginal tissue, form biofilm and inhibit adhesion of C. albicans by several mechanisms, including competition for receptors, displacement of adhered pathogenic microorganisms, and avoiding their re-adhesion (Boris et al., 1998; Coudeyras et al., 2008; Borges et al., 2014; De Seta et al., 2014).

Future directions

In addition to the above-mentioned interplays in the microbiota, we think the following directions deserve further investigations in the future.

Effect of anti-fungal use on the microbiota

Many antibiotics can disrupt the microbiota equilibrium and eliminate “good” gut bacteria which inhibit fungal growth. Therefore, abuse of antibiotics promotes fungal overgrowth and pathogenicity. Few studies have looked at the effect of anti-fungal use on the microbiota. Some studies indicate fungal depletion due to application of antifungal drugs is associated with altered microbiome and some diseases. For example, in one study, mice treated with antifungal drug fluconazole had severe dextran sulfate sodium-induced colitis (DSS-colitis), while mice treated with antibiotics had less severe DSS-colitis. One hypothesis was fluconazole caused the deletion of commensal fungi in the gut. The depletion led to overgrowth of pathogenic bacteria, which exacerbated colitis (Qiu et al., 2015b). Similarly, in another study, treatment of mice with fluconazole caused more aggravated DSS-induced acute colitis, as well as T cell transfer-mediated chronic colitis, and house dust mite-induced intratracheal allergic airway disease (Iliev et al., 2012; Qiu et al., 2015b; Wheeler et al., 2016). Further investigations indicated that fluconazole caused dysbiosis of both mycobiota and bacteriobiota. Among the mycobiota, Candida spp. were decreased but other fungi, such as Aspergillus, Wallemia, and Epicoccum, were increased. Among the bacteriobiota, the overall diversity remained unchanged, but Bacteroides, Allobaculum, Clostridium, Desulfovibrio, and Lactobacillus spp. had decreased relative detection, while Anaerostipes, Coprococcus, and Streptococcus had increased relative detection (Wheeler et al., 2016)

Therapy against mixed biofilms

Both singularly or co-existed, fungi and bacteria usually form biofilms in the human body. Forming of mixed species biofilms protects both bacteria and fungi from external assaults, such as host clearance and antimicrobial agents, and facilitates intra- and inter-species interactions (Levison and Pitsakis, 1987; Shirtliff et al., 2009). For example, it was found that mixed species biofilms of C. albicans and oral streptococci were more resistant to antibiotics than single species biofilms (Morales and Hogan, 2010). The most common mixed fungal infections are caused by mixing of C. albicans with S. aureus, P. aeruginosa or Escherichia coli (Dhamgaye et al., 2016; Kalan and Grice, 2018). Mixed infections of two or three Candida spp. are also found in patients with orogastric cancer (de Sousa et al., 2016). The overall prevalence of fungal mixed fungal infections almost doubled over the 10-year period, with 10.3% of all infections in 2015 compared to 5.7% in 2006 (Gawaz and Weisel, 2018). However, traditional therapies against microbial infections generally target individual causative agents. Considerations for efficacy on a polymicrobial cause or on individual members of microbial communities are largely lacking. Treatment of mixed species infections should be the combinations of antimicrobials, and the drugs of choice should be synergistic. Potential antagonism between drugs and undesirable side effects of the drugs also needs to be taken into consideration.

Restoring microbiota equilibrium

In addition to candidiasis, imbalanced microbiota has been linked directly and indirectly to various human diseases including mental illnesses (Clapp et al., 2017), autoimmune diseases (Xu et al., 2019), and chronic diseases such as rheumatoid arthritis and asthma (Vijay and Valdes, 2022). Numerous approaches have been developed with the purpose of rehabilitating the perturbed microbiota, such as the administration of probiotics, prebiotics, and synbiotics, fecal microbiota transplantation, phages, predatory bacteria (Gagliardi et al., 2018; Gomaa, 2020; Sestito et al., 2020).

Probiotics can be used to prevent the onset of dysbiosis and to restore the balance in the dysbiosis (Gomaa, 2020). Vaginal administration of probiotics is a promising therapy of VVC. In VVC patients, probiotics could potentiate the efficiency of azoles (Kovachev and Vatcheva-Dobrevska, 2015). L. acidophilus, L. rhamnosus, and L. reuteri are the most-researched strains of probiotics for the purpose of restoring and maintaining vaginal microbiota equilibrium (Reid et al., 2001, 2003). Probiotics can stick to vaginal surfaces and inhibit growth of harmful bacteria on vaginal surfaces. Lactobacillus may also adhere directly to harmful bacteria and prevent them from spreading (Maudsdotter et al., 2011). In addition, L. rhamnosus GG can interfere with hyphal formation of C. albicans via production of exopolysaccharides and chitinases (Allonsius et al., 2017, 2019). In addition to probiotic bacteria, some yeasts are also potential probiotics. For example, both live and inactivated Saccharomyces cerevisiae can co-aggregate with C. albicans and inhibit adherence of C. albicans to epithelial cells. S. cerevisiae can also inhibit the switch from yeast to mycelial form, production of aspartyl proteases (SAP) in C. albicans (Pericolini et al., 2017). Another well characterized probiotic yeast, Saccharomyces boulardii, is often used to alleviate GI tract disorders. It also inhibits adhesion of C. albicans to and biofilm formation on polystyrene surfaces (Krasowska et al., 2009).

Fecal microbiota transplantation (FMT) can restore the compositions and functions of gut microbiota. It has been successfully used for the treatment of several intestinal diseases, such as Clostridium difficile infection, ulcerative colitis, and Crohn’s disease (Tan et al., 2020). Autologous FMT has the potential of restoring a pre-antibiotic microbiome baseline after disruption (Bulow et al., 2018; Suez et al., 2018; Taur et al., 2018). The presence of C. albicans in the donor stool decreases the efficacy of FMT, suggesting the outcome of FMT is largely affected by fungal dysbiosis (Zuo et al., 2018). FMT of commensal bacteria prevents C. albicans colonization in the gut of mice (Matsuo et al., 2019), and serial FMT of C. albicans in mice treated with antibiotics results in mutualistic interaction of C. albicans and the host: C. albicans become a “better” commensal with much lower virulence due to loss-of-function of FLO8, which resulted in failure of filamentation and less damage to host cells. The mouse gut-evolved C. albicans strains raised trained protective immune responses, and the host was protected against systemic candidiasis (Tso et al., 2018).

Phages account for approximately 90% of the human virome. They can modulate the composition the microbiota at least in part due to their antimicrobial activity (Scarpellini et al., 2015). Predatory bacteria can eat other bacteria or yeasts by actively hunting and consuming their biomass as nutrients (Rossen et al., 2015). In the gut where Gram-negative bacteria predominates, predatory bacteria can be used to restore gut dysbiosis in which Gram-negative bacteria predominates (Atterbury et al., 2011).

Conclusion

In recent years, due to the treatments for organ transplantations, malignant diseases and HIV/AIDS, and the advances in intensive care unit (ICU) interventions, opportunistic infections are increasing. Candida spp. are among the leading pathogens (Pfaller and Diekema, 2007). Colonization is a prerequisite for subsequent infection. Candida spp. is a commensal organism in up to 60% of healthy individuals (Kullberg and Arendrup, 2015; McCarty and Pappas, 2016). The shift from a commensal to pathogen is regulated by multifactorial mechanisms, mainly the interplay with the host immune defense and with the microbiota. Uncovering the molecular mechanism of the interactions between C. albicans and the resident microbiota will largely facilitate the understanding of the etiology and the searching for novel therapeutic options of fungal as well as bacterial infections. Furthermore, the effect of antifungals on the microbiota also deserves more investigations. New approaches such as drug combination therapy against polymicrobial infections need to be developed. Finally, several therapies including probiotics and FMT are under development with the aim of restoring the microbiota equilibrium.

Author contributions

FY, Y-yJ, and T-hY: conceptualization. HL, M-xM, C-lJ, and Y-bC: writing—original draft preparation. HL, Y-yJ, T-hY, and FY: writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by National Natural Science Foundation of China (No. 82020108032) and the Innovation Program of Shanghai Municipal Education Commission (No. 202101070007E00094) to Y-yJ. National Natural Science Foundation of China (No. 81872910) and Shanghai Key Basic Research Project (No. 19JC1414900) to Y-bC.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Abu-Elteen, K. H., and Abu-Alteen, R. M. (1998). The prevalence of Candida albicans populations in the mouths of complete denture wearers. New Microbiol. 21, 41–48.

PubMed Abstract | Google Scholar

Aguiar Cordeiro, R., de Jesus Evangelista, A. J., Serpa, R., Colares de Andrade, A. R., Leite Mendes, P. B., et al. (2018). beta-lactam antibiotics & vancomycin increase the growth & virulence of Candida spp. Future Microbiol. 13, 869–875. doi: 10.2217/fmb-2018-2019

CrossRef Full Text | Google Scholar

Allison, D. L., Scheres, N., Willems, H. M. E., Bode, C. S., Krom, B. P., and Shirtliff, M. E. (2019). The host immune system facilitates disseminated Staphylococcus aureus disease due to phagocytic attraction to Candida albicans during coinfection: a case of bait and switch. Infect. Immun. 87:e00137-19. doi: 10.1128/iai.00137-119

CrossRef Full Text | Google Scholar

Allonsius, C. N., van den Broek, M. F. L., De Boeck, I., Kiekens, S., Oerlemans, E. F. M., Kiekens, F., et al. (2017). Interplay between Lactobacillus rhamnosus GG and Candida and the involvement of exopolysaccharides. Microb Biotechnol. 10, 1753–1763. doi: 10.1111/1751-7915.12799

PubMed Abstract | CrossRef Full Text | Google Scholar

Allonsius, C. N., Vandenheuvel, D., Oerlemans, E. F. M., Petrova, M. I., Donders, G. G. G., Cos, P., et al. (2019). Inhibition of Candida albicans morphogenesis by chitinase from Lactobacillus rhamnosus GG. Sci. Rep. 9:2900. doi: 10.1038/s41598-019-39625-39620

CrossRef Full Text | Google Scholar

Andrioli, J. L., Oliveira, G. S., Barreto, C. S., Sousa, Z. L., Oliveira, M. C., Cazorla, I. M., et al. (2009). [Frequency of yeasts in vaginal fluid of women with and without clinical suspicion of vulvovaginal candidiasis]. Rev. Bras Ginecol. Obstet 31, 300–304. doi: 10.1590/s0100-72032009000600006

PubMed Abstract | CrossRef Full Text | Google Scholar

Atterbury, R. J., Hobley, L., Till, R., Lambert, C., Capeness, M. J., Lerner, T. R., et al. (2011). Effects of orally administered Bdellovibrio bacteriovorus on the well-being and Salmonella colonization of young chicks. Appl. Environ. Microbiol. 77, 5794–5803. doi: 10.1128/aem.00426-411

CrossRef Full Text | Google Scholar

Bachtiar, E. W., Bachtiar, B. M., Jarosz, L. M., Amir, L. R., Sunarto, H., Ganin, H., et al. (2014). AI-2 of Aggregatibacter actinomycetemcomitans inhibits Candida albicans biofilm formation. Front. Cell Infect. Microbiol. 4:94. doi: 10.3389/fcimb.2014.00094

PubMed Abstract | CrossRef Full Text | Google Scholar

Ballou, E. R., Avelar, G. M., Childers, D. S., Mackie, J., Bain, J. M., Wagener, J., et al. (2016). Lactate signalling regulates fungal beta-glucan masking and immune evasion. Nat. Microbiol. 2:16238. doi: 10.1038/nmicrobiol.2016.238

PubMed Abstract | CrossRef Full Text | Google Scholar

Bamford, C. V., d’Mello, A., Nobbs, A. H., Dutton, L. C., Vickerman, M. M., and Jenkinson, H. F. (2009). Streptococcus gordonii modulates Candida albicans biofilm formation through intergeneric communication. Infect. Immun. 77, 3696–3704. doi: 10.1128/iai.00438-439

CrossRef Full Text | Google Scholar

Bamford, C. V., Nobbs, A. H., Barbour, M. E., Lamont, R. J., and Jenkinson, H. F. (2015). Functional regions of Candida albicans hyphal cell wall protein Als3 that determine interaction with the oral bacterium Streptococcus gordonii. Microbiology (Reading) 161, 18–29. doi: 10.1099/mic.0.083378-83370

CrossRef Full Text | Google Scholar

Barbosa, J. O., Rossoni, R. D., Vilela, S. F., de Alvarenga, J. A., Velloso Mdos, S., Prata, M. C., et al. (2016). Streptococcus mutans can modulate biofilm formation and attenuate the virulence of Candida albicans. PLoS One 11:e0150457. doi: 10.1371/journal.pone.0150457

PubMed Abstract | CrossRef Full Text | Google Scholar

Beaussart, A., Herman, P., El-Kirat-Chatel, S., Lipke, P. N., Kucharikova, S., Van Dijck, P., et al. (2013). Single-cell force spectroscopy of the medically important Staphylococcus epidermidis-Candida albicans interaction. Nanoscale 5, 10894–10900. doi: 10.1039/c3nr03272h

PubMed Abstract | CrossRef Full Text | Google Scholar

Benítez-Chao, D. F., León-Buitimea, A., Lerma-Escalera, J. A., and Morones-Ramírez, J. R. (2021). Bacteriocins: an overview of antimicrobial, toxicity, and biosafety assessment by in vivo models. Front. Microbiol. 12:630695. doi: 10.3389/fmicb.2021.630695

PubMed Abstract | CrossRef Full Text | Google Scholar

Bertolini, M., Ranjan, A., Thompson, A., Diaz, P. I., Sobue, T., Maas, K., et al. (2019). Candida albicans induces mucosal bacterial dysbiosis that promotes invasive infection. PLoS Pathog 15:e1007717. doi: 10.1371/journal.ppat.1007717

PubMed Abstract | CrossRef Full Text | Google Scholar

Bhardwaj, R. G., Ellepolla, A., Drobiova, H., and Karched, M. (2020). Biofilm growth and IL-8 & TNF-alpha-inducing properties of Candida albicans in the presence of oral gram-positive and gram-negative bacteria. BMC Microbiol. 20:156. doi: 10.1186/s12866-020-01834-1833

CrossRef Full Text | Google Scholar

Bor, B., Cen, L., Agnello, M., Shi, W., and He, X. (2016). Morphological and physiological changes induced by contact-dependent interaction between Candida albicans and Fusobacterium nucleatum. Sci. Rep. 6:27956. doi: 10.1038/srep27956

PubMed Abstract | CrossRef Full Text | Google Scholar

Borges, S., Silva, J., and Teixeira, P. (2014). The role of lactobacilli and probiotics in maintaining vaginal health. Arch. Gynecol. Obstet. 289, 479–489. doi: 10.1007/s00404-013-3064-3069

CrossRef Full Text | Google Scholar

Boris, S., and Barbés, C. (2000). Role played by lactobacilli in controlling the population of vaginal pathogens. Microbes Infect. 2, 543–546. doi: 10.1016/s1286-4579(00)00313-310

CrossRef Full Text | Google Scholar

Boris, S., Suárez, J. E., Vázquez, F., and Barbés, C. (1998). Adherence of human vaginal lactobacilli to vaginal epithelial cells and interaction with uropathogens. Infect. Immun. 66, 1985–1989. doi: 10.1128/iai.66.5.1985-1989.1998

PubMed Abstract | CrossRef Full Text | Google Scholar

Bowen, W. H., and Koo, H. (2011). Biology of Streptococcus mutans-derived glucosyltransferases: role in extracellular matrix formation of cariogenic biofilms. Caries Res. 45, 69–86. doi: 10.1159/000324598

PubMed Abstract | CrossRef Full Text | Google Scholar

Brandt, M. E., and Lockhart, S. R. (2012). Recent taxonomic developments with candida and other opportunistic yeasts. Curr. Fungal Infect. Rep. 6, 170–177. doi: 10.1007/s12281-012-0094-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Bulow, C., Langdon, A., Hink, T., Wallace, M., Reske, K. A., Patel, S., et al. (2018). Impact of amoxicillin-clavulanate followed by autologous fecal microbiota transplantation on fecal microbiome structure and metabolic potential. mSphere 3:e00588-18. doi: 10.1128/mSphereDirect.00588-518

CrossRef Full Text | Google Scholar

Byadarahally Raju, S., and Rajappa, S. (2011). Isolation and identification of Candida from the oral cavity. ISRN Dent. 2011:487921. doi: 10.5402/2011/487921

PubMed Abstract | CrossRef Full Text | Google Scholar

Cavalcanti, I. M., Del Bel Cury, A. A., Jenkinson, H. F., and Nobbs, A. H. (2017). Interactions between Streptococcus oralis, Actinomyces oris, and Candida albicans in the development of multispecies oral microbial biofilms on salivary pellicle. Mol. Oral Microbiol. 32, 60–73. doi: 10.1111/omi.12154

PubMed Abstract | CrossRef Full Text | Google Scholar

Ceccarani, C., Foschi, C., Parolin, C., D’Antuono, A., Gaspari, V., Consolandi, C., et al. (2019). Diversity of vaginal microbiome and metabolome during genital infections. Sci. Rep. 9:14095. doi: 10.1038/s41598-019-50410-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Z., Wu, C., Cao, X., Wen, G., Guo, D., Yao, Z., et al. (2018). Risk factors for neonatal group B streptococcus vertical transmission: a prospective cohort study of 1815 mother-baby pairs. J. Perinatol. 38, 1309–1317. doi: 10.1038/s41372-018-0182-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Chinnici, J., Yerke, L., Tsou, C., Busarajan, S., Mancuso, R., Sadhak, N. D., et al. (2019). Candida albicans cell wall integrity transcription factors regulate polymicrobial biofilm formation with Streptococcus gordonii. PeerJ 7:e7870. doi: 10.7717/peerj.7870

PubMed Abstract | CrossRef Full Text | Google Scholar

Clapp, M., Aurora, N., Herrera, L., Bhatia, M., Wilen, E., and Wakefield, S. (2017). Gut microbiota’s effect on mental health: the gut-brain axis. Clin. Pract. 7:987. doi: 10.4081/cp.2017.987

PubMed Abstract | CrossRef Full Text | Google Scholar

Cordeiro, R. A., Evangelista, A. J. J., Serpa, R., de Andrade, A. R. C., Mendes, P. B. L., de Oliveira, J. S., et al. (2019). Cefepime and amoxicillin increase metabolism and enhance caspofungin tolerance of Candida albicans biofilms. Front. Microbiol. 10:1337. doi: 10.3389/fmicb.2019.01337

PubMed Abstract | CrossRef Full Text | Google Scholar

Coudeyras, S., Jugie, G., Vermerie, M., and Forestier, C. (2008). Adhesion of human probiotic Lactobacillus rhamnosus to cervical and vaginal cells and interaction with vaginosis-associated pathogens. Infect. Dis. Obstet. Gynecol. 2008:549640. doi: 10.1155/2008/549640

PubMed Abstract | CrossRef Full Text | Google Scholar

De Cesare, G. B., Chebaro, Y., Guha, S., Cruz, M., Garsin, D., and Lorenz, M. (2021). Characterization of the mechanism of action of the E. faecalis bacteriocin EntV on C. albicans. Access Microbiology. 3:0153. doi: 10.1099/acmi.cc2021.po0153

CrossRef Full Text | Google Scholar

De Gregorio, P. R., Parolin, C., Abruzzo, A., Luppi, B., Protti, M., Mercolini, L., et al. (2020). Biosurfactant from vaginal Lactobacillus crispatus BC1 as a promising agent to interfere with Candida adhesion. Microb Cell Fact. 19:133. doi: 10.1186/s12934-020-01390-1395

CrossRef Full Text | Google Scholar

De Seta, F., Parazzini, F., De Leo, R., Banco, R., Maso, G. P., De Santo, D., et al. (2014). Lactobacillus plantarum P17630 for preventing Candida vaginitis recurrence: a retrospective comparative study. Eur. J. Obstet. Gynecol. Reprod. Biol. 182, 136–139. doi: 10.1016/j.ejogrb.2014.09.018

PubMed Abstract | CrossRef Full Text | Google Scholar

de Sousa, L., Santos, V. L., de Souza Monteiro, A., Dias-Souza, M. V., Marques, S. G., de Faria, E. S., et al. (2016). Isolation and identification of Candida species in patients with orogastric cancer: susceptibility to antifungal drugs, attributes of virulence in vitro and immune response phenotype. BMC Infect. Dis. 16:86. doi: 10.1186/s12879-016-1431-1434

CrossRef Full Text | Google Scholar

DeGruttola, A. K., Low, D., Mizoguchi, A., and Mizoguchi, E. (2016). Current understanding of dysbiosis in disease in human and animal models. Inflamm. Bowel Dis. 22, 1137–1150. doi: 10.1097/mib.0000000000000750

PubMed Abstract | CrossRef Full Text | Google Scholar

Deidda, F., Amoruso, A., Allesina, S., Pane, M., Graziano, T., Del Piano, M., et al. (2016). In vitro activity of Lactobacillus fermentum LF5 against different candida species and Gardnerella vaginalis: a new perspective to approach mixed vaginal infections? J. Clin. Gastroenterol. 2015, S168–S170. doi: 10.1097/mcg.0000000000000692

PubMed Abstract | CrossRef Full Text | Google Scholar

d’Enfert, C., Kaune, A. K., Alaban, L. R., Chakraborty, S., Cole, N., Delavy, M., et al. (2021). The impact of the Fungus-Host-Microbiota interplay upon Candida albicans infections: current knowledge and new perspectives. FEMS Microbiol. Rev. 45:fuaa060. doi: 10.1093/femsre/fuaa060

PubMed Abstract | CrossRef Full Text | Google Scholar

Denning, D. W., Kneale, M., Sobel, J. D., and Rautemaa-Richardson, R. (2018). Global burden of recurrent vulvovaginal candidiasis: a systematic review. Lancet Infect. Dis. 18, e339–e347. doi: 10.1016/S1473-3099(18)30103-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Desai, J. D., and Banat, I. M. (1997). Microbial production of surfactants and their commercial potential. Microbiol. Mol. Biol. Rev. 61, 47–64.

Google Scholar

Dhamgaye, S., Qu, Y., and Peleg, A. Y. (2016). Polymicrobial infections involving clinically relevant Gram-negative bacteria and fungi. Cell Microbiol. 18, 1716–1722. doi: 10.1111/cmi.12674

PubMed Abstract | CrossRef Full Text | Google Scholar

Diaz, P. I., Strausbaugh, L. D., and Dongari-Bagtzoglou, A. (2014). Fungal-bacterial interactions and their relevance to oral health: linking the clinic and the bench. Front. Cell Infect. Microbiol. 4:101. doi: 10.3389/fcimb.2014.00101

PubMed Abstract | CrossRef Full Text | Google Scholar

Diaz, P. I., Xie, Z., Sobue, T., Thompson, A., Biyikoglu, B., Ricker, A., et al. (2012). Synergistic interaction between Candida albicans and commensal oral streptococci in a novel in vitro mucosal model. Infect. Immun. 80, 620–632. doi: 10.1128/iai.05896-5811

CrossRef Full Text | Google Scholar

Dollive, S., Chen, Y. Y., Grunberg, S., Bittinger, K., Hoffmann, C., Vandivier, L., et al. (2013). Fungi of the murine gut: episodic variation and proliferation during antibiotic treatment. PLoS One 8:e71806. doi: 10.1371/journal.pone.0071806

PubMed Abstract | CrossRef Full Text | Google Scholar

Donaldson, G. P., Lee, S. M., and Mazmanian, S. K. (2016). Gut biogeography of the bacterial microbiota. Nat. Rev. Microbiol. 14, 20–32. doi: 10.1038/nrmicro3552

PubMed Abstract | CrossRef Full Text | Google Scholar

Dos Santos, J. D., Fugisaki, L. R. O., Medina, R. P., Scorzoni, L., Alves, M. S., de Barros, P. P., et al. (2020). Streptococcus mutans secreted products inhibit candida albicans induced oral candidiasis. Front. Microbiol. 11:1605. doi: 10.3389/fmicb.2020.01605

PubMed Abstract | CrossRef Full Text | Google Scholar

Dufresne, S. F., Cole, D. C., Denning, D. W., and Sheppard, D. C. (2017). Serious fungal infections in Canada. Eur. J. Clin. Microbiol. Infect. Dis. 36, 987–992. doi: 10.1007/s10096-017-2922-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Dutton, L. C., Nobbs, A. H., Jepson, K., Jepson, M. A., Vickerman, M. M., Aqeel Alawfi, S., et al. (2014). O-mannosylation in Candida albicans enables development of interkingdom biofilm communities. mBio 5:e00911-14. doi: 10.1128/mBio.00911-914

CrossRef Full Text | Google Scholar

El-Azizi, M. A., Starks, S. E., and Khardori, N. (2004). Interactions of Candida albicans with other Candida spp. and bacteria in the biofilms. J. Appl. Microbiol. 96, 1067–1073. doi: 10.1111/j.1365-2672.2004.02213.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Ellepola, K., Liu, Y., Cao, T., Koo, H., and Seneviratne, C. J. (2017). Bacterial GtfB augments Candida albicans accumulation in cross-kingdom biofilms. J. Dent. Res. 96, 1129–1135. doi: 10.1177/0022034517714414

PubMed Abstract | CrossRef Full Text | Google Scholar

Ellepola, K., Truong, T., Liu, Y., Lin, Q., Lim, T. K., Lee, Y. M., et al. (2019). Multi-omics analyses reveal synergistic carbohydrate metabolism in Streptococcus mutans-Candida albicans mixed-species biofilms. Infect. Immun. 87:e00339-19. doi: 10.1128/IAI.00339-319

CrossRef Full Text | Google Scholar

El-Sayed, A., Aleya, L., and Kamel, M. (2021). Microbiota’s role in health and diseases. Environ. Sci. Pollut. Res. Int. 28, 36967–36983. doi: 10.1007/s11356-021-14593-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Falsetta, M. L., Klein, M. I., Colonne, P. M., Scott-Anne, K., Gregoire, S., Pai, C. H., et al. (2014). Symbiotic relationship between Streptococcus mutans and Candida albicans synergizes virulence of plaque biofilms in vivo. Infect. Immun. 82, 1968–1981. doi: 10.1128/iai.00087-14

PubMed Abstract | CrossRef Full Text | Google Scholar

Fan, D., Coughlin, L. A., Neubauer, M. M., Kim, J., Kim, M. S., Zhan, X., et al. (2015). Activation of HIF-1alpha and LL-37 by commensal bacteria inhibits Candida albicans colonization. Nat. Med. 21, 808–814. doi: 10.1038/nm.3871

PubMed Abstract | CrossRef Full Text | Google Scholar

Fanello, S., Bouchara, J. P., Sauteron, M., Delbos, V., Parot, E., Marot-Leblond, A., et al. (2006). Predictive value of oral colonization by Candida yeasts for the onset of a nosocomial infection in elderly hospitalized patients. J. Med. Microbiol. 55, 223–228. doi: 10.1099/jmm.0.46155-46150

CrossRef Full Text | Google Scholar

Fernandes, R. A., Monteiro, D. R., Arias, L. S., Fernandes, G. L., Delbem, A. C., and Barbosa, D. B. (2016). Biofilm formation by Candida albicans and Streptococcus mutans in the presence of farnesol: a quantitative evaluation. Biofouling 32, 329–338. doi: 10.1080/08927014.2016.1144053

PubMed Abstract | CrossRef Full Text | Google Scholar

Flint, S. R., Tappuni, A., Leigh, J., Schmidt-Westhausen, A. M., and MacPhail, L. (2006). (B3) Markers of immunodeficiency and mechanisms of HAART therapy on oral lesions. Adv. Dent. Res. 19, 146–151. doi: 10.1177/154407370601900126

PubMed Abstract | CrossRef Full Text | Google Scholar

Frandsen, E. V., Pedrazzoli, V., and Kilian, M. (1991). Ecology of viridans streptococci in the oral cavity and pharynx. Oral. Microbiol. Immunol. 6, 129–133. doi: 10.1111/j.1399-302x.1991.tb00466.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Fuochi, V., Cardile, V., Petronio Petronio, G., and Furneri, P. M. (2019). Biological properties and production of bacteriocins-like-inhibitory substances by Lactobacillus sp. strains from human vagina. J. Appl. Microbiol. 126, 1541–1550. doi: 10.1111/jam.14164

PubMed Abstract | CrossRef Full Text | Google Scholar

Gagliardi, A., Totino, V., Cacciotti, F., Iebba, V., Neroni, B., Bonfiglio, G., et al. (2018). Rebuilding the gut microbiota ecosystem. Int. J. Environ. Res. Public Health 15:1679. doi: 10.3390/ijerph15081679

PubMed Abstract | CrossRef Full Text | Google Scholar

Gawaz, A., and Weisel, G. (2018). Mixed infections are a critical factor in the treatment of superficial mycoses. Mycoses 61, 731–735. doi: 10.1111/myc.12794

PubMed Abstract | CrossRef Full Text | Google Scholar

Giuliano, M., Barza, M., Jacobus, N. V., and Gorbach, S. L. (1987). Effect of broad-spectrum parenteral antibiotics on composition of intestinal microflora of humans. Antimicrob. Agents Chemother. 31, 202–206. doi: 10.1128/aac.31.2.202

PubMed Abstract | CrossRef Full Text | Google Scholar

Goldman, D. L., and Huffnagle, G. B. (2009). Potential contribution of fungal infection and colonization to the development of allergy. Med. Mycol. 47, 445–456. doi: 10.1080/13693780802641904

PubMed Abstract | CrossRef Full Text | Google Scholar

Gomaa, E. Z. (2020). Human gut microbiota/microbiome in health and diseases: a review. Antonie Van Leeuwenhoek 113, 2019–2040. doi: 10.1007/s10482-020-01474-1477

CrossRef Full Text | Google Scholar

Graf, K., Last, A., Gratz, R., Allert, S., Linde, S., Westermann, M., et al. (2019). Keeping Candida commensal: how lactobacilli antagonize pathogenicity of Candida albicans in an in vitro gut model. Dis. Model. Mech. 12:dmm039719. doi: 10.1242/dmm.039719

PubMed Abstract | CrossRef Full Text | Google Scholar

Graham, C. E., Cruz, M. R., Garsin, D. A., and Lorenz, M. C. (2017). Enterococcus faecalis bacteriocin EntV inhibits hyphal morphogenesis, biofilm formation, and virulence of Candida albicans. Proc. Natl. Acad. Sci. U S A. 114, 4507–4512. doi: 10.1073/pnas.1620432114

PubMed Abstract | CrossRef Full Text | Google Scholar

Greenbaum, S., Greenbaum, G., Moran-Gilad, J., and Weintraub, A. Y. (2019). Ecological dynamics of the vaginal microbiome in relation to health and disease. Am. J. Obstet. Gynecol. 220, 324–335. doi: 10.1016/j.ajog.2018.11.1089

PubMed Abstract | CrossRef Full Text | Google Scholar

Gregoire, S., Xiao, J., Silva, B. B., Gonzalez, I., Agidi, P. S., Klein, M. I., et al. (2011). Role of glucosyltransferase B in interactions of Candida albicans with Streptococcus mutans and with an experimental pellicle on hydroxyapatite surfaces. Appl. Environ. Microbiol. 77, 6357–6367. doi: 10.1128/aem.05203-5211

CrossRef Full Text | Google Scholar

Guggenbichler, J. P., Kofler, J., and Allerberger, F. (1985). The influence of third-generation cephalosporins on the aerobic intestinal flora. Infection 13(Suppl. 1), S137–S139. doi: 10.1007/bf01644235

PubMed Abstract | CrossRef Full Text | Google Scholar

Guinan, J., and Thangamani, S. (2018). Antibiotic-induced alterations in taurocholic acid levels promote gastrointestinal colonization of Candida albicans. FEMS Microbiol. Lett. 365:fny196. doi: 10.1093/femsle/fny196

CrossRef Full Text | Google Scholar

Guinan, J., Villa, P., and Thangamani, S. (2018). Secondary bile acids inhibit Candida albicans growth and morphogenesis. Pathog Dis. 76. doi: 10.1093/femspd/fty038

PubMed Abstract | CrossRef Full Text | Google Scholar

Gutierrez, D., Weinstock, A., Antharam, V. C., Gu, H., Jasbi, P., Shi, X., et al. (2020). Antibiotic-induced gut metabolome and microbiome alterations increase the susceptibility to Candida albicans colonization in the gastrointestinal tract. FEMS Microbiol. Ecol. 96:fiz187. doi: 10.1093/femsec/fiz187

PubMed Abstract | CrossRef Full Text | Google Scholar

Hall, R. A., and Noverr, M. C. (2017). Fungal interactions with the human host: exploring the spectrum of symbiosis. Curr. Opin. Microbiol. 40, 58–64. doi: 10.1016/j.mib.2017.10.020

PubMed Abstract | CrossRef Full Text | Google Scholar

Healy, M. G., Devine, C. M., and Murphy, R. (1996). Microbial production of biosurfactants. Resources Conserv. Recycling 18, 41–57.

Google Scholar

Hellstein, J. W., and Marek, C. L. (2019). Candidiasis: red and white manifestations in the oral cavity. Head Neck Pathol. 13, 25–32. doi: 10.1007/s12105-019-01004-1006

CrossRef Full Text | Google Scholar

Högenauer, C., Hammer, H. F., Krejs, G. J., and Reisinger, E. C. (1998). Mechanisms and management of antibiotic-associated diarrhea. Clin. Infect. Dis. 27, 702–710. doi: 10.1086/514958

PubMed Abstract | CrossRef Full Text | Google Scholar

Holmes, A. R., Gilbert, C., Wells, J. M., and Jenkinson, H. F. (1998). Binding properties of Streptococcus gordonii SspA and SspB (antigen I/II family) polypeptides expressed on the cell surface of Lactococcus lactis MG1363. Infect. Immun. 66, 4633–4639. doi: 10.1128/IAI.66.10.4633-4639.1998

PubMed Abstract | CrossRef Full Text | Google Scholar

Horn, D. L., Neofytos, D., Anaissie, E. J., Fishman, J. A., Steinbach, W. J., Olyaei, A. J., et al. (2009). Epidemiology and outcomes of candidemia in 2019 patients: data from the prospective antifungal therapy alliance registry. Clin. Infect. Dis. 48, 1695–1703. doi: 10.1086/599039

PubMed Abstract | CrossRef Full Text | Google Scholar

Hsieh, S. H., Brunke, S., and Brock, M. (2017). Encapsulation of antifungals in micelles protects Candida albicans during gall-bladder infection. Front. Microbiol. 8:117. doi: 10.3389/fmicb.2017.00117

PubMed Abstract | CrossRef Full Text | Google Scholar

Hube, B. (2004). From commensal to pathogen: stage- and tissue-specific gene expression of Candida albicans. Curr. Opin. Microbiol. 7, 336–341. doi: 10.1016/j.mib.2004.06.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Hwang, G., Liu, Y., Kim, D., Li, Y., Krysan, D. J., and Koo, H. (2017). Candida albicans mannans mediate Streptococcus mutans exoenzyme GtfB binding to modulate cross-kingdom biofilm development in vivo. PLoS Pathog 13:e1006407. doi: 10.1371/journal.ppat.1006407

PubMed Abstract | CrossRef Full Text | Google Scholar

Hwang, G., Marsh, G., Gao, L., Waugh, R., and Koo, H. (2015). Binding force dynamics of Streptococcus mutans-glucosyltransferase B to Candida albicans. J. Dent. Res. 94, 1310–1317. doi: 10.1177/0022034515592859

PubMed Abstract | CrossRef Full Text | Google Scholar

Iliev, I. D., Funari, V. A., Taylor, K. D., Nguyen, Q., Reyes, C. N., Strom, S. P., et al. (2012). Interactions between commensal fungi and the C-type lectin receptor Dectin-1 influence colitis. Science 336, 1314–1317. doi: 10.1126/science.1221789

PubMed Abstract | CrossRef Full Text | Google Scholar

Jabra-Rizk, M. A., Meiller, T. F., James, C. E., and Shirtliff, M. E. (2006). Effect of farnesol on Staphylococcus aureus biofilm formation and antimicrobial susceptibility. Antimicrob. Agents Chemother. 50, 1463–1469. doi: 10.1128/AAC.50.4.1463-1469.2006

PubMed Abstract | CrossRef Full Text | Google Scholar

Jack, A. A., Daniels, D. E., Jepson, M. A., Vickerman, M. M., Lamont, R. J., Jenkinson, H. F., et al. (2015). Streptococcus gordonii comCDE (competence) operon modulates biofilm formation with Candida albicans. Microbiology (Reading) 161, 411–421. doi: 10.1099/mic.0.000010

PubMed Abstract | CrossRef Full Text | Google Scholar

Jang, S. J., Lee, K., Kwon, B., You, H. J., and Ko, G. (2019). Vaginal lactobacilli inhibit growth and hyphae formation of Candida albicans. Sci. Rep. 9:8121. doi: 10.1038/s41598-019-44579-44574

CrossRef Full Text | Google Scholar

Jesionowski, A. M., Mansfield, J. M., Brittan, J. L., Jenkinson, H. F., and Vickerman, M. M. (2016). Transcriptome analysis of Streptococcus gordonii Challis DL1 indicates a role for the biofilm-associated fruRBA operon in response to Candida albicans. Mol. Oral Microbiol. 31, 314–328. doi: 10.1111/omi.12125

PubMed Abstract | CrossRef Full Text | Google Scholar

Kalan, L., and Grice, E. A. (2018). Fungi in the wound microbiome. Adv. Wound Care (New Rochelle) 7, 247–255. doi: 10.1089/wound.2017.0756

PubMed Abstract | CrossRef Full Text | Google Scholar

Kalia, N., Kaur, M., Sharma, S., and Singh, J. (2018). A comprehensive in silico analysis of regulatory SNPs of human CLEC7A gene and its validation as genotypic and phenotypic disease marker in recurrent vulvovaginal infections. Front. Cell Infect. Microbiol. 8:65. doi: 10.3389/fcimb.2018.00065

PubMed Abstract | CrossRef Full Text | Google Scholar

Kalia, N., Singh, J., Sharma, S., Arora, H., and Kaur, M. (2017). Genetic and phenotypic screening of mannose-binding lectin in relation to risk of recurrent vulvovaginal infections in women of north india: a prospective cohort study. Front. Microbiol. 8:75. doi: 10.3389/fmicb.2017.00075

PubMed Abstract | CrossRef Full Text | Google Scholar

Kalia, N., Singh, J., Sharma, S., and Kaur, M. (2019). SNPs in 3’-UTR region of MBL2 increases susceptibility to recurrent vulvovaginal infections by altering sMBL levels. Immunobiology 224, 42–49. doi: 10.1016/j.imbio.2018.10.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Kapitan, M., Niemiec, M. J., Steimle, A., Frick, J. S., and Jacobsen, I. D. (2019). Fungi as part of the microbiota and interactions with intestinal bacteria. Curr. Top. Microbiol. Immunol. 422, 265–301. doi: 10.1007/82_2018_117

CrossRef Full Text | Google Scholar

Khan, I., Bai, Y., Zha, L., Ullah, N., Ullah, H., Shah, S. R. H., et al. (2021). Mechanism of the gut microbiota colonization resistance and enteric pathogen infection. Front. Cell Infect. Microbiol. 11:716299. doi: 10.3389/fcimb.2021.716299

PubMed Abstract | CrossRef Full Text | Google Scholar

Khoury, Z. H., Vila, T., Puthran, T. R., Sultan, A. S., Montelongo-Jauregui, D., Melo, M. A. S., et al. (2020). The role of Candida albicans secreted polysaccharides in augmenting Streptococcus mutans adherence and mixed biofilm formation: in vitro and in vivo studies. Front. Microbiol. 11:307. doi: 10.3389/fmicb.2020.00307

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, D., Liu, Y., Benhamou, R. I., Sanchez, H., Simon-Soro, A., Li, Y., et al. (2018). Bacterial-derived exopolysaccharides enhance antifungal drug tolerance in a cross-kingdom oral biofilm. ISME J. 12, 1427–1442. doi: 10.1038/s41396-018-0113-111

CrossRef Full Text | Google Scholar

Kim, D., Sengupta, A., Niepa, T. H., Lee, B. H., Weljie, A., Freitas-Blanco, V. S., et al. (2017). Candida albicans stimulates Streptococcus mutans microcolony development via cross-kingdom biofilm-derived metabolites. Sci. Rep. 7:41332. doi: 10.1038/srep41332

PubMed Abstract | CrossRef Full Text | Google Scholar

Klotz, S. A., Chasin, B. S., Powell, B., Gaur, N. K., and Lipke, P. N. (2007). Polymicrobial bloodstream infections involving Candida species: analysis of patients and review of the literature. Diagn. Microbiol. Infect. Dis. 59, 401–406. doi: 10.1016/j.diagmicrobio.2007.07.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Kong, E. F., Tsui, C., Kucharikova, S., Andes, D., Van Dijck, P., and Jabra-Rizk, M. A. (2016). Commensal protection of Staphylococcus aureus against antimicrobials by Candida albicans biofilm matrix. mBio 7:e01365-16. doi: 10.1128/mBio.01365-1316

CrossRef Full Text | Google Scholar

Kong, E. F., Tsui, C., Kucharíková, S., Van Dijck, P., and Jabra-Rizk, M. A. (2017). Modulation of Staphylococcus aureus response to antimicrobials by the Candida albicans quorum sensing molecule farnesol. Antimicrob. Agents Chemother. 61:e01573-17. doi: 10.1128/aac.01573-1517

CrossRef Full Text | Google Scholar

Koo, H., Hayacibara, M. F., Schobel, B. D., Cury, J. A., Rosalen, P. L., Park, Y. K., et al. (2003). Inhibition of Streptococcus mutans biofilm accumulation and polysaccharide production by apigenin and tt-farnesol. J. Antimicrob Chemother 52, 782–789. doi: 10.1093/jac/dkg449

PubMed Abstract | CrossRef Full Text | Google Scholar

Kovachev, S. M., and Vatcheva-Dobrevska, R. S. (2015). Local probiotic therapy for vaginal Candida albicans infections. Probiotics Antimicrob Proteins 7, 38–44. doi: 10.1007/s12602-014-9176-9170

CrossRef Full Text | Google Scholar

Kraneveld, E. A., Buijs, M. J., Bonder, M. J., Visser, M., Keijser, B. J., Crielaard, W., et al. (2012). The relation between oral Candida load and bacterial microbiome profiles in Dutch older adults. PLoS One 7:e42770. doi: 10.1371/journal.pone.0042770

PubMed Abstract | CrossRef Full Text | Google Scholar

Krasowska, A., Murzyn, A., Dyjankiewicz, A., Łukaszewicz, M., and Dziadkowiec, D. (2009). The antagonistic effect of Saccharomyces boulardii on Candida albicans filamentation, adhesion and biofilm formation. FEMS Yeast Res. 9, 1312–1321. doi: 10.1111/j.1567-1364.2009.00559.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Krause, J., Geginat, G., and Tammer, I. (2015). Prostaglandin E2 from Candida albicans stimulates the growth of Staphylococcus aureus in mixed biofilms. PLoS One 10:e0135404. doi: 10.1371/journal.pone.0135404

PubMed Abstract | CrossRef Full Text | Google Scholar

Kullberg, B. J., and Arendrup, M. C. (2015). Invasive candidiasis. N. Engl. J. Med. 373, 1445–1456. doi: 10.1056/NEJMra1315399

PubMed Abstract | CrossRef Full Text | Google Scholar

Levison, M. E., and Pitsakis, P. G. (1987). Susceptibility to experimental Candida albicans urinary tract infection in the rat. J. Infect. Dis. 155, 841–846. doi: 10.1093/infdis/155.5.841

PubMed Abstract | CrossRef Full Text | Google Scholar

Lewis, K. (2000). Programmed death in bacteria. Microbiol. Mol. Biol. Rev. 64, 503–514. doi: 10.1128/mmbr.64.3.503-514.2000

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, J., Chen, D., Yu, B., He, J., Zheng, P., Mao, X., et al. (2018a). Fungi in gastrointestinal tracts of human and mice: from community to functions. Microb. Ecol. 75, 821–829. doi: 10.1007/s00248-017-1105-1109

CrossRef Full Text | Google Scholar

Li, X., Leonardi, I., Semon, A., Doron, I., Gao, I. H., Putzel, G. G., et al. (2018b). Response to fungal dysbiosis by gut-resident CX3CR1(+) mononuclear phagocytes aggravates allergic airway disease. Cell Host Microbe 24, 847–856.e4. doi: 10.1016/j.chom.2018.11.003.

PubMed Abstract | CrossRef Full Text | Google Scholar

Lidbeck, A., and Nord, C. E. (1993). Lactobacilli and the normal human anaerobic microflora. Clin. Infect. Dis. 16, (Suppl. 4), S181–S187.

Google Scholar

Linhares, I. M., Sisti, G., Minis, E., de Freitas, G. B., Moron, A. F., and Witkin, S. S. (2019). Contribution of epithelial cells to defense mechanisms in the human vagina. Curr. Infect. Dis. Rep. 21:30. doi: 10.1007/s11908-019-0686-685

CrossRef Full Text | Google Scholar

Lo, H. J., Köhler, J. R., DiDomenico, B., Loebenberg, D., Cacciapuoti, A., Fink, G. R., et al. (1997). albicans mutants are avirulent. Cell 90, 939–949.

Google Scholar

Loesche, W. J. (1986). Role of Streptococcus mutans in human dental decay. Microbiol. Rev. 50, 353–380.

Google Scholar

Ma, S., Zhao, Y., Xia, X., Dong, X., Ge, W., and Li, H. (2015). Effects of Streptococcus sanguinis bacteriocin on cell surface hydrophobicity, membrane permeability, and ultrastructure of Candida thallus. Biomed. Res. Int. 2015:514152. doi: 10.1155/2015/514152

PubMed Abstract | CrossRef Full Text | Google Scholar

Magill, S. S., Edwards, J. R., Bamberg, W., Beldavs, Z. G., Dumyati, G., Kainer, M. A., et al. (2014). Multistate point-prevalence survey of health care-associated infections. N. Engl. J. Med. 370, 1198–1208. doi: 10.1056/NEJMoa1306801

PubMed Abstract | CrossRef Full Text | Google Scholar

Magill, S. S., O’Leary, E., Janelle, S. J., Thompson, D. L., Dumyati, G., Nadle, J., et al. (2018). Changes in prevalence of health care-associated infections in U.S. Hospitals. N. Engl. J. Med. 379, 1732–1744. doi: 10.1056/NEJMoa1801550

PubMed Abstract | CrossRef Full Text | Google Scholar

Markey, L., Hooper, A., Melon, L. C., Baglot, S., Hill, M. N., Maguire, J., et al. (2020). Colonization with the commensal fungus Candida albicans perturbs the gut-brain axis through dysregulation of endocannabinoid signaling. Psychoneuroendocrinology 121:104808. doi: 10.1016/j.psyneuen.2020.104808

PubMed Abstract | CrossRef Full Text | Google Scholar

Marsh, P. D., and Zaura, E. (2017). Dental biofilm: ecological interactions in health and disease. J. Clin. Periodontol. 44(Suppl. 18), S12–S22. doi: 10.1111/jcpe.12679

PubMed Abstract | CrossRef Full Text | Google Scholar

Mason, K. L., Erb Downward, J. R., Mason, K. D., Falkowski, N. R., Eaton, K. A., Kao, J. Y., et al. (2012). Candida albicans and bacterial microbiota interactions in the cecum during recolonization following broad-spectrum antibiotic therapy. Infect. Immun. 80, 3371–3380. doi: 10.1128/iai.00449-412

CrossRef Full Text | Google Scholar

Matsuo, K., Haku, A., Bi, B., Takahashi, H., Kamada, N., Yaguchi, T., et al. (2019). Fecal microbiota transplantation prevents Candida albicans from colonizing the gastrointestinal tract. Microbiol. Immunol. 63, 155–163. doi: 10.1111/1348-0421.12680

PubMed Abstract | CrossRef Full Text | Google Scholar

Maudsdotter, L., Jonsson, H., Roos, S., and Jonsson, A. B. (2011). Lactobacilli reduce cell cytotoxicity caused by Streptococcus pyogenes by producing lactic acid that degrades the toxic component lipoteichoic acid. Antimicrob. Agents Chemother. 55, 1622–1628. doi: 10.1128/aac.00770-710

CrossRef Full Text | Google Scholar

McCarty, T. P., and Pappas, P. G. (2016). Invasive candidiasis. Infect. Dis. Clin. North Am. 30, 103–124. doi: 10.1016/j.idc.2015.10.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Mendling, W. (2016). Vaginal microbiota. Adv. Exp. Med. Biol. 902, 83–93. doi: 10.1007/978-3-319-31248-4_6

CrossRef Full Text | Google Scholar

Miller, E. A., Beasley, D. E., Dunn, R. R., and Archie, E. A. (2016). Lactobacilli dominance and vaginal pH: why is the human vaginal microbiome unique? Front. Microbiol. 7:1936. doi: 10.3389/fmicb.2016.01936

PubMed Abstract | CrossRef Full Text | Google Scholar

Millsop, J. W., and Fazel, N. (2016). Oral candidiasis. Clin. Dermatol. 34, 487–494. doi: 10.1016/j.clindermatol.2016.02.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Miranda, L. N., van der Heijden, I. M., Costa, S. F., Sousa, A. P., Sienra, R. A., Gobara, S., et al. (2009). Candida colonisation as a source for candidaemia. J. Hosp. Infect. 72, 9–16. doi: 10.1016/j.jhin.2009.02.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Mizutani, T., Ishizaka, A., Koga, M., Tsutsumi, T., and Yotsuyanagi, H. (2022). Role of microbiota in viral infections and pathological progression. Viruses 14:950. doi: 10.3390/v14050950

PubMed Abstract | CrossRef Full Text | Google Scholar

Montelongo-Jauregui, D., Saville, S. P., and Lopez-Ribot, J. L. (2019). Contributions of Candida albicans dimorphism, adhesive interactions, and extracellular matrix to the formation of dual-species biofilms with Streptococcus gordonii. mBio 10:e01179-19. doi: 10.1128/mBio.01179-1119

CrossRef Full Text | Google Scholar

Montelongo-Jauregui, D., Srinivasan, A., Ramasubramanian, A. K., and Lopez-Ribot, J. L. (2016). An in vitro model for oral mixed biofilms of Candida albicans and Streptococcus gordonii in synthetic saliva. Front. Microbiol. 7:686. doi: 10.3389/fmicb.2016.00686

PubMed Abstract | CrossRef Full Text | Google Scholar

Montelongo-Jauregui, D., Srinivasan, A., Ramasubramanian, A. K., and Lopez-Ribot, J. L. (2018). An in vitro model for Candida albicansStreptococcus gordonii biofilms on titanium surfaces. J. Fungi (Basel) 4:66. doi: 10.3390/jof4020066

PubMed Abstract | CrossRef Full Text | Google Scholar

Morales, D. K., and Hogan, D. A. (2010). Candida albicans interactions with bacteria in the context of human health and disease. PLoS Pathog 6:e1000886. doi: 10.1371/journal.ppat.1000886

PubMed Abstract | CrossRef Full Text | Google Scholar

Morgan, J., Meltzer, M. I., Plikaytis, B. D., Sofair, A. N., Huie-White, S., Wilcox, S., et al. (2005). Excess mortality, hospital stay, and cost due to candidemia: a case-control study using data from population-based candidemia surveillance. Infect. Control Hosp. Epidemiol. 26, 540–547. doi: 10.1086/502581

PubMed Abstract | CrossRef Full Text | Google Scholar

Morse, D. J., Wilson, M. J., Wei, X., Bradshaw, D. J., Lewis, M. A. O., and Williams, D. W. (2019). Modulation of Candida albicans virulence in in vitro biofilms by oral bacteria. Lett. Appl. Microbiol. 68, 337–343. doi: 10.1111/lam.13145

PubMed Abstract | CrossRef Full Text | Google Scholar

Nader-Macías, M. E., and Juárez Tomás, M. S. (2015). Profiles and technological requirements of urogenital probiotics. Adv. Drug Deliv. Rev. 92, 84–104. doi: 10.1016/j.addr.2015.03.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Netea, M. G., Brown, G. D., Kullberg, B. J., and Gow, N. A. (2008). An integrated model of the recognition of Candida albicans by the innate immune system. Nat. Rev. Microbiol. 6, 67–78. doi: 10.1038/nrmicro1815

PubMed Abstract | CrossRef Full Text | Google Scholar

Nobbs, A. H., Vickerman, M. M., and Jenkinson, H. F. (2010). Heterologous expression of Candida albicans cell wall-associated adhesins in Saccharomyces cerevisiae reveals differential specificities in adherence and biofilm formation and in binding oral Streptococcus gordonii. Eukaryot. Cell 9, 1622–1634. doi: 10.1128/EC.00103-110

CrossRef Full Text | Google Scholar

Nobile, C. J., and Johnson, A. D. (2015). Candida albicans biofilms and human disease. Annu. Rev. Microbiol. 69, 71–92. doi: 10.1146/annurev-micro-091014-104330

PubMed Abstract | CrossRef Full Text | Google Scholar

O’Donnell, L. E., Millhouse, E., Sherry, L., Kean, R., Malcolm, J., Nile, C. J., et al. (2015). Polymicrobial Candida biofilms: friends and foe in the oral cavity. FEMS Yeast Res. 15:fov077. doi: 10.1093/femsyr/fov077

PubMed Abstract | CrossRef Full Text | Google Scholar

Orrhage, K., and Nord, C. E. (2000). Bifidobacteria and lactobacilli in human health. Drugs Exp. Clin. Res. 26, 95–111.

Google Scholar

Pammi, M., Liang, R., Hicks, J., Mistretta, T. A., and Versalovic, J. (2013). Biofilm extracellular DNA enhances mixed species biofilms of Staphylococcus epidermidis and Candida albicans. BMC Microbiol. 13:257. doi: 10.1186/1471-2180-13-257

PubMed Abstract | CrossRef Full Text | Google Scholar

Paniágua, A. L., Correia, A. F., Pereira, L. C., de Alencar, B. M., Silva, F. B. A., Almeida, R. M., et al. (2021). Inhibitory effects of Lactobacillus casei Shirota against both Candida auris and Candida spp. isolates that cause vulvovaginal candidiasis and are resistant to antifungals. BMC Complement Med. Ther. 21:237. doi: 10.1186/s12906-021-03405-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Pappas, P. G., Lionakis, M. S., Arendrup, M. C., Ostrosky-Zeichner, L., and Kullberg, B. J. (2018). Invasive candidiasis. Nat. Rev. Dis. Primers 4:18026. doi: 10.1038/nrdp.2018.26

PubMed Abstract | CrossRef Full Text | Google Scholar

Pascual, L. M., Daniele, M. B., Giordano, W., Pájaro, M. C., and Barberis, I. L. (2008). Purification and partial characterization of novel bacteriocin L23 produced by Lactobacillus fermentum L23. Curr. Microbiol. 56, 397–402. doi: 10.1007/s00284-007-9094-9094

CrossRef Full Text | Google Scholar

Payne, S., Gibson, G., Wynne, A., Hudspith, B., Brostoff, J., and Tuohy, K. (2003). In vitro studies on colonization resistance of the human gut microbiota to Candida albicans and the effects of tetracycline and Lactobacillus plantarum LPK. Curr. Issues Intest. Microbiol. 4, 1–8.

