28
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Filamentation in Candida auris, an emerging fungal pathogen of humans: passage through the mammalian body induces a heritable phenotypic switch

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Morphological plasticity has historically been an indicator of increased virulence among fungal pathogens, allowing rapid adaptation to changing environments. Candida auris has been identified as an emerging multidrug-resistant human pathogen of global importance. Since the discovery of this species, it has been thought that C. auris is incapable of filamentous growth. Here, we report the discovery of filamentation and three distinct cell types in C. auris: typical yeast, filamentation-competent (FC) yeast, and filamentous cells. These cell types form a novel phenotypic switching system that contains a heritable (typical yeast-filament) and a nonheritable (FC-filament) switch. Intriguingly, the heritable switch between the typical yeast and the FC/filamentous phenotype is triggered by passage through a mammalian body, whereas the switch between the FC and filamentous phenotype is nonheritable and temperature-dependent. Low temperatures favor the filamentous phenotype, whereas high temperatures promote the FC yeast phenotype. Systemic in vivo and in vitro investigations were used to characterize phenotype-specific variations in global gene expression, secreted aspartyl proteinase (SAP) activity, and changes in virulence, indicating potential for niche-specific adaptations. Taken together, our study not only sheds light on the pathogenesis and biology of C. auris but also provides a novel example of morphological and epigenetic switching in fungi.

          Related collections

          Most cited references26

          • Record: found
          • Abstract: found
          • Article: not found

          Nonfilamentous C. albicans mutants are avirulent.

          Candida albicans and Saccharomyces cerevisiae switch from a yeast to a filamentous form. In Saccharomyces, this switch is controlled by two regulatory proteins, Ste12p and Phd1p. Single-mutant strains, ste12/ste12 or phd1/phd1, are partially defective, whereas the ste12/ste12 phd1/phd1 double mutant is completely defective in filamentous growth and is noninvasive. The equivalent cph1/cph1 efg1/efg1 double mutant in Candida (Cph1p is the Ste12p homolog and Efg1p is the Phd1p homolog) is also defective in filamentous growth, unable to form hyphae or pseudohyphae in response to many stimuli, including serum or macrophages. This Candida cph1/cph1 efg1/efg1 double mutant, locked in the yeast form, is avirulent in a mouse model.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Isogenic strain construction and gene mapping in Candida albicans.

            Genetic manipulation of Candida albicans is constrained by its diploid genome and asexual life cycle. Recessive mutations are not expressed when heterozygous and undesired mutations introduced in the course of random mutagenesis cannot be removed by genetic back-crossing. To circumvent these problems, we developed a genotypic screen that permitted identification of a heterozygous recessive mutation at the URA3 locus. The mutation was introduced by targeted mutagenesis, homologous integration of transforming DNA, to avoid introduction of extraneous mutations. The ura3 mutation was rendered homozygous by a second round of transformation resulting in a Ura- strain otherwise isogenic with the parental clinical isolate. Subsequent mutation of the Ura- strain was achieved by targeted mutagenesis using the URA3 gene as a selectable marker. URA3 selection was used repeatedly for the sequential introduction of mutations by flanking the URA3 gene with direct repeats of the Salmonella typhimurium hisG gene. Spontaneous intrachromosomal recombination between the flanking repeats excised the URA3 gene restoring a Ura- phenotype. These Ura- segregants were selected on 5-fluoroorotic acid-containing medium and used in the next round of mutagenesis. To permit the physical mapping of disrupted genes, the 18-bp recognition sequence of the endonuclease I-SceI was incorporated into the hisG repeats. Site-specific cleavage of the chromosome with I-SceI revealed the position of the integrated sequences.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Candida auris: A rapidly emerging cause of hospital-acquired multidrug-resistant fungal infections globally

              Candidiasis, which includes both superficial infections and invasive disease, is the most common cause of fungal infection worldwide. Candida bloodstream infections (BSI) cause significant mortality and elicit a major threat to intensive care unit (ICU) patients [1]. The annual global burden of Candida spp. BSIs is about 400,000 cases, with most cases reported from the developed world. Although Candida albicans remains the most frequently isolated Candida species in the clinical setting, in some countries, a marked shift towards species of Candida that have increased resistance to azoles such as fluconazole (FLU), the standard antifungal drug of choice in many countries, and to the recently introduced antifungