Next Article in Journal
COVID-19-Associated Pulmonary Aspergillosis (CAPA) in Northern Greece during 2020–2022: A Comparative Study According to the Main Consensus Criteria and Definitions
Previous Article in Journal
Combined Application of Tacrolimus with Cyproconazole, Hymexazol and Novel {2-(3-R-1H-1,2,4-triazol-5-yl)phenyl}amines as Antifungals: In Vitro Growth Inhibition and In Silico Molecular Docking Analysis to Fungal Chitin Deacetylase
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Candida parapsilosis Virulence and Antifungal Resistance Mechanisms: A Comprehensive Review of Key Determinants

by
Joana Branco
1,2,*,
Isabel M. Miranda
3,† and
Acácio G. Rodrigues
1,2,†
1
Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
2
Center for Health Technology and Services Research—CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
3
Cardiovascular Research & Development Centre—UnIC@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Fungi 2023, 9(1), 80; https://doi.org/10.3390/jof9010080
Submission received: 30 November 2022 / Revised: 29 December 2022 / Accepted: 3 January 2023 / Published: 5 January 2023

Abstract

:
Candida parapsilosis is the second most common Candida species isolated in Asia, Southern Europe, and Latin America and is often involved in invasive infections that seriously impact human health. This pathogen is part of the psilosis complex, which also includes Candida orthopsilosis and Candida metapsilosis. C. parapsilosis infections are particularly prevalent among neonates with low birth weights, individuals who are immunocompromised, and patients who require prolonged use of a central venous catheter or other indwelling devices, whose surfaces C. parapsilosis exhibits an enhanced capacity to adhere to and form biofilms. Despite this well-acknowledged prevalence, the biology of C. parapsilosis has not been as extensively explored as that of Candida albicans. In this paper, we describe the molecular mechanistic pathways of virulence in C. parapsilosis and show how they differ from those of C. albicans. We also describe the mode of action of antifungal drugs used for the treatment of Candida infections, namely, polyenes, echinocandins, and azoles, as well as the resistance mechanisms developed by C. parapsilosis to overcome them. Finally, we stress the importance of the ongoing search for species-specific features that may aid the development of effective control strategies and thus reduce the burden on patients and healthcare costs.

1. Candida and Human Disease

Fungi can cause a diversity of health disorders in humans, ranging from allergic syndromes and mucocutaneous infections to invasive diseases that seriously threaten life. It is estimated that fungal diseases annually affect over a billion people and cause 1.5 million deaths worldwide [1]. Invasive fungal infections caused by Candida species are widely associated with high rates of severe illness and may be responsible for as many as 30% of all deaths from fungal disease. In the United States, the health cost attributable to prolonged hospitalizations resulting from candidaemia is estimated at USD 46,684 per patient [2].
Candidosis is a broad term that refers to cutaneous, mucosal, and deep-seated organ infections caused by opportunistic pathogens of the Candida genus [3]. Candida spp. are commensal yeasts commonly found in the human gastrointestinal tract, mucous membranes, and skin. Disruption of the gastrointestinal and cutaneous barriers following shock, localized infections, or the replacement of an intravascular catheter can all promote invasive candidosis, which is widely recognized as a major cause of morbidity and mortality. The patient populations most at risk are the elderly, premature newborns, and those with compromised immune systems due to HIV, chemotherapy, or transplant-necessitated immunosuppression therapy [4]. Invasive candidosis is a disorder that can potentially affect any organ. Each distinct Candida species exhibits its own unique characteristics in terms of its invasive potential, virulence, and antifungal susceptibility pattern [3].
The distribution of Candida species varies geographically, with notable differences between hospital centers. The underlying condition of the patient and whether they have experienced previous antifungal therapy both have an effect on the distribution and frequency of Candida spp. [5]. While C. albicans is the most common pathogen associated with nosocomial invasive candidosis worldwide, an increasing number of infections by non-albicans Candida species (NACs) have also been reported in recent years, including Candida glabrata, Candida parapsilosis, Candida tropicalis, Candida krusei, and Candida auris, among others [6]. Of these, C. glabrata predominates in Northern European countries and in the United States, but C. parapsilosis and/or C. tropicalis are more prevalent in India, Pakistan, Latin America, and Mediterranean countries [3].

2. Candida parapsilosis

Since its discovery in 1928, C. parapsilosis has undergone several changes in phylogenetic classification. Initially isolated from the stool of a patient with diarrhea in Puerto Rico, the species was first classified as Monilia parapsilosis (i.e., a species of the Monilia genus, incapable of fermenting maltose) to distinguish it from Monilia psilosis, which is today known as C. albicans [7]. In 1932, it was renamed Candida parapsilosis. In 2005, Tavanti et al. [8] confirmed, through multilocus sequence typing, the existence of a C. parapsilosis complex comprising three distinct species: Candida parapsilosis sensu stricto, Candida orthopsilosis, and Candida metapsilosis. In this paper, we focus on Candida parapsilosis.
C. parapsilosis is widely distributed in nature and is often isolated from a variety of non-human sources, such as domestic animals, insects, soil, and marine environments [9]. This yeast successfully colonizes the human skin and mucosal membranes as a commensal microorganism, wherein the hands of healthcare professionals are recognized as a major vector for C. parapsilosis nosocomial acquisition [10,11,12]. In addition, the selective ability of C. parapsilosis to grow in hyperalimentation solutions promotes the infection risk by this pathogen [13]. C. parapsilosis represents a high risk for immunocompromised individuals, such as HIV sufferers and surgical patients, particularly those subjected to gastrointestinal track surgery. Additionally, patients requiring prolonged use of a central venous catheter or other indwelling devices are at high risk, due to the innate ability of C. parapsilosis to adhere to prosthetic surfaces and implanted medical devices. In such cases, biofilm formation typically begins soon after attachment. When the structure is mature, it greatly decreases the ability of antifungals to reach cells, with potentially life-threatening consequences in the host [14,15,16]. Because C. parapsilosis is responsible for one-third of neonatal Candida infections, with a mortality rate of approximately 10%, low-birth-weight neonates are at especially high risk [17].
The distribution of C. parapsilosis recovered from patients with bloodstream infections in various studies conducted in different geographical areas shows that its relative dominance differs according to region [5]. It is the second most common Candida isolate in Latin America countries, such as Argentina, Peru and Brazil. In Venezuela and Colombia, C. parapsilosis even outranks C. albicans infections [5,18,19]. The incidence of C. parapsilosis infections in Europe is region-dependent; in Southern European hospitals (Portugal, Spain, Italy, and Greece) it is the second most isolated species [20,21,22,23], and in central and northern countries of Europe the incidence of C. parapsilosis ranks third, after that of C. albicans and C. glabrata [24,25,26]. A different prevalence was also reported in North American countries, Canada and USA, where C. parapsilosis ranks second and third, respectively [27,28,29,30]. According to studies of bloodstream fungal infections in Asia (China and Japan), C. parapsilosis is commonly found after C. albicans [31,32], while in India it ranks third [33]. A similar incidence of infection was observed in Australia [34].
The two cryptic psilosis species, Candida orthopsilosis and Candida metapsilosis, are also opportunistic pathogens, associated with local and systemic diseases. As with C. parapsilosis, their frequency and distribution reportedly differ in distinct geographical areas [35,36].
C. parapsilosis is a diploid pathogen, with eight chromosome pairs and an estimated genome size of 13.1 Mb. From the 5837 ORFs identified in this species, only 107 (1.83%) have actually been characterized [37]. Its genome is highly conserved; compared to other Candida spp., it exhibits a remarkably low level of heterozygosity with just one single nucleotide polymorphism (SNP) per 15,553 bases, more than 70 times less than the corresponding number in the closely related Lodderomyces elongisporus [38].
The yeast cells of C. parapsilosis display an oval, round, or cylindrical shape, and their colony phenotypes have been identified as crepe, concentric, smooth, or crater [13,39]. Unlike C. albicans, C. parapsilosis does not form true hyphae; it only exists as yeast or in pseudohyphal forms. Form and colony phenotypes are intimately linked; cells exhibiting crepe and concentric phenotypes are almost entirely pseudohyphal, whereas those with smooth and crater phenotypes are mostly yeast-like [39].

3. Virulence Attributes

Similarly to other microorganisms, Candida species have developed several specific and effective strategies to enhance their pathogenicity. The virulence of C. parapsilosis is mainly attributed to its intrinsic ability to adhere to the abiotic surfaces of medical devices and prosthetic materials, and to the host’s mucosal epithelium. This ability is crucial for biofilm formation and consequently damage to the host [15,40].
Researchers have found that the ability to colonize upon mucosal surfaces or inert materials varies among Candida species [41]. An unusually high intraspecies variation in terms of adhesion ability has also been identified among clinical isolates of C. parapsilosis, compared with other Candida species. A correlation between the site of isolation and the rate of adhesion has also been reported, as C. parapsilosis mucocutaneous isolates demonstrate higher adhesiveness [41].

3.1. Cell Adhesion

Adhesion is an important, multifactorial process that is mediated by the characteristics of fungal and host (biotic or abiotic) cells, including cell surface hydrophobicity, cell wall composition, and growth conditions [42]. Initially, the adhesion of the yeast cells is highly dependent upon hydrophobic interactions between the microorganism and host surfaces. Cell surface hydrophobicity is strongly correlated with adhesion to both polystyrene/polyetherurethane surfaces and to epithelial cells. Candida species generally exhibit a high degree of cell surface hydrophobicity [43].
In adhesion, the key trigger interaction is promoted by specific cell wall proteins, namely adhesins. This process promotes the attachment of the fungal cells to other microorganisms, the host’s epithelium, and abiotic surfaces [40]. Among Candida spp., several adhesin families are involved in adherence. Important adhesin families include: (i) the hyphal wall protein (Hwp) family, which includes five proteins, namely, Hwp1, Hwp2, Rbt1, Eap1, and Ywp1, that play a role in C. albicans biofilm formation [42,44]; (ii) the adhesins of the EPA (epithelial adhesion) family in C. glabrata, comprising 23 genes, of which EPA1, EPA6, and EPA7 are described as the most important for the adhesion process in this species [42,44,45]; and (iii) the Als-like (agglutinin-like sequence) family encoding large-cell-surface glycoproteins involved in Candida adhesion, including C. albicans, C. parapsilosis, C. tropicalis, C. dubliniensis, C. lusitaniae, and C. guilliermondii [42,44]. Among the eight Als members described in C. albicans, Als3 has the most profound impact on biofilm formation; its deletion causes a severe biofilm formation defect [46]. In C. parapsilosis, five Als proteins are present on the surface of the pseudohyphae, and the ortholog CaAls7 has been described as a determinant for adhesion to host epithelial cells [47,48]. Other adhesion proteins and non-protein factors with similar properties, such as Eap1, Iff4, Mp65, Ecm33, Utr2, Int1, and Mnt1, have also been identified in Candida species; however, these have not been widely studied to date [49].

3.2. Secretion of Hydrolytic Enzymes

Candida species can produce and secrete several hydrolytic enzymes, including secreted aspartyl proteases (SAPs), lipases (LIPs), and phospholipases. The activity of these enzymes is closely linked with Candida’s pathogenicity, such adhesion, cell damage, and the invasion of host tissues [40].
The production of SAPs by Candida cells aims to degrade structural and immunological defense proteins in the host, facilitating the invasion and colonization of the host tissue. Compared to C. albicans, C. parapsilosis expresses less SAP activity [50]. To date, three aspartyl protease-encoding genes (SAPP1 to SAPP3) have been identified in C. parapsilosis, with a wide variability in expression among different isolates [51]. Isolates from body surfaces, such as skin or vaginal mucosa, are more invasive than those recovered from systemic infections or from environmental surfaces, due to the production of such enzymes [52].
In addition to SAPs, enzymes categorized as lipases catalyze both the hydrolysis and synthesis of triacylglycerols. Of the four secreted-lipase-encoding genes identified in the C. parapsilosis genome, only two (LIP1 and LIP2) have been confirmed as able to encode functionally active proteins. Although the production of LIPs varies greatly among C. parapsilosis isolates, ranging from 36% to 80%, their role in enhanced pathogenicity has been confirmed [53]. The putative roles played by LIPs in a successful host invasion include the digestion of lipids for nutrient acquisition, the enhancement of adhesion and biofilm formation, and the suppression of immune response, among others [54,55].
Other hydrolytic enzymes have also been described, including secreted phospholipases, which hydrolyze phospholipids and fatty acids, thereby exposing host receptors and facilitating adhesion; however, these are still poorly understood in C. parapsilosis [56].

3.3. Biofilm Formation

Biofilms have been described as an organized community, comprising a dense network of microbial cells embedded in an extracellular matrix (ECM) of polymers [13]. Biofilm formation is a potent virulence attribute of several Candida species. Biofilm formation during infection has been linked to higher mortality rates in cases involving such species when compared with isolates incapable of forming biofilm [57]. Biofilm development is a well-regulated process comprising three sequential stages (Figure 1): an early phase, involving the entire adhesion process of the cells, as described above; an intermediated phase, and, finally, a maturation/dispersion phase [40]. In the intermediate phase, following initial fungal adhesion, yeast cells undergo a morphology transition from yeast to filamentous or pseudohyphal forms, forming a mixed population with a multilayer formation (Figure 1). Afterwards, biofilm maturation begins through the production and secretion of a polysaccharide-rich extracellular matrix, formed by polysaccharides, proteins, lipids, and nucleic acids, which provides structural and functional stability to the biofilm [40,58].
The biofilm’s architecture, morphology, and thickness also vary widely among Candida species and between strains [58]. These features are influenced by several host and Candida-derived variables, including: (i) physiological conditions, such as pH and oxygen concentration; (ii) fluid flow at the infection site, which influences nutrient exchange and impacts the biofilm’s structural integrity; (iii) available nutrients in the growth media, including sugars, lipids, and serum; and (iv) the material on which the biofilm grows (those typically used in medical devices include silicone, latex, and polyurethane, among others); and (v) community microbial interactions, either fungal–fungal or fungal–bacterial, which modulate the ability of Candida to form biofilm and also represent a promising topic for future research [58,59,60].
C. parapsilosis biofilm growth is especially common in patients fitted with a central venous catheter who receive total parenteral nutrition [61,62]. The biofilm structure of C. parapsilosis exhibits high variability among clinical isolates. Because C. parapsilosis does not form true hyphae, its biofilm is composed of aggregated blastoconidia and pseudohyphae that occupy a volume lower than that of other Candida species [63,64]. In addition, the extracellular matrix of C. parapsilosis biofilm is mainly composed of carbohydrates and low levels of protein [63].
The ability to form biofilms is closely related to its virulence potential, because only limited penetration of substances is possible through the biofilm matrix, resulting in a greatly decreased susceptibility to antimicrobial agents [65,66]. The development of the biofilm also serves to counter the host immune response by inhibiting macrophage phagocytosis and antibody activity [65].
The process of biofilm development involves a massive cell detachment during the final maturation phase, with consequent dispersion that promotes the colonization of new locations and surfaces [40]. However, Uppuluri et al. [67] found that dispersion was not confined to the maturation phase and occurs continuously during the biofilm development process. A more robust biofilm is produced by dispersed cells compared with the biofilm formed by initial planktonic mother cells such that the virulence potential increases over generations. All of these findings represent matters of serious clinical concern, not only for the treatment of patient infections but also in terms of public health [66].
The complexity of all stages of biofilm formation, involving such phenomena as the control of adhesion, morphology changes, and ECM production, among others, requires an extensive and complex regulatory network [68]. The biofilm formation regulatory process has been extensively studied in C. albicans; however, as with other characteristics, such knowledge cannot be simply transposed to other Candida species. For example, the four transcription factors BRG1, TEC1, ROB1, and FLO8 are all involved in the biofilm regulatory network of C. albicans but play no role in the biofilm regulation of C. parapsilosis [68,69]. Conversely, CZF1, UME6, GZF3, and CPH2 have been highlighted as key contributors to biofilm formation in C. parapsilosis, but these genes play a negligible role in this process in C. albicans. One recent report identified the direct role of Ndt80 as a repressor of C. parapsilosis virulence attributes, thereby diverging functionally from its homolog in the closely related fungal pathogen C. albicans [70]. However, other genes required for biofilm development, such as ACE2, BCR1, and EFG1, have been found to perform a similar function in both species [68,71].

4. Antifungals and Resistance Mechanisms

Despite ongoing research efforts concerning new therapeutic compounds and treatment strategies, only a limited number of options of antifungal drugs are available for the treatment of candidosis [72]. Currently, the arsenal of systemic antifungals available for clinical use consists of only three major drug classes: polyenes, echinocandins, and azoles [73].

4.1. Polyenes

Amphotericin B (AmB) is the most used member of the class of polyenes, being clinically used for more than 55 years [73]. Its potent fungicidal activity is derived from its interaction with the ergosterol of fungal cells by binding to the lipid bilayer, forming pores in the cell membrane and facilitating the leakage of intracellular components, such as potassium ions (K+), into the extracellular medium (Figure 2A) [74]. Consequently, this interaction results in a drastic change in cell permeability, ultimately leading to cell lysis. This antifungal has low solubility and is highly toxic to the host cell due to the close structural relationship between ergosterol and cholesterol, the mammalian membrane sterol. This limits its use in long-term antifungal therapy [75]. However, less toxic, lipid-based polyene formulations have now been developed, including liposomal amphotericin B (LAmB), which has become the first-line treatment for various types of invasive fungal infections [76].
The development of fungal resistance to polyenes is rare. Most Candida spp., including C. albicans, C. glabrata, and C. parapsilosis, are generally considered to be susceptible to AmB, with surveillance studies reporting an AmB susceptibility rate close to 100% [77]. Recently, a global pooled prevalence meta-analysis estimated C. parapsilosis AmB-resistance at 1.3% [78]. Emerging AmB resistance has been reported in species, such as C. auris [79]. The resistance mechanisms of this class are less well understood than those of echinocandins and azoles; nevertheless, several hypotheses have been forwarded to explain resistance, as illustrated in Figure 2A. These include: (i) sterol composition modulation through the depletion or replacement of ergosterol triggered by mutations in genes involved in the ergosterol biosynthesis pathway, specifically in ERG1 to ERG4, ERG6, and ERG11 [80,81,82]; (ii) enhanced defense against oxidative damage to break down the reactive oxygen species (ROS) that are produced under AmB exposure, either by means of catalase activity and/or by the molecular chaperones of the heat shock protein (HSP) family, namely, Hsp90 and Hsp70 [83,84,85].

4.2. Echinocandins

Echinocandins, i.e., caspofungin, micafungin, and anidulafungin, are the newest class of antifungal drugs available for the treatment of invasive fungal infections and offer an excellent safety profile combined with high fungicidal activity [86,87]. They noncompetitively inhibit (1,3)-β-D-glucan synthase, which is responsible for the biosynthesis of 1,3-β-D-glucan, a crucial structural component of fungal cell walls [88,89]. Specifically, echinocandins target the catalytic subunits FKS1 of β-D-glucan synthase, encoded by FKS1 and FKS2 genes, leading to the disruption of cell wall glucan, osmotic instability, cell lysis, and death for most species (Figure 2B) [90,91]. Although their antifungal spectrum is limited, echinocandins are fungicidal against most Candida spp., including azole-resistant strains and biofilm [92,93]. However, as the use of these drugs has expanded, reports of resistance to echinocandin treatments among Candida spp. have increased [93]. In particular, C. parapsilosis tends to be associated with increased in vitro minimum inhibitory concentrations (MICs) of echinocandin [94,95], raising concerns that such drugs may facilitated the development of high levels of resistance [96,97,98].
Decreased echinocandin susceptibility can occur via two main mechanisms (Figure 2B): (i) an adaptive stress response mechanism, involving a compensatory increase in the synthesis of chitin (an essential cell wall component) that is mediated, for example, via the activation of the calcineurin (Ca2+) signaling pathway. The activation of this pathway is initially signaled by the Hsp90 chaperone, a key regulator of cellular stress response, and thus confers protection against the antifungal agent [99,100,101]; (ii) acquired or intrinsic mutations in genes encoding FKS1 and FKS2, characterized by amino acid substitutions in specific regions clustered around two highly conserved regions (termed hot spots 1 and 2) of Fksp, which is generally correlated with increased resistance to such drugs [95,102,103]. Acquired mutations have been reported for C. albicans, C. tropicalis, C. krusei, and C. glabrata [102,104] but not yet for C. parapsilosis [96,105]. In C. parapsilosis, naturally occurring FKS1 mutations in the hot spot 1 region were found to be responsible for the intrinsic reduced susceptibility of this species to echinocandins [106].

4.3. Azoles

Azoles represent the largest class of antifungal agents in clinical use due to their broad spectrum of activity, favorable safety profile, and bioavailability [73]. The clinically approved azoles include fluconazole (FLC), voriconazole (VRC), posaconazole (PSC), itraconazole, and isavuconazole. Azoles exhibit mainly fungistatic activity against Candida [107]. Due to differences between the membranes of fungal and human cells (mainly composed of cholesterol), the use of azoles does not interfere with human body cells during treatment. They bind to and inhibit the activity of the enzyme lanosterol 14α-demethylase (encoded by the ERG11 gene in yeasts), which is a key enzyme in the ergosterol biosynthetic pathway (Figure 2C) [108,109,110]. Ergosterol is an important component of fungal cell membranes [111]. The interruption of its synthesis enables the accumulation of a toxic 14α-methyl sterol, which impairs the membrane integrity and also the function of some membrane-bound proteins (such as those involved in cell wall synthesis), with consequences in terms of cell growth [108,111,112].
The emergence of azole resistance in Candida species represents a major challenge to treatment [113,114,115,116]. Candida spp. azole resistance has been linked to different molecular mechanisms that include (Figure 2C): (i) mutations in the gene encoding the azole target enzyme lanosterol 14α-demethylase (ERG11), with resulting overexpression, and reduced azole binding, which also results in the reduction in or loss of affinity with azoles, preventing azole binding; (ii) alterations in the ergosterol biosynthetic pathway, caused by loss-of-function point mutations in ERG3, leading to a depletion of ergosterol and to the accumulation of 14α-methyl fecosterol, which is less damaging to cell membranes, thus enabling continued growth in the presence of azoles; and (iii) the upregulation of multidrug efflux pumps CDR1 and CDR2 (Candida drug resistance) and MDR1 (multidrug resistance) genes that transport the drug out of the cells [117,118]. The analysis of serial isolates from individual patients has revealed that acquired azole resistance commonly relies on multiple and often-combined molecular mechanisms [119].
Similarly to C. albicans, C. parapsilosis harbors several genes that have been found to be involved in resistance development. For example, Mrr1p (multidrug resistance regulator 1) is a zinc cluster transcription factor that controls MDR1 expression [120]. Several authors have demonstrated that gain-of-function mutations in the MRR1 gene, which render the transcription factor constitutively active, are responsible for the upregulation of the MDR1 efflux pump and thus play a central role in the development of drug resistance [121,122,123,124]. The hyperactivation of the Tac1 (transcriptional activator of CDR genes 1) transcription factor is also conferred by gain-of-function mutations that consequently promote the overexpression of CDR1 and CDR2 genes [125,126]. Recently, researchers described a new azole resistance mechanism in Candida, particularly among C. parapsilosis isolates, involving another Cdr1-like gene, the CDR1B (CPAR2_304370). Expression of a GOF mutation in the MRR1 gene impacts the fluconazole susceptibility in C. parapsilosis through CDR1B overexpression [114,127]. CDR1 (CLUG_03113) expression in Candida lusitaniae is also shown to be regulated by GOF mutation in MRR1 [128]. In addition, several pieces of evidence point to another mechanism involved in C. parapsilosis antifungal resistance: allele copy number variation. Our group observed an increase in the CDR1B copy number, resulting in CDR1B overexpression and a consequent reduction in fluconazole susceptibility [114]. The copy number variation mechanism has not only been associated with the drug fluconazole but also with miltefosine, a drug recently approved by the FDA for the treatment of invasive candidiasis [129].
Upc2 (Sterol uptake control protein 2), another member of the zinc cluster transcription factor family, is a key regulator of ergosterol metabolism that controls the expression of the azole target ERG11 gene [130,131,132]. Gain-of-function mutations in UPC2 lead to the increased ERG11 expression, contributing to fluconazole resistance in this species [133,134,135]. As with UPC2, the transcription factor Ndt80 also modulates the expression of several ergosterol metabolism genes [132,136]. Moreover, Chen et al. (2004) demonstrated the involvement of this regulatory factor in azole tolerance by controlling the expression of the CDR1 gene in C. albicans [137].
Alterations in the ergosterol biosynthetic pathway, including mutations in the ERG11 gene or its overexpression, have also been linked to azole resistance [138]. The amino acid Y132F substitution in ERG11 is frequently reported among Candida spp., including C. parapsilosis [113,139,140,141,142]. The persistence of C. parapsilosis isolates harboring the Y132F mutation in clinical settings has been associated with outbreaks of infections in hospitals, with fatal consequences [115,116,143].

5. Final Remarks

Candida parapsilosis is a predominant species within NACs that is responsible for invasive candidosis in low-birth-weight neonates, transplant recipients, critical care patients, and those receiving parenteral nutrition. The high prevalence of C. parapsilosis is also promoted by its well-documented ability to persist and thrive in the hospital environments for long periods. Its remarkable ability to adhere to abiotic surfaces, such as catheters, and to form biofilms constitutes a gateway to systemic colonization. The extensive use of antifungals, both prophylactically and therapeutically, is also recognized as a major cause of worldwide antifungal resistance in this pathogen.
In light of the above, there can be no doubt that further comprehensive research efforts addressing the epidemiology, pathogenic attributes, antimicrobial susceptibility profile, and genetic resistance mechanisms of Candida parapsilosis will contribute to improved treatments for and the prevention of infections, leading to improved patient outcomes and lower burdens upon healthcare systems.

Author Contributions

Writing—original draft, J.B.; Writing—review and editing, J.B., I.M.M. and A.G.R. All authors have read and agreed to the published version of the manuscript.

Funding

J.B. is supported by an FCT—Portuguese Foundation for Science and Technology—grant (SFRH/BD/135883/2018). This manuscript was also supported by National Funds through FCT under the scope of the CINTESIS R&D Unit (UIDB/04255/2020 and UIDP/04255/2020).

Data Availability Statement

All data are publicly available.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bongomin, F.; Gago, S.; Oladele, R.O.; Denning, D.W. Global and Multi-National Prevalence of Fungal Diseases-Estimate Precision. J. Fungi 2017, 3, 57. [Google Scholar] [CrossRef]
  2. Strollo, S.; Lionakis, M.S.; Adjemian, J.; Steiner, C.A.; Prevots, D.R. Epidemiology of Hospitalizations Associated with Invasive Candidiasis, United States, 2002-2012(1). Emerg. Infect. Dis. 2016, 23, 7–13. [Google Scholar] [CrossRef] [Green Version]
  3. Pappas, P.G.; Lionakis, M.S.; Arendrup, M.C.; Ostrosky-Zeichner, L.; Kullberg, B.J. Invasive candidiasis. Nat. Rev. Dis. Primers 2018, 4, 18026. [Google Scholar] [CrossRef]
  4. McCarty, T.P.; White, C.M.; Pappas, P.G. Candidemia and Invasive Candidiasis. Infect. Dis. Clin. N. Am. 2021, 35, 389–413. [Google Scholar] [CrossRef]
  5. Guinea, J. Global trends in the distribution of Candida species causing candidemia. Clin. Microbiol. Infect. 2014, 20 (Suppl. S6), 5–10. [Google Scholar] [CrossRef] [Green Version]
  6. Koehler, P.; Stecher, M.; Cornely, O.A.; Koehler, D.; Vehreschild, M.; Bohlius, J.; Wisplinghoff, H.; Vehreschild, J.J. Morbidity and mortality of candidaemia in Europe: An epidemiologic meta-analysis. Clin. Microbiol. Infect. 2019, 25, 1200–1212. [Google Scholar] [CrossRef]
  7. Ashford, B.K. Certain conditions of the gastrointestinal tract in Puerto Rico and their relation to tropical sprue. Am. J. Trop. Med. Hyg. 1928, 8, 507–538. [Google Scholar] [CrossRef]
  8. Tavanti, A.; Davidson, A.D.; Gow, N.A.; Maiden, M.C.; Odds, F.C. Candida orthopsilosis and Candida metapsilosis spp. nov. to replace Candida parapsilosis groups II and III. J. Clin. Microbiol. 2005, 43, 284–292. [Google Scholar] [CrossRef] [Green Version]
  9. van Asbeck, E.C.; Clemons, K.V.; Stevens, D.A. Candida parapsilosis: A review of its epidemiology, pathogenesis, clinical aspects, typing and antimicrobial susceptibility. Crit. Rev. Microbiol. 2009, 35, 283–309. [Google Scholar] [CrossRef]
  10. Bonassoli, L.A.; Bertoli, M.; Svidzinski, T.I. High frequency of Candida parapsilosis on the hands of healthy hosts. J. Hosp. Infect. 2005, 59, 159–162. [Google Scholar] [CrossRef]
  11. van Asbeck, E.C.; Huang, Y.C.; Markham, A.N.; Clemons, K.V.; Stevens, D.A. Candida parapsilosis fungemia in neonates: Genotyping results suggest healthcare workers hands as source, and review of published studies. Mycopathologia 2007, 164, 287–293. [Google Scholar] [CrossRef]
  12. Levin, A.S.; Costa, S.F.; Mussi, N.S.; Basso, M.; Sinto, S.I.; Machado, C.; Geiger, D.C.; Villares, M.C.; Schreiber, A.Z.; Barone, A.A.; et al. Candida parapsilosis fungemia associated with implantable and semi-implantable central venous catheters and the hands of healthcare workers. Diagn. Microbiol. Infect. Dis. 1998, 30, 243–249. [Google Scholar] [CrossRef]
  13. Trofa, D.; Gacser, A.; Nosanchuk, J.D. Candida parapsilosis, an emerging fungal pathogen. Clin. Microbiol. Rev. 2008, 21, 606–625. [Google Scholar] [CrossRef] [Green Version]
  14. Ramage, G.; Martinez, J.P.; Lopez-Ribot, J.L. Candida biofilms on implanted biomaterials: A clinically significant problem. FEMS Yeast Res. 2006, 6, 979–986. [Google Scholar] [CrossRef] [Green Version]
  15. Nemeth, T.; Toth, A.; Szenzenstein, J.; Horvath, P.; Nosanchuk, J.D.; Grozer, Z.; Toth, R.; Papp, C.; Hamari, Z.; Vagvolgyi, C.; et al. Characterization of virulence properties in the C. parapsilosis sensu lato species. PLoS ONE 2013, 8, e68704. [Google Scholar] [CrossRef] [Green Version]
  16. Cuellar-Cruz, M.; Lopez-Romero, E.; Villagomez-Castro, J.C.; Ruiz-Baca, E. Candida species: New insights into biofilm formation. Future Microbiol. 2012, 7, 755–771. [Google Scholar] [CrossRef]
  17. Pammi, M.; Holland, L.; Butler, G.; Gacser, A.; Bliss, J.M. Candida parapsilosis is a Significant Neonatal Pathogen: A Systematic Review and Meta-Analysis. Pediatr. Infect. Dis. J. 2013, 32, e206–e216. [Google Scholar] [CrossRef] [Green Version]
  18. Nucci, M.; Queiroz-Telles, F.; Alvarado-Matute, T.; Tiraboschi, I.N.; Cortes, J.; Zurita, J.; Guzman-Blanco, M.; Santolaya, M.E.; Thompson, L.; Sifuentes-Osornio, J.; et al. Epidemiology of Candidemia in Latin America: A Laboratory-Based Survey. PLoS ONE 2013, 8, e59373. [Google Scholar] [CrossRef] [Green Version]
  19. Rodriguez, L.; Bustamante, B.; Huaroto, L.; Agurto, C.; Illescas, R.; Ramirez, R.; Diaz, A.; Hidalgo, J. A multi-centric Study of Candida bloodstream infection in Lima-Callao, Peru: Species distribution, antifungal resistance and clinical outcomes. PLoS ONE 2017, 12, e0175172. [Google Scholar] [CrossRef] [Green Version]
  20. Faria-Ramos, I.; Neves-Maia, J.; Ricardo, E.; Santos-Antunes, J.; Silva, A.T.; Costa-de-Oliveira, S.; Canton, E.; Rodrigues, A.G.; Pina-Vaz, C. Species distribution and in vitro antifungal susceptibility profiles of yeast isolates from invasive infections during a Portuguese multicenter survey. Eur. J. Clin. Microbiol. Infect. Dis. 2014, 33, 2241–2247. [Google Scholar] [CrossRef]
  21. Guinea, J.; Zaragoza, Ó.; Escribano, P.; Martín-Mazuelos, E.; Pemán, J.; Sánchez-Reus, F.; Cuenca-Estrella, M. Molecular Identification and Antifungal Susceptibility of Yeast Isolates Causing Fungemia Collected in a Population-Based Study in Spain in 2010 and 2011. Antimicrob. Agents Chemother. 2014, 58, 1529–1537. [Google Scholar] [CrossRef] [Green Version]
  22. Tedeschi, S.; Tumietto, F.; Giannella, M.; Bartoletti, M.; Cristini, F.; Cioni, G.; Ambretti, S.; Carretto, E.; Sambri, V.; Sarti, M.; et al. Epidemiology and outcome of candidemia in internal medicine wards: A regional study in Italy. Eur. J. Intern. Med. 2016, 34, 39–44. [Google Scholar] [CrossRef]
  23. Vogiatzi, L.; Ilia, S.; Sideri, G.; Vagelakoudi, E.; Vassilopoulou, M.; Sdougka, M.; Briassoulis, G.; Papadatos, I.; Kalabalikis, P.; Sianidou, L.; et al. Invasive candidiasis in pediatric intensive care in Greece: A nationwide study. Intensive Care Med. 2013, 39, 2188–2195. [Google Scholar] [CrossRef]
  24. Arendrup, M.C.; Bruun, B.; Christensen, J.J.; Fuursted, K.; Johansen, H.K.; Kjældgaard, P.; Knudsen, J.D.; Kristensen, L.; Møller, J.; Nielsen, L.; et al. National Surveillance of Fungemia in Denmark (2004 to 2009). J. Clin. Microbiol. 2011, 49, 325–334. [Google Scholar] [CrossRef] [Green Version]
  25. Ericsson, J.; Chryssanthou, E.; Klingspor, L.; Johansson, A.G.; Ljungman, P.; Svensson, E.; Sjolin, J. Candidaemia in Sweden: A nationwide prospective observational survey. Clin. Microbiol. Infect. 2013, 19, E218–E221. [Google Scholar] [CrossRef] [Green Version]
  26. Chalmers, C.; Gaur, S.; Chew, J.; Wright, T.; Kumar, A.; Mathur, S.; Wan, W.Y.; Gould, I.M.; Leanord, A.; Bal, A.M. Epidemiology and management of candidaemia--a retrospective, multicentre study in five hospitals in the UK. Mycoses 2011, 54, e795–e800. [Google Scholar] [CrossRef]
  27. Toda, M.; Williams, S.R.; Berkow, E.L.; Farley, M.M.; Harrison, L.H.; Bonner, L.; Marceaux, K.M.; Hollick, R.; Zhang, A.Y.; Schaffner, W.; et al. Population-Based Active Surveillance for Culture-Confirmed Candidemia—Four Sites, United States, 2012–2016. MMWR Surveill. Summ. 2019, 68, 1–15. [Google Scholar] [CrossRef]
  28. Ting, J.Y.; Roberts, A.; Synnes, A.; Canning, R.; Bodani, J.; Monterossa, L.; Shah, P.S.; Canadian Neonatal Network, I. Invasive Fungal Infections in Neonates in Canada: Epidemiology and Outcomes. Pediatr. Infect. Dis. J. 2018, 37, 1154–1159. [Google Scholar] [CrossRef]
  29. Tsay, S.V.; Mu, Y.; Williams, S.; Epson, E.; Nadle, J.; Bamberg, W.M.; Barter, D.M.; Johnston, H.L.; Farley, M.M.; Harb, S.; et al. Burden of Candidemia in the United States, 2017. Clin. Infect. Dis. 2020, 71, e449–e453. [Google Scholar] [CrossRef]
  30. Pfaller, M.A.; Diekema, D.J.; Turnidge, J.D.; Castanheira, M.; Jones, R.N. Twenty Years of the SENTRY Antifungal Surveillance Program: Results for Candida Species From 1997–2016. Open Forum Infect. Dis. 2019, 6, S79–S94. [Google Scholar] [CrossRef]
  31. Kakeya, H.; Yamada, K.; Kaneko, Y.; Yanagihara, K.; Tateda, K.; Maesaki, S.; Takesue, Y.; Tomono, K.; Kadota, J.I.; Kaku, M.; et al. National Trends in the Distribution of Candida Species Causing Candidemia in Japan from 2003 to 2014. Med. Mycol. J. 2018, 59, E19–E22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Xiao, M.; Chen, S.C.; Kong, F.; Xu, X.L.; Yan, L.; Kong, H.S.; Fan, X.; Hou, X.; Cheng, J.W.; Zhou, M.L.; et al. Distribution and Antifungal Susceptibility of Candida Species Causing Candidemia in China: An Update From the CHIF-NET Study. J. Infect. Dis. 2020, 221, S139–S147. [Google Scholar] [CrossRef] [PubMed]
  33. Rajni, E.; Chaudhary, P.; Garg, V.K.; Sharma, R.; Malik, M. A complete clinico-epidemiological and microbiological profile of candidemia cases in a tertiary-care hospital in Western India. Antimicrob. Steward. Healthc. Epidemiol. 2022, 2, e37. [Google Scholar] [CrossRef] [PubMed]
  34. Chapman, B.; Slavin, M.; Marriott, D.; Halliday, C.; Kidd, S.; Arthur, I.; Bak, N.; Heath, C.H.; Kennedy, K.; Morrissey, C.O.; et al. Changing epidemiology of candidaemia in Australia. J. Antimicrob. Chemother. 2017, 72, 1103–1108. [Google Scholar] [CrossRef] [Green Version]
  35. Neji, S.; Hadrich, I.; Trabelsi, H.; Abbes, S.; Cheikhrouhou, F.; Sellami, H.; Makni, F.; Ayadi, A. Virulence factors, antifungal susceptibility and molecular mechanisms of azole resistance among Candida parapsilosis complex isolates recovered from clinical specimens. J. Biomed. Sci. 2017, 24, 67. [Google Scholar] [CrossRef] [Green Version]
  36. Németh, T.M.; Gacser, A.; Nosanchuk, J.D. Candida psilosis complex. In Reference Module in Life Sciences; Roitberg, B.D., Ed.; Elsevier: Amsterdam, The Netherlands, 2018. [Google Scholar] [CrossRef]
  37. Skrzypek, M.S.; Binkley, J.; Binkley, G.; Miyasato, S.R.; Simison, M.; Sherlock, G. The Candida Genome Database (CGD): Incorporation of Assembly 22, systematic identifiers and visualization of high throughput sequencing data. Nucleic Acids Res. 2017, 45, D592–D596. [Google Scholar] [CrossRef] [Green Version]
  38. Butler, G.; Rasmussen, M.D.; Lin, M.F.; Santos, M.A.; Sakthikumar, S.; Munro, C.A.; Rheinbay, E.; Grabherr, M.; Forche, A.; Reedy, J.L.; et al. Evolution of pathogenicity and sexual reproduction in eight Candida genomes. Nature 2009, 459, 657–662. [Google Scholar] [CrossRef] [Green Version]
  39. Laffey, S.F.; Butler, G. Phenotype switching affects biofilm formation by Candida parapsilosis. Microbiology 2005, 151, 1073–1081. [Google Scholar] [CrossRef] [Green Version]
  40. Cavalheiro, M.; Teixeira, M.C. Candida Biofilms: Threats, Challenges, and Promising Strategies. Front. Med. 2018, 5, 28. [Google Scholar] [CrossRef]
  41. Silva-Dias, A.; Miranda, I.M.; Branco, J.; Monteiro-Soares, M.; Pina-Vaz, C.; Rodrigues, A.G. Adhesion, biofilm formation, cell surface hydrophobicity, and antifungal planktonic susceptibility: Relationship among Candida spp. Front. Microbiol. 2015, 6, 205. [Google Scholar] [CrossRef] [Green Version]
  42. Silva, S.; Negri, M.; Henriques, M.; Oliveira, R.; Williams, D.W.; Azeredo, J. Adherence and biofilm formation of non-Candida albicans Candida species. Trends Microbiol. 2011, 19, 241–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Chandra, J.; Patel, J.D.; Li, J.; Zhou, G.; Mukherjee, P.K.; McCormick, T.S.; Anderson, J.M.; Ghannoum, M.A. Modification of surface properties of biomaterials influences the ability of Candida albicans to form biofilms. Appl. Environ. Microbiol. 2005, 71, 8795–8801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. de Groot, P.W.; Bader, O.; de Boer, A.D.; Weig, M.; Chauhan, N. Adhesins in human fungal pathogens: Glue with plenty of stick. Eukaryot. Cell 2013, 12, 470–481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Modrzewska, B.; Kurnatowski, P. Adherence of Candida sp. to host tissues and cells as one of its pathogenicity features. Ann. Parasitol. 2015, 61, 3–9. [Google Scholar]
  46. Nobile, C.J.; Andes, D.R.; Nett, J.E.; Smith, F.J.; Yue, F.; Phan, Q.T.; Edwards, J.E.; Filler, S.G.; Mitchell, A.P. Critical role of Bcr1-dependent adhesins in C. albicans biofilm formation in vitro and in vivo. PLoS Pathog. 2006, 2, e63. [Google Scholar] [CrossRef] [PubMed]
  47. Bertini, A.; Zoppo, M.; Lombardi, L.; Rizzato, C.; De Carolis, E.; Vella, A.; Torelli, R.; Sanguinetti, M.; Tavanti, A. Targeted gene disruption in Candida parapsilosis demonstrates a role for CPAR2_404800 in adhesion to a biotic surface and in a murine model of ascending urinary tract infection. Virulence 2016, 7, 85–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Neale, M.N.; Glass, K.A.; Longley, S.J.; Kim, D.J.; Laforce-Nesbitt, S.S.; Wortzel, J.D.; Shaw, S.K.; Bliss, J.M. Role of the Inducible Adhesin CpAls7 in Binding of Candida parapsilosis to the Extracellular Matrix under Fluid Shear. Infect. Immun. 2018, 86, e00892-17. [Google Scholar] [CrossRef] [Green Version]
  49. Czechowicz, P.; Nowicka, J.; Gosciniak, G. Virulence Factors of Candida spp. and Host Immune Response Important in the Pathogenesis of Vulvovaginal Candidiasis. Int. J. Mol. Sci. 2022, 23, 5895. [Google Scholar] [CrossRef]
  50. Ruchel, R.; Boning, B.; Borg, M. Characterization of a secretory proteinase of Candida parapsilosis and evidence for the absence of the enzyme during infection in vitro. Infect. Immun. 1986, 53, 411–419. [Google Scholar] [CrossRef] [Green Version]
  51. Horvath, P.; Nosanchuk, J.D.; Hamari, Z.; Vagvolgyi, C.; Gacser, A. The identification of gene duplication and the role of secreted aspartyl proteinase 1 in Candida parapsilosis virulence. J. Infect. Dis. 2012, 205, 923–933. [Google Scholar] [CrossRef]
  52. Dagdeviren, M.; Cerikcioglu, N.; Karavus, M. Acid proteinase, phospholipase and adherence properties of Candida parapsilosis strains isolated from clinical specimens of hospitalised patients. Mycoses 2005, 48, 321–326. [Google Scholar] [CrossRef] [PubMed]
  53. Tosun, I.; Akyuz, Z.; Guler, N.C.; Gulmez, D.; Bayramoglu, G.; Kaklikkaya, N.; Arikan-Akdagli, S.; Aydin, F. Distribution, virulence attributes and antifungal susceptibility patterns of Candida parapsilosis complex strains isolated from clinical samples. Med. Mycol. 2013, 51, 483–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Gacser, A.; Trofa, D.; Schafer, W.; Nosanchuk, J.D. Targeted gene deletion in Candida parapsilosis demonstrates the role of secreted lipase in virulence. J. Clin. Investig. 2007, 117, 3049–3058. [Google Scholar] [CrossRef] [Green Version]
  55. Trofa, D.; Agovino, M.; Stehr, F.; Schafer, W.; Rykunov, D.; Fiser, A.; Hamari, Z.; Nosanchuk, J.D.; Gacser, A. Acetylsalicylic acid (aspirin) reduces damage to reconstituted human tissues infected with Candida species by inhibiting extracellular fungal lipases. Microbes. Infect. 2009, 11, 1131–1139. [Google Scholar] [CrossRef] [Green Version]
  56. Ghannoum, M.A. Potential role of phospholipases in virulence and fungal pathogenesis. Clin. Microbiol. Rev. 2000, 13, 122–143. [Google Scholar] [CrossRef]
  57. Mukherjee, P.K.; Chandra, J. Candida biofilm resistance. Drug Resist. Updat. 2004, 7, 301–309. [Google Scholar] [CrossRef]
  58. Chandra, J.; Mukherjee, P.K. Candida Biofilms: Development, Architecture, and Resistance. Microbiol. Spectr. 2015, 3. [Google Scholar] [CrossRef] [Green Version]
  59. Douglas, L.J. Candida biofilms and their role in infection. Trends Microbiol. 2003, 11, 30–36. [Google Scholar] [CrossRef]
  60. Estivill, D.; Arias, A.; Torres-Lana, A.; Carrillo-Munoz, A.J.; Arevalo, M.P. Biofilm formation by five species of Candida on three clinical materials. J. Microbiol. Methods 2011, 86, 238–242. [Google Scholar] [CrossRef]
  61. Shin, J.H.; Kee, S.J.; Shin, M.G.; Kim, S.H.; Shin, D.H.; Lee, S.K.; Suh, S.P.; Ryang, D.W. Biofilm production by isolates of Candida species recovered from nonneutropenic patients: Comparison of bloodstream isolates with isolates from other sources. J. Clin. Microbiol. 2002, 40, 1244–1248. [Google Scholar] [CrossRef]
  62. Branchini, M.L.; Pfaller, M.A.; Rhine-Chalberg, J.; Frempong, T.; Isenberg, H.D. Genotypic variation and slime production among blood and catheter isolates of Candida parapsilosis. J. Clin. Microbiol. 1994, 32, 452–456. [Google Scholar] [CrossRef] [Green Version]
  63. Silva, S.; Henriques, M.; Martins, A.; Oliveira, R.; Williams, D.; Azeredo, J. Biofilms of non-Candida albicans Candida species: Quantification, structure and matrix composition. Med. Mycol. 2009, 47, 681–689. [Google Scholar] [CrossRef] [Green Version]
  64. Kuhn, D.M.; Chandra, J.; Mukherjee, P.K.; Ghannoum, M.A. Comparison of biofilms formed by Candida albicans and Candida parapsilosis on bioprosthetic surfaces. Infect. Immun. 2002, 70, 878–888. [Google Scholar] [CrossRef] [Green Version]
  65. Mitchell, K.F.; Zarnowski, R.; Sanchez, H.; Edward, J.A.; Reinicke, E.L.; Nett, J.E.; Mitchell, A.P.; Andes, D.R. Community participation in biofilm matrix assembly and function. Proc. Natl. Acad. Sci. USA 2015, 112, 4092–4097. [Google Scholar] [CrossRef] [Green Version]
  66. Silva, S.; Rodrigues, C.F.; Araujo, D.; Rodrigues, M.E.; Henriques, M. Candida Species Biofilms’ Antifungal Resistance. J. Fungi 2017, 3, 8. [Google Scholar] [CrossRef] [Green Version]
  67. Uppuluri, P.; Chaturvedi, A.K.; Srinivasan, A.; Banerjee, M.; Ramasubramaniam, A.K.; Kohler, J.R.; Kadosh, D.; Lopez-Ribot, J.L. Dispersion as an important step in the Candida albicans biofilm developmental cycle. PLoS Pathog. 2010, 6, e1000828. [Google Scholar] [CrossRef]