PubMed Abstract | Google Scholar

Pellon, A., Sadeghi Nasab, S. D., and Moyes, D. L. (2020). New insights in Candida albicans innate immunity at the mucosa: toxins, epithelium, metabolism, and beyond. Front. Cell Infect. Microbiol. 10:81. doi: 10.3389/fcimb.2020.00081

PubMed Abstract | CrossRef Full Text | Google Scholar

Pericolini, E., Gabrielli, E., Ballet, N., Sabbatini, S., Roselletti, E., Cayzeele Decherf, A., et al. (2017). Therapeutic activity of a Saccharomyces cerevisiae-based probiotic and inactivated whole yeast on vaginal candidiasis. Virulence 8, 74–90. doi: 10.1080/21505594.2016.1213937

PubMed Abstract | CrossRef Full Text | Google Scholar

Peters, B. M., Jabra-Rizk, M. A., Scheper, M. A., Leid, J. G., Costerton, J. W., and Shirtliff, M. E. (2010). Microbial interactions and differential protein expression in Staphylococcus aureus -Candida albicans dual-species biofilms. FEMS Immunol. Med. Microbiol. 59, 493–503. doi: 10.1111/j.1574-695X.2010.00710.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Peters, B. M., and Noverr, M. C. (2013). Candida albicans-Staphylococcus aureus polymicrobial peritonitis modulates host innate immunity. Infect. Immun. 81, 2178–2189. doi: 10.1128/IAI.00265-213

CrossRef Full Text | Google Scholar

Peters, B. M., Ovchinnikova, E. S., Krom, B. P., Schlecht, L. M., Zhou, H., Hoyer, L. L., et al. (2012). Staphylococcus aureus adherence to Candida albicans hyphae is mediated by the hyphal adhesin Als3p. Microbiology (Reading) 158, 2975–2986. doi: 10.1099/mic.0.062109-62100

CrossRef Full Text | Google Scholar

Pfaller, M. A., and Diekema, D. J. (2007). Epidemiology of invasive candidiasis: a persistent public health problem. Clin. Microbiol. Rev. 20, 133–163. doi: 10.1128/cmr.00029-26

CrossRef Full Text | Google Scholar

Pfaller, M. A., Diekema, D. J., Turnidge, J. D., Castanheira, M., and Jones, R. N. (2019). Twenty years of the SENTRY antifungal surveillance program: results for candida species from 1997-2016. Open Forum Infect. Dis. 6, S79–S94. doi: 10.1093/ofid/ofy358

PubMed Abstract | CrossRef Full Text | Google Scholar

Pidwill, G. R., Rego, S., Jenkinson, H. F., Lamont, R. J., and Nobbs, A. H. (2018). Coassociation between Group B Streptococcus and Candida albicans promotes interactions with vaginal epithelium. Infect. Immun. 86:e00669-17. doi: 10.1128/iai.00669-617

CrossRef Full Text | Google Scholar

Qin, Y., Zhang, L., Xu, Z., Zhang, J., Jiang, Y. Y., Cao, Y., et al. (2016). Innate immune cell response upon Candida albicans infection. Virulence 7, 512–526. doi: 10.1080/21505594.2016.1138201

PubMed Abstract | CrossRef Full Text | Google Scholar

Qiu, R., Li, W., Lin, Y., Yu, D., and Zhao, W. (2015a). Genotypic diversity and cariogenicity of Candida albicans from children with early childhood caries and caries-free children. BMC Oral Health 15:144. doi: 10.1186/s12903-015-0134-133

CrossRef Full Text | Google Scholar

Qiu, X., Zhang, F., Yang, X., Wu, N., Jiang, W., et al. (2015b). Changes in the composition of intestinal fungi and their role in mice with dextran sulfate sodium-induced colitis. Sci. Rep. 5:10416. doi: 10.1038/srep10416

PubMed Abstract | CrossRef Full Text | Google Scholar

Raja, M., Hannan, A., and Ali, K. (2010). Association of oral candidal carriage with dental caries in children. Caries Res. 44, 272–276. doi: 10.1159/000314675

PubMed Abstract | CrossRef Full Text | Google Scholar

Ramage, G., Saville, S. P., Wickes, B. L., and López-Ribot, J. L. (2002). Inhibition of Candida albicans biofilm formation by farnesol, a quorum-sensing molecule. Appl. Environ. Microbiol. 68, 5459–5463. doi: 10.1128/aem.68.11.5459-5463.2002

PubMed Abstract | CrossRef Full Text | Google Scholar

Rego, S., Heal, T. J., Pidwill, G. R., Till, M., Robson, A., Lamont, R. J., et al. (2016). Structural and functional analysis of cell wall-anchored polypeptide adhesin BspA in Streptococcus agalactiae. J. Biol. Chem. 291, 15985–16000. doi: 10.1074/jbc.M116.726562

PubMed Abstract | CrossRef Full Text | Google Scholar

Reid, G., Beuerman, D., Heinemann, C., and Bruce, A. W. (2001). Probiotic Lactobacillus dose required to restore and maintain a normal vaginal flora. FEMS Immunol. Med. Microbiol. 32, 37–41. doi: 10.1111/j.1574-695X.2001.tb00531.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Reid, G., Charbonneau, D., Erb, J., Kochanowski, B., Beuerman, D., Poehner, R., et al. (2003). Oral use of Lactobacillus rhamnosus GR-1 and L. fermentum RC-14 significantly alters vaginal flora: randomized, placebo-controlled trial in 64 healthy women. FEMS Immunol. Med. Microbiol. 35, 131–134. doi: 10.1016/s0928-8244(02)00465-460

CrossRef Full Text | Google Scholar

Reno, J., Doshi, S., Tunali, A. K., Stein, B., Farley, M. M., Ray, S. M., et al. (2015). Epidemiology of methicillin-resistant Staphylococcus aureus bloodstream coinfection among adults with candidemia in Atlanta, GA, 2008-2012. Infect. Control Hosp. Epidemiol. 36, 1298–1304. doi: 10.1017/ice.2015.185

PubMed Abstract | CrossRef Full Text | Google Scholar

Richardson, J. P., and Moyes, D. L. (2015). Adaptive immune responses to Candida albicans infection. Virulence 6, 327–337. doi: 10.1080/21505594.2015.1004977

PubMed Abstract | CrossRef Full Text | Google Scholar

Ricker, A., Vickerman, M., and Dongari-Bagtzoglou, A. (2014). Streptococcus gordonii glucosyltransferase promotes biofilm interactions with Candida albicans. J. Oral Microbiol. 6. doi: 10.3402/jom.v6.23419

PubMed Abstract | CrossRef Full Text | Google Scholar

Ricotta, E. E., Lai, Y. L., Babiker, A., Strich, J. R., Kadri, S. S., Lionakis, M. S., et al. (2020). Invasive candidiasis species distribution and trends, United States, 2009-2017. J. Infect Dis. 223, 1295–1302. doi: 10.1093/infdis/jiaa502

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodríguez-Cerdeira, C., Gregorio, M. C., Molares-Vila, A., López-Barcenas, A., Fabbrocini, G., Bardhi, B., et al. (2019). Biofilms and vulvovaginal candidiasis. Colloids Surf. B Biointerfaces 174, 110–125. doi: 10.1016/j.colsurfb.2018.11.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Romo, J. A., and Kumamoto, C. A. (2020). On commensalism of Candida. J. Fungi (Basel) 6:16. doi: 10.3390/jof6010016

PubMed Abstract | CrossRef Full Text | Google Scholar

Rossen, N. G., Fuentes, S., van der Spek, M. J., Tijssen, J. G., Hartman, J. H., Duflou, A., et al. (2015). Findings from a randomized controlled trial of fecal transplantation for patients with ulcerative colitis. Gastroenterology 149, 110–118.e4. doi: 10.1053/j.gastro.2015.03.045.

PubMed Abstract | CrossRef Full Text | Google Scholar

Sabbatini, S., Visconti, S., Gentili, M., Lusenti, E., Nunzi, E., Ronchetti, S., et al. (2021). Lactobacillus iners cell-free supernatant enhances biofilm formation and hyphal/pseudohyphal growth by Candida albicans vaginal isolates. Microorganisms 9:2577. doi: 10.3390/microorganisms9122577

PubMed Abstract | CrossRef Full Text | Google Scholar

Salvatori, O., Kumar, R., Metcalfe, S., Vickerman, M., Kay, J. G., and Edgerton, M. (2020). Bacteria modify Candida albicans hypha formation, microcolony properties, and survival within macrophages. mSphere 5:e00689-20. doi: 10.1128/mSphere.00689-620

CrossRef Full Text | Google Scholar

Samonis, G., Gikas, A., Anaissie, E. J., Vrenzos, G., Maraki, S., Tselentis, Y., et al. (1993). Prospective evaluation of effects of broad-spectrum antibiotics on gastrointestinal yeast colonization of humans. Antimicrob. Agents Chemother. 37, 51–53. doi: 10.1128/aac.37.1.51

PubMed Abstract | CrossRef Full Text | Google Scholar

Scarpellini, E., Ianiro, G., Attili, F., Bassanelli, C., De Santis, A., and Gasbarrini, A. (2015). The human gut microbiota and virome: potential therapeutic implications. Dig. Liver Dis. 47, 1007–1012. doi: 10.1016/j.dld.2015.07.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Schlecht, L. M., Peters, B. M., Krom, B. P., Freiberg, J. A., Hänsch, G. M., Filler, S. G., et al. (2015). Systemic Staphylococcus aureus infection mediated by Candida albicans hyphal invasion of mucosal tissue. Microbiology (Reading) 161, 168–181. doi: 10.1099/mic.0.083485-83480

CrossRef Full Text | Google Scholar

Scully, C., el-Kabir, M., and Samaranayake, L. P. (1994). Candida and oral candidosis: a review. Crit. Rev. Oral Biol. Med. 5, 125–157. doi: 10.1177/10454411940050020101

PubMed Abstract | CrossRef Full Text | Google Scholar

Seelig, M. S. (1966a). Mechanisms by which antibiotics increase the incidence and severity of candidiasis and alter the immunological defenses. Bacteriol. Rev. 30, 442–459. doi: 10.1128/br.30.2.442-459.1966

PubMed Abstract | CrossRef Full Text | Google Scholar

Seelig, M. S. (1966b). The role of antibiotics in the pathogenesis of Candidainfections. Am. J. Med. 40, 887–917. doi: 10.1016/0002-9343(66)90204-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Sestito, S., D’Auria, E., Baldassarre, M. E., Salvatore, S., Tallarico, V., Stefanelli, E., et al. (2020). The role of prebiotics and probiotics in prevention of allergic diseases in infants. Front. Pediatr. 8:583946. doi: 10.3389/fped.2020.583946