  68. Holland, L.M.; Schroder, M.S.; Turner, S.A.; Taff, H.; Andes, D.; Grozer, Z.; Gacser, A.; Ames, L.; Haynes, K.; Higgins, D.G.; et al. Comparative phenotypic analysis of the major fungal pathogens Candida parapsilosis and Candida albicans. PLoS Pathog. 2014, 10, e1004365. [Google Scholar] [CrossRef] [Green Version]
  69. Nobile, C.J.; Fox, E.P.; Nett, J.E.; Sorrells, T.R.; Mitrovich, Q.M.; Hernday, A.D.; Tuch, B.B.; Andes, D.R.; Johnson, A.D. A recently evolved transcriptional network controls biofilm development in Candida albicans. Cell 2012, 148, 126–138. [Google Scholar] [CrossRef] [Green Version]
  70. Branco, J.; Martins-Cruz, C.; Rodrigues, L.; Silva, R.M.; Araujo-Gomes, N.; Goncalves, T.; Miranda, I.M.; Rodrigues, A.G. The transcription factor Ndt80 is a repressor of Candida parapsilosis virulence attributes. Virulence 2021, 12, 601–614. [Google Scholar] [CrossRef]
  71. Ding, C.; Vidanes, G.M.; Maguire, S.L.; Guida, A.; Synnott, J.M.; Andes, D.R.; Butler, G. Conserved and divergent roles of Bcr1 and CFEM proteins in Candida parapsilosis and Candida albicans. PLoS ONE 2011, 6, e28151. [Google Scholar] [CrossRef]
  72. Denning, D.W.; Hope, W.W. Therapy for fungal diseases: Opportunities and priorities. Trends Microbiol. 2010, 18, 195–204. [Google Scholar] [CrossRef] [PubMed]
  73. Ben-Ami, R.; Kontoyiannis, D.P. Resistance to Antifungal Drugs. Infect. Dis. Clin. N. Am. 2021, 35, 279–311. [Google Scholar] [CrossRef] [PubMed]
  74. Carolus, H.; Pierson, S.; Lagrou, K.; Van Dijck, P. Amphotericin B and Other Polyenes-Discovery, Clinical Use, Mode of Action and Drug Resistance. J. Fungi 2020, 6, 321. [Google Scholar] [CrossRef] [PubMed]
  75. Fanos, V.; Cataldi, L. Amphotericin B-induced nephrotoxicity: A review. J. Chemother. 2000, 12, 463–470. [Google Scholar] [CrossRef]
  76. Groll, A.H.; Rijnders, B.J.A.; Walsh, T.J.; Adler-Moore, J.; Lewis, R.E.; Bruggemann, R.J.M. Clinical Pharmacokinetics, Pharmacodynamics, Safety and Efficacy of Liposomal Amphotericin B. Clin. Infect. Dis. 2019, 68, S260–S274. [Google Scholar] [CrossRef]
  77. Lockhart, S.R.; Iqbal, N.; Cleveland, A.A.; Farley, M.M.; Harrison, L.H.; Bolden, C.B.; Baughman, W.; Stein, B.; Hollick, R.; Park, B.J.; et al. Species identification and antifungal susceptibility testing of Candida bloodstream isolates from population-based surveillance studies in two U.S. cities from 2008 to 2011. J. Clin. Microbiol. 2012, 50, 3435–3442. [Google Scholar] [CrossRef] [Green Version]
  78. Yamin, D.; Akanmu, M.H.; Al Mutair, A.; Alhumaid, S.; Rabaan, A.A.; Hajissa, K. Global Prevalence of Antifungal-Resistant Candida parapsilosis: A Systematic Review and Meta-Analysis. Trop. Med. Infect. Dis. 2022, 7, 188. [Google Scholar] [CrossRef]
  79. Chowdhary, A.; Prakash, A.; Sharma, C.; Kordalewska, M.; Kumar, A.; Sarma, S.; Tarai, B.; Singh, A.; Upadhyaya, G.; Upadhyay, S.; et al. A multicentre study of antifungal susceptibility patterns among 350 Candida auris isolates (2009-17) in India: Role of the ERG11 and FKS1 genes in azole and echinocandin resistance. J. Antimicrob. Chemother. 2018, 73, 891–899. [Google Scholar] [CrossRef]
  80. Martel, C.M.; Parker, J.E.; Bader, O.; Weig, M.; Gross, U.; Warrilow, A.G.; Kelly, D.E.; Kelly, S.L. A clinical isolate of Candida albicans with mutations in ERG11 (encoding sterol 14alpha-demethylase) and ERG5 (encoding C22 desaturase) is cross resistant to azoles and amphotericin B. Antimicrob. Agents Chemother. 2010, 54, 3578–3583. [Google Scholar] [CrossRef] [Green Version]
  81. Young, L.Y.; Hull, C.M.; Heitman, J. Disruption of ergosterol biosynthesis confers resistance to amphotericin B in Candida lusitaniae. Antimicrob. Agents Chemother. 2003, 47, 2717–2724. [Google Scholar] [CrossRef]
  82. Perlin, D.S. Echinocandin Resistance in Candida. Clin. Infect. Dis. 2015, 61 (Suppl. S6), S612–S617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Sokol-Anderson, M.; Sligh, J.E., Jr.; Elberg, S.; Brajtburg, J.; Kobayashi, G.S.; Medoff, G. Role of cell defense against oxidative damage in the resistance of Candida albicans to the killing effect of amphotericin B. Antimicrob. Agents Chemother. 1988, 32, 702–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Kristanc, L.; Bozic, B.; Jokhadar, S.Z.; Dolenc, M.S.; Gomiscek, G. The pore-forming action of polyenes: From model membranes to living organisms. Biochim. Biophys. Acta Biomembr. 2019, 1861, 418–430. [Google Scholar] [CrossRef] [PubMed]
  85. Cowen, L.E.; Lindquist, S. Hsp90 potentiates the rapid evolution of new traits: Drug resistance in diverse fungi. Science 2005, 309, 2185–2189. [Google Scholar] [CrossRef]
  86. Denning, D.W. Echinocandin antifungal drugs. Lancet 2003, 362, 1142–1151. [Google Scholar] [CrossRef]
  87. Pappas, P.G.; Kauffman, C.A.; Andes, D.R.; Clancy, C.J.; Marr, K.A.; Ostrosky-Zeichner, L.; Reboli, A.C.; Schuster, M.G.; Vazquez, J.A.; Walsh, T.J.; et al. Clinical Practice Guideline for the Management of Candidiasis: 2016 Update by the Infectious Diseases Society of America. Clin. Infect. Dis. 2016, 62, e1–e50. [Google Scholar] [CrossRef] [Green Version]
  88. Chen, S.A.; Slavin, M.; Sorrell, T. Echinocandin Antifungal Drugs in Fungal Infections. Drugs 2011, 71, 11–41. [Google Scholar] [CrossRef]
  89. Beyda, N.D.; Lewis, R.E.; Garey, K.W. Echinocandin Resistance in Candida Species: Mechanisms of Reduced Susceptibility and Therapeutic Approaches. Ann. Pharmacother. 2012, 46, 1086–1096. [Google Scholar] [CrossRef]
  90. Pfaller, M.A.; Diekema, D.J.; Andes, D.; Arendrup, M.C.; Brown, S.D.; Lockhart, S.R.; Motyl, M.; Perlin, D.S. Clinical breakpoints for the echinocandins and Candida revisited: Integration of molecular, clinical, and microbiological data to arrive at species-specific interpretive criteria. Drug Resist. Updat. 2011, 14, 164–176. [Google Scholar] [CrossRef]
  91. Douglas, C.M.; D'Ippolito, J.A.; Shei, G.J.; Meinz, M.; Onishi, J.; Marrinan, J.A.; Li, W.; Abruzzo, G.K.; Flattery, A.; Bartizal, K.; et al. Identification of the FKS1 gene of Candida albicans as the essential target of 1,3-beta-D-glucan synthase inhibitors. Antimicrob. Agents Chemother. 1997, 41, 2471–2479. [Google Scholar] [CrossRef]
  92. Bachmann, S.P.; VandeWalle, K.; Ramage, G.; Patterson, T.F.; Wickes, B.L.; Graybill, J.R.; López-Ribot, J.L. In Vitro Activity of Caspofungin against Candida albicans Biofilms. Antimicrob. Agents Chemother. 2002, 46, 3591–3596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Perlin, D.S. Current perspectives on echinocandin class drugs. Future Microbiol. 2011, 6, 441–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Ostrosky-Zeichner, L.; Rex, J.H.; Pappas, P.G.; Hamill, R.J.; Larsen, R.A.; Horowitz, H.W.; Powderly, W.G.; Hyslop, N.; Kauffman, C.A.; Cleary, J.; et al. Antifungal Susceptibility Survey of 2,000 Bloodstream Candida Isolates in the United States. Antimicrob. Agents Chemother. 2003, 47, 3149–3154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Perlin, D.S. Resistance to echinocandin-class antifungal drugs. Drug Resist. Updat. 2007, 10, 121–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Garcia-Effron, G.; Canton, E.; Pemán, J.; Dilger, A.; Romá, E.; Perlin, D.S. Epidemiology and echinocandin susceptibility of Candida parapsilosis sensu lato species isolated from bloodstream infections at a Spanish university hospital. J. Antimicrob. Chemother. 2012, 67, 2739–2748. [Google Scholar] [CrossRef] [Green Version]
  97. Siopi, M.; Papadopoulos, A.; Spiliopoulou, A.; Paliogianni, F.; Abou-Chakra, N.; Arendrup, M.C.; Damoulari, C.; Tsioulos, G.; Giannitsioti, E.; Frantzeskaki, F.; et al. Pan-Echinocandin Resistant C. parapsilosis Harboring an F652S Fks1 Alteration in a Patient with Prolonged Echinocandin Therapy. J. Fungi 2022, 8, 931. [Google Scholar] [CrossRef]
  98. Ning, Y.; Xiao, M.; Perlin, D.S.; Zhao, Y.; Lu, M.; Li, Y.; Luo, Z.; Dai, R.; Li, S.; Xu, J.; et al. Decreased echinocandin susceptibility in Candida parapsilosis causing candidemia and emergence of a pan-echinocandin resistant case in China. Emerg. Microbes Infect. 2023, 12, 2153086. [Google Scholar] [CrossRef]
  99. Singh, S.D.; Robbins, N.; Zaas, A.K.; Schell, W.A.; Perfect, J.R.; Cowen, L.E. Hsp90 governs echinocandin resistance in the pathogenic yeast Candida albicans via calcineurin. PLoS Pathog. 2009, 5, e1000532. [Google Scholar] [CrossRef]
  100. Munro, C.A.; Selvaggini, S.; De Bruijn, I.; Walker, L.; Lenardon, M.D.; Gerssen, B.; Milne, S.; Brown, A.J.P.; Gow, N.A.R. The PKC, HOG and Ca2+ signalling pathways co-ordinately regulate chitin synthesis in Candida albicans. Mol. Microbiol. 2007, 63, 1399–1413. [Google Scholar] [CrossRef] [Green Version]
  101. Lee, K.K.; Maccallum, D.M.; Jacobsen, M.D.; Walker, L.A.; Odds, F.C.; Gow, N.A.; Munro, C.A. Elevated cell wall chitin in Candida albicans confers echinocandin resistance in vivo. Antimicrob. Agents Chemother. 2012, 56, 208–217. [Google Scholar] [CrossRef]
  102. Park, S.; Kelly, R.; Kahn, J.N.; Robles, J.; Hsu, M.-J.; Register, E.; Li, W.; Vyas, V.; Fan, H.; Abruzzo, G.; et al. Specific Substitutions in the Echinocandin Target Fks1p Account for Reduced Susceptibility of Rare Laboratory and Clinical Candida sp. Isolates. Antimicrob. Agents Chemother. 2005, 49, 3264–3273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Katiyar, S.; Pfaller, M.; Edlind, T. Candida albicans and Candida glabrata Clinical Isolates Exhibiting Reduced Echinocandin Susceptibility. Antimicrob. Agents Chemother. 2006, 50, 2892–2894. [Google Scholar] [CrossRef] [Green Version]
  104. Garcia-Effron, G.; Kontoyiannis, D.P.; Lewis, R.E.; Perlin, D.S. Caspofungin-Resistant Candida tropicalis Strains Causing Breakthrough Fungemia in Patients at High Risk for Hematologic Malignancies. Antimicrob. Agents Chemother. 2008, 52, 4181–4183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Pfaller, M.A.; Messer, S.A.; Woosley, L.N.; Jones, R.N.; Castanheira, M. Echinocandin and Triazole Antifungal Susceptibility Profiles for Clinical Opportunistic Yeast and Mold Isolates Collected from 2010 to 2011: Application of New CLSI Clinical Breakpoints and Epidemiological Cutoff Values for Characterization of Geographic and Temporal Trends of Antifungal Resistance. J. Clin. Microbiol. 2013, 51, 2571–2581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Garcia-Effron, G.; Katiyar, S.K.; Park, S.; Edlind, T.D.; Perlin, D.S. A Naturally Occurring Proline-to-Alanine Amino Acid Change in Fks1p in Candida parapsilosis, Candida orthopsilosis, and Candida metapsilosis Accounts for Reduced Echinocandin Susceptibility. Antimicrob. Agents Chemother. 2008, 52, 2305–2312. [Google Scholar] [CrossRef] [Green Version]
  107. Robbins, N.; Caplan, T.; Cowen, L.E. Molecular Evolution of Antifungal Drug Resistance. Annu. Rev. Microbiol. 2017, 71, 753–775. [Google Scholar] [CrossRef] [Green Version]
  108. Odds, F.C.; Brown, A.J.P.; Gow, N.A.R. Antifungal agents: Mechanisms of action. Trends Microbiol. 2003, 11, 272–279. [Google Scholar] [CrossRef] [PubMed]
  109. Cowen, L.E.; Sanglard, D.; Howard, S.J.; Rogers, P.D.; Perlin, D.S. Mechanisms of Antifungal Drug Resistance. Cold Spring Harb. Perspect. Med. 2014, 5, a019752. [Google Scholar] [CrossRef]
  110. Xiao, L.; Madison, V.; Chau, A.S.; Loebenberg, D.; Palermo, R.E.; McNicholas, P.M. Three-Dimensional Models of Wild-Type and Mutated Forms of Cytochrome P450 14α-Sterol Demethylases from Aspergillus fumigatus and Candida albicans Provide Insights into Posaconazole Binding. Antimicrob. Agents Chemother. 2004, 48, 568–574. [Google Scholar] [CrossRef] [Green Version]
  111. Akins, R.A. An update on antifungal targets and mechanisms of resistance in Candida albicans. Med. Mycol. 2005, 43, 285–318. [Google Scholar] [CrossRef]
  112. Xie, J.L.; Polvi, E.J.; Shekhar-Guturja, T.; Cowen, L.E. Elucidating drug resistance in human fungal pathogens. Future Microbiol. 2014, 9, 523–542. [Google Scholar] [CrossRef] [PubMed]
  113. Magobo, R.E.; Lockhart, S.R.; Govender, N.P. Fluconazole-resistant Candida parapsilosis strains with a Y132F substitution in the ERG11 gene causing invasive infections in a neonatal unit, South Africa. Mycoses 2020, 63, 471–477. [Google Scholar] [CrossRef]
  114. Branco, J.; Ryan, A.P.; Silva, A.P.; Butler, G.; Miranda, I.M.; Rodrigues, A.G. Clinical azole cross-resistance in Candida parapsilosis is related to a novel MRR1 gain-of-function mutation. Clin. Microbiol. Infect. 2022, 28, 1655. [Google Scholar] [CrossRef] [PubMed]
  115. Martini, C.; Torelli, R.; de Groot, T.; De Carolis, E.; Morandotti, G.A.; De Angelis, G.; Posteraro, B.; Meis, J.F.; Sanguinetti, M. Prevalence and Clonal Distribution of Azole-Resistant Candida parapsilosis Isolates Causing Bloodstream Infections in a Large Italian Hospital. Front. Cell Infect. Microbiol. 2020, 10, 232. [Google Scholar] [CrossRef] [PubMed]
  116. Fekkar, A.; Blaize, M.; Bougle, A.; Normand, A.C.; Raoelina, A.; Kornblum, D.; Kamus, L.; Piarroux, R.; Imbert, S. Hospital outbreak of fluconazole-resistant Candida parapsilosis: Arguments for clonal transmission and long-term persistence. Antimicrob. Agents Chemother. 2021, 65, e02036-20. [Google Scholar] [CrossRef]
  117. Sanglard, D.; Odds, F.C. Resistance of Candida species to antifungal agents: Molecular mechanisms and clinical consequences. Lancet Infect. Dis. 2002, 2, 73–85. [Google Scholar] [CrossRef]
  118. Pfaller, M.A. Antifungal Drug Resistance: Mechanisms, Epidemiology, and Consequences for Treatment. Am. J. Med. 2012, 125, S3–S13. [Google Scholar] [CrossRef]
  119. Morio, F.; Pagniez, F.; Besse, M.; Gay-andrieu, F.; Miegeville, M.; Le Pape, P. Deciphering azole resistance mechanisms with a focus on transcription factor-encoding genes TAC1, MRR1 and UPC2 in a set of fluconazole-resistant clinical isolates of Candida albicans. Int. J. Antimicrob. Agents 2013, 42, 410–415. [Google Scholar] [CrossRef]
  120. Morschhäuser, J.; Barker, K.S.; Liu, T.T.; Blaß-Warmuth, J.; Homayouni, R.; Rogers, P.D. The Transcription Factor Mrr1p Controls Expression of the MDR1 Efflux Pump and Mediates Multidrug Resistance in Candida albicans. PLoS Pathog. 2007, 3, e164. [Google Scholar] [CrossRef] [Green Version]
  121. Dunkel, N.; Blaß, J.; Rogers, P.D.; Morschhäuser, J. Mutations in the multi-drug resistance regulator MRR1, followed by loss of heterozygosity, are the main cause of MDR1 overexpression in fluconazole-resistant Candida albicans strains. Mol. Microbiol. 2008, 69, 827–840. [Google Scholar] [CrossRef]
  122. Schubert, S.; Rogers, P.D.; Morschhäuser, J. Gain-of-Function Mutations in the Transcription Factor MRR1 Are Responsible for Overexpression of the MDR1 Efflux Pump in Fluconazole-Resistant Candida dubliniensis Strains. Antimicrob. Agents Chemother. 2008, 52, 4274–4280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Branco, J.; Silva, A.P.; Silva, R.M.; Silva-Dias, A.; Pina-Vaz, C.; Butler, G.; Rodrigues, A.G.; Miranda, I.M. Fluconazole and Voriconazole Resistance in Candida parapsilosis Is Conferred by Gain-of-Function Mutations in MRR1 Transcription Factor Gene. Antimicrob. Agents Chemother. 2015, 59, 6629–6633. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Papp, C.; Bohner, F.; Kocsis, K.; Varga, M.; Szekeres, A.; Bodai, L.; Willis, J.R.; Gabaldon, T.; Toth, R.; Nosanchuk, J.D.; et al. Triazole Evolution of Candida parapsilosis Results in Cross-Resistance to Other Antifungal Drugs, Influences Stress Responses, and Alters Virulence in an Antifungal Drug-Dependent Manner. mSphere 2020, 5, e00821-20. [Google Scholar] [CrossRef] [PubMed]
  125. Coste, A.T.; Karababa, M.; Ischer, F.; Bille, J.; Sanglard, D. TAC1, transcriptional activator of CDR genes, is a new transcription factor involved in the regulation of Candida albicans ABC transporters CDR1 and CDR2. Eukaryot. Cell 2004, 3, 1639–1652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Berkow, E.L.; Manigaba, K.; Parker, J.E.; Barker, K.S.; Kelly, S.L.; Rogers, P.D. Multidrug Transporters and Alterations in Sterol Biosynthesis Contribute to Azole Antifungal Resistance in Candida parapsilosis. Antimicrob. Agents Chemother. 2015, 59, 5942–5950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Doorley, L.A.; Rybak, J.M.; Berkow, E.L.; Zhang, Q.; Morschhauser, J.; Rogers, P.D. Candida parapsilosis Mdr1B and Cdr1B Are Drivers of Mrr1-Mediated Clinical Fluconazole Resistance. Antimicrob. Agents Chemother. 2022, 66, e0028922. [Google Scholar] [CrossRef] [PubMed]
  128. Borgeat, V.; Brandalise, D.; Grenouillet, F.; Sanglard, D. Participation of the ABC Transporter CDR1 in Azole Resistance of Candida lusitaniae. J. Fungi 2021, 7, 760. [Google Scholar] [CrossRef]
  129. Bergin, S.A.; Zhao, F.; Ryan, A.P.; Muller, C.A.; Nieduszynski, C.A.; Zhai, B.; Rolling, T.; Hohl, T.M.; Morio, F.; Scully, J.; et al. Systematic Analysis of Copy Number Variations in the Pathogenic Yeast Candida parapsilosis Identifies a Gene Amplification in RTA3 That is Associated with Drug Resistance. mBio 2022, 13, e0177722. [Google Scholar] [CrossRef]
  130. Silver, P.M.; Oliver, B.G.; White, T.C. Role of Candida albicans Transcription Factor Upc2p in Drug Resistance and Sterol Metabolism. Eukaryot. Cell 2004, 3, 1391–1397. [Google Scholar] [CrossRef] [Green Version]
  131. Schubert, S.; Barker, K.S.; Znaidi, S.; Schneider, S.; Dierolf, F.; Dunkel, N.; Aïd, M.; Boucher, G.; Rogers, P.D.; Raymond, M.; et al. Regulation of Efflux Pump Expression and Drug Resistance by the Transcription Factors Mrr1, Upc2, and Cap1 in Candida albicans. Antimicrob. Agents Chemother. 2011, 55, 2212–2223. [Google Scholar] [CrossRef]
  132. Branco, J.; Ola, M.; Silva, R.M.; Fonseca, E.; Gomes, N.C.; Martins-Cruz, C.; Silva, A.P.; Silva-Dias, A.; Pina-Vaz, C.; Erraught, C.; et al. Impact of ERG3 mutations and expression of ergosterol genes controlled by UPC2 and NDT80 in Candida parapsilosis azole resistance. Clin. Microbiol. Infect. 2017, 23, 575. [Google Scholar] [CrossRef] [PubMed]
  133. Heilmann, C.J.; Schneider, S.; Barker, K.S.; Rogers, P.D.; Morschhäuser, J. An A643T Mutation in the Transcription Factor Upc2p Causes Constitutive ERG11 Upregulation and Increased Fluconazole Resistance in Candida albicans. Antimicrob. Agents Chemother. 2010, 54, 353–359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Flowers, S.A.; Barker, K.S.; Berkow, E.L.; Toner, G.; Chadwick, S.G.; Gygax, S.E.; Morschhäuser, J.; Rogers, P.D. Gain-of-Function Mutations in UPC2 Are a Frequent Cause of ERG11 Upregulation in Azole-Resistant Clinical Isolates of Candida albicans. Eukaryot. Cell 2012, 11, 1289–1299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Dunkel, N.; Liu, T.T.; Barker, K.S.; Homayouni, R.; Morschhäuser, J.; Rogers, P.D. A Gain-of-Function Mutation in the Transcription Factor Upc2p Causes Upregulation of Ergosterol Biosynthesis Genes and Increased Fluconazole Resistance in a Clinical Candida albicans Isolate. Eukaryot. Cell 2008, 7, 1180–1190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Sellam, A.; Tebbji, F.; Nantel, A. Role of Ndt80p in Sterol Metabolism Regulation and Azole Resistance in Candida albicans. Eukaryot. Cell 2009, 8, 1174–1183. [Google Scholar] [CrossRef] [Green Version]
  137. Chen, C.-G.; Yang, Y.-L.; Shih, H.-I.; Su, C.-L.; Lo, H.-J. CaNdt80 Is Involved in Drug Resistance in Candida albicans by Regulating CDR1. Antimicrob. Agents Chemother. 2004, 48, 4505–4512. [Google Scholar] [CrossRef] [Green Version]
  138. Pristov, K.E.; Ghannoum, M.A. Resistance of Candida to azoles and echinocandins worldwide. Clin. Microbiol. Infect. 2019, 25, 792–798. [Google Scholar] [CrossRef]
  139. Morio, F.; Loge, C.; Besse, B.; Hennequin, C.; Le Pape, P. Screening for amino acid substitutions in the Candida albicans Erg11 protein of azole-susceptible and azole-resistant clinical isolates: New substitutions and a review of the literature. Diagn. Microbiol. Infect. Dis. 2010, 66, 373–384. [Google Scholar] [CrossRef]
  140. Vandeputte, P.; Larcher, G.; Berges, T.; Renier, G.; Chabasse, D.; Bouchara, J.P. Mechanisms of azole resistance in a clinical isolate of Candida tropicalis. Antimicrob. Agents Chemother. 2005, 49, 4608–4615. [Google Scholar] [CrossRef] [Green Version]
  141. Healey, K.R.; Kordalewska, M.; Jimenez Ortigosa, C.; Singh, A.; Berrio, I.; Chowdhary, A.; Perlin, D.S. Limited ERG11 Mutations Identified in Isolates of Candida auris Directly Contribute to Reduced Azole Susceptibility. Antimicrob. Agents Chemother. 2018, 62, e01427-18. [Google Scholar] [CrossRef]
  142. Singh, A.; Singh, P.K.; de Groot, T.; Kumar, A.; Mathur, P.; Tarai, B.; Sachdeva, N.; Upadhyaya, G.; Sarma, S.; Meis, J.F.; et al. Emergence of clonal fluconazole-resistant Candida parapsilosis clinical isolates in a multicentre laboratory-based surveillance study in India. J. Antimicrob. Chemother. 2019, 74, 1260–1268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Arastehfar, A.; Daneshnia, F.; Hilmioglu-Polat, S.; Fang, W.; Yasar, M.; Polat, F.; Metin, D.Y.; Rigole, P.; Coenye, T.; Ilkit, M.; et al. First Report of Candidemia Clonal Outbreak Caused by Emerging Fluconazole-Resistant Candida parapsilosis Isolates Harboring Y132F and/or Y132F+K143R in Turkey. Antimicrob. Agents Chemother. 2020, 64, e01001-20. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Illustration of biofilm formation cycle in Candida spp. Biofilm development consists of three stages: an early phase, in which cells adhere to biotic or abiotic surfaces; an intermediate phase, involving cell proliferation and the formation of a mixed population; and, finally, a maturation/dispersion phase, characterized by the production of the extracellular matrix and the massive dispersion of cells. The detachment and dispersion of daughter cells occurs in all stages of biofilm development.
Figure 1. Illustration of biofilm formation cycle in Candida spp. Biofilm development consists of three stages: an early phase, in which cells adhere to biotic or abiotic surfaces; an intermediate phase, involving cell proliferation and the formation of a mixed population; and, finally, a maturation/dispersion phase, characterized by the production of the extracellular matrix and the massive dispersion of cells. The detachment and dispersion of daughter cells occurs in all stages of biofilm development.
Jof 09 00080 g001
Figure 2. Mechanism of action of antifungals against Candida spp. and mechanisms underlying drug resistance. (A) Polyenes act by forming polyene/ergosterol aggregates, destabilizing the fungal membrane. The action of polyenes can be overcome through mutations in ergosterol biosynthesis genes responsible for altered sterol composition and by the activation of stress response pathways, such as catalase and Hsp. (B) Echinocandins act as noncompetitive inhibitors of (1,3)-β-D-glucan synthase, encoded by FKS genes, causing a depletion of the 1,3-β-glucan in the cell wall. Echinocandin resistance in Candida is associated with mutations in FKS genes and the activation of cell wall stress response mediator pathways, such as Hsp90 and calcineurin (Ca2+), increasing the chitin content. (C) Azoles target and inhibit the enzyme lanosterol 14α-demethylase, encoded by the ERG11 gene, leading to the accumulation of toxic sterol. Azole resistance involves: (i) point mutations in the ERG11 gene, which can be responsible for its overexpression and/or the inhibition of enzyme lanosterol 14α-demethylase, due to the decrease in azole–target binding affinity; (ii) mutations in ERG genes involved in the ergosterol biosynthesis pathway, particularly in ERG3; and (iii) increased efflux of the azole drugs from the fungal cell through the overexpression of multidrug efflux pumps. Red T-shaped bars indicate inhibition. Star icon indicates gene mutation.
Figure 2. Mechanism of action of antifungals against Candida spp. and mechanisms underlying drug resistance. (A) Polyenes act by forming polyene/ergosterol aggregates, destabilizing the fungal membrane. The action of polyenes can be overcome through mutations in ergosterol biosynthesis genes responsible for altered sterol composition and by the activation of stress response pathways, such as catalase and Hsp. (B) Echinocandins act as noncompetitive inhibitors of (1,3)-β-D-glucan synthase, encoded by FKS genes, causing a depletion of the 1,3-β-glucan in the cell wall. Echinocandin resistance in Candida is associated with mutations in FKS genes and the activation of cell wall stress response mediator pathways, such as Hsp90 and calcineurin (Ca2+), increasing the chitin content. (C) Azoles target and inhibit the enzyme lanosterol 14α-demethylase, encoded by the ERG11 gene, leading to the accumulation of toxic sterol. Azole resistance involves: (i) point mutations in the ERG11 gene, which can be responsible for its overexpression and/or the inhibition of enzyme lanosterol 14α-demethylase, due to the decrease in azole–target binding affinity; (ii) mutations in ERG genes involved in the ergosterol biosynthesis pathway, particularly in ERG3; and (iii) increased efflux of the azole drugs from the fungal cell through the overexpression of multidrug efflux pumps. Red T-shaped bars indicate inhibition. Star icon indicates gene mutation.
Jof 09 00080 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Branco, J.; Miranda, I.M.; Rodrigues, A.G. Candida parapsilosis Virulence and Antifungal Resistance Mechanisms: A Comprehensive Review of Key Determinants. J. Fungi 2023, 9, 80. https://doi.org/10.3390/jof9010080

AMA Style

Branco J, Miranda IM, Rodrigues AG. Candida parapsilosis Virulence and Antifungal Resistance Mechanisms: A Comprehensive Review of Key Determinants. Journal of Fungi. 2023; 9(1):80. https://doi.org/10.3390/jof9010080

Chicago/Turabian Style

Branco, Joana, Isabel M. Miranda, and Acácio G. Rodrigues. 2023. "Candida parapsilosis Virulence and Antifungal Resistance Mechanisms: A Comprehensive Review of Key Determinants" Journal of Fungi 9, no. 1: 80. https://doi.org/10.3390/jof9010080

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

Article Metrics

Back to TopTop