PubMed Abstract | CrossRef Full Text | Google Scholar

Severance, E. G., Gressitt, K. L., Stallings, C. R., Katsafanas, E., Schweinfurth, L. A., Savage, C. L., et al. (2016). Candida albicans exposures, sex specificity and cognitive deficits in schizophrenia and bipolar disorder. NPJ Schizophr. 2:16018. doi: 10.1038/npjschz.2016.18

PubMed Abstract | CrossRef Full Text | Google Scholar

Sheehan, G., Tully, L., and Kavanagh, K. A. (2020). Candida albicans increases the pathogenicity of Staphylococcus aureus during polymicrobial infection of Galleria mellonella larvae. Microbiology (Reading) 166, 375–385. doi: 10.1099/mic.0.000892

PubMed Abstract | CrossRef Full Text | Google Scholar

Shirtliff, M. E., Peters, B. M., and Jabra-Rizk, M. A. (2009). Cross-kingdom interactions: Candida albicans and bacteria. FEMS Microbiol. Lett. 299, 1–8. doi: 10.1111/j.1574-6968.2009.01668.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Silverman, R. J., Nobbs, A. H., Vickerman, M. M., Barbour, M. E., and Jenkinson, H. F. (2010). Interaction of Candida albicans cell wall Als3 protein with Streptococcus gordonii SspB adhesin promotes development of mixed-species communities. Infect. Immun. 78, 4644–4652. doi: 10.1128/iai.00685-610

CrossRef Full Text | Google Scholar

Sjövall, J., Huitfeldt, B., Magni, L., and Nord, C. E. (1986). Effect of beta-lactam prodrugs on human intestinal microflora. Scand. J. Infect. Dis. Suppl. 49, 73–84.

Google Scholar

Sobel, J. D. (2007). Vulvovaginal candidosis. Lancet 369, 1961–1971.

Google Scholar

Sobel, J. D. (2016). Recurrent vulvovaginal candidiasis. Am. J. Obstet. Gynecol. 214, 15–21. doi: 10.1016/j.ajog.2015.06.067

PubMed Abstract | CrossRef Full Text | Google Scholar

Souza, J. G. S., Bertolini, M., Thompson, A., Mansfield, J. M., Grassmann, A. A., Maas, K., et al. (2020). Role of glucosyltransferase R in biofilm interactions between Streptococcus oralis and Candida albicans. ISME J. 14, 1207–1222. doi: 10.1038/s41396-020-0608-604

CrossRef Full Text | Google Scholar

Suez, J., Zmora, N., Zilberman-Schapira, G., Mor, U., Dori-Bachash, M., Bashiardes, S., et al. (2018). Post-Antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell 174, 1406–1423.e16. doi: 10.1016/j.cell.2018.08.047.

PubMed Abstract | CrossRef Full Text | Google Scholar

Sullivan, A., Edlund, C., and Nord, C. E. (2001). Effect of antimicrobial agents on the ecological balance of human microflora. Lancet Infect. Dis. 1, 101–114.

Google Scholar

Tan, C. T., Xu, X., Qiao, Y., and Wang, Y. (2021). A peptidoglycan storm caused by β-lactam antibiotic’s action on host microbiota drives Candida albicans infection. Nat. Commun. 12:2560. doi: 10.1038/s41467-021-22845-22842

CrossRef Full Text | Google Scholar

Tan, P., Li, X., Shen, J., and Feng, Q. (2020). Fecal microbiota transplantation for the treatment of inflammatory bowel disease: an update. Front. Pharmacol. 11:574533. doi: 10.3389/fphar.2020.574533

PubMed Abstract | CrossRef Full Text | Google Scholar

Taur, Y., Coyte, K., Schluter, J., Robilotti, E., Figueroa, C., Gjonbalaj, M., et al. (2018). Reconstitution of the gut microbiota of antibiotic-treated patients by autologous fecal microbiota transplant. Sci. Transl. Med. 10:eaa9489. doi: 10.1126/scitranslmed.aap9489

PubMed Abstract | CrossRef Full Text | Google Scholar

Todd, O. A., Fidel, P. L. Jr., Harro, J. M., Hilliard, J. J., Tkaczyk, C., et al. (2019a). Candida albicans augments Staphylococcus aureus virulence by engaging the Staphylococcal agr quorum sensing system. mBio 10:e00910-19. doi: 10.1128/mBio.00910-919

CrossRef Full Text | Google Scholar

Todd, O. A., Noverr, M. C., and Peters, B. M. (2019b). Candida albicans impacts Staphylococcus aureus alpha-toxin production via extracellular alkalinization. mSphere 4:e00780-19. doi: 10.1128/mSphere.00780-719

CrossRef Full Text | Google Scholar

Tso, G. H. W., Reales-Calderon, J. A., Tan, A. S. M., Sem, X., Le, G. T. T., Tan, T. G., et al. (2018). Experimental evolution of a fungal pathogen into a gut symbiont. Science 362, 589–595. doi: 10.1126/science.aat0537

PubMed Abstract | CrossRef Full Text | Google Scholar

van der Waaij, D. (1989). The ecology of the human intestine and its consequences for overgrowth by pathogens such as Clostridium difficile. Annu. Rev. Microbiol. 43, 69–87. doi: 10.1146/annurev.mi.43.100189.000441

PubMed Abstract | CrossRef Full Text | Google Scholar

van Ogtrop, M. L., Guiot, H. F., Mattie, H., and van Furth, R. (1991). Modulation of the intestinal flora of mice by parenteral treatment with broad-spectrum cephalosporins. Antimicrob. Agents Chemother. 35, 976–982. doi: 10.1128/aac.35.5.976

PubMed Abstract | CrossRef Full Text | Google Scholar

Vijay, A., and Valdes, A. M. (2022). Role of the gut microbiome in chronic diseases: a narrative review. Eur. J. Clin. Nutr. 76, 489–501. doi: 10.1038/s41430-021-00991-996

CrossRef Full Text | Google Scholar

Vila, T., Kong, E. F., Ibrahim, A., Piepenbrink, K., Shetty, A. C., McCracken, C., et al. (2019). Candida albicans quorum-sensing molecule farnesol modulates staphyloxanthin production and activates the thiol-based oxidative-stress response in Staphylococcus aureus. Virulence 10, 625–642. doi: 10.1080/21505594.2019.1635418

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, S., Wang, Q., Yang, E., Yan, L., Li, T., and Zhuang, H. (2017). 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. 8:564. doi: 10.3389/fmicb.2017.00564

PubMed Abstract | CrossRef Full Text | Google Scholar

Wheeler, M. L., Limon, J. J., Bar, A. S., Leal, C. A., Gargus, M., Tang, J., et al. (2016). Immunological consequences of intestinal fungal dysbiosis. Cell Host Microbe 19, 865–873. doi: 10.1016/j.chom.2016.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Wisplinghoff, H., Bischoff, T., Tallent, S. M., Seifert, H., Wenzel, R. P., and Edmond, M. B. (2004). Nosocomial bloodstream infections in US hospitals: analysis of 24,179 cases from a prospective nationwide surveillance study. Clin. Infect. Dis. 39, 309–317. doi: 10.1086/421946

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, P. F., Liu, W. L., Hsieh, M. H., Hii, I. M., Lee, Y. L., Lin, Y. T., et al. (2017). Epidemiology and antifungal susceptibility of candidemia isolates of non-albicans Candida species from cancer patients. Emerg. Microbes Infect. 6:e87. doi: 10.1038/emi.2017.74

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, H., Liu, M., Cao, J., Li, X., Fan, D., Xia, Y., et al. (2019). The dynamic interplay between the gut microbiota and autoimmune diseases. J. Immunol. Res. 2019:7546047. doi: 10.1155/2019/7546047

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, X. L., Lee, R. T., Fang, H. M., Wang, Y. M., Li, R., Zou, H., et al. (2008). Bacterial peptidoglycan triggers Candida albicans hyphal growth by directly activating the adenylyl cyclase Cyr1p. Cell Host Microbe 4, 28–39. doi: 10.1016/j.chom.2008.05.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Zeise, K. D., Woods, R. J., and Huffnagle, G. B. (2021). Interplay between Candida albicans and lactic acid bacteria in the gastrointestinal tract: impact on colonization resistance, microbial carriage, opportunistic infection, and host immunity. Clin. Microbiol. Rev. 34:e0032320. doi: 10.1128/CMR.00323-320

CrossRef Full Text | Google Scholar

Zuo, T., Wong, S. H., Cheung, C. P., Lam, K., Lui, R., Cheung, K., et al. (2018). Gut fungal dysbiosis correlates with reduced efficacy of fecal microbiota transplantation in Clostridium difficile infection. Nat. Commun. 9:3663. doi: 10.1038/s41467-018-06103-6106

CrossRef Full Text | Google Scholar

Keywords: Candida albicans, microbiota, biofilm, candidiasis, polymicrobial disease

Citation: Li H, Miao M-x, Jia C-l, Cao Y-b, Yan T-h, Jiang Y-y and Yang F (2022) Interactions between Candida albicans and the resident microbiota. Front. Microbiol. 13:930495. doi: 10.3389/fmicb.2022.930495

Received: 28 April 2022; Accepted: 31 August 2022;
Published: 20 September 2022.

Edited by:

Ilse Denise Jacobsen, Leibniz Institute for Natural Product Research and Infection Biology, Germany

Reviewed by:

Maria Joanna Niemiec, Leibniz Institute for Natural Product Research and Infection Biology, Germany
Bastiaan P. Krom, VU Amsterdam, Netherlands

Copyright © 2022 Li, Miao, Jia, Cao, Yan, Jiang and Yang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Tian-hua Yan, 1020050806@cpu.edu.cn; Yuan-ying Jiang, jiangyy@tongji.edu.cn; Feng Yang, feng.yang0405@gmail.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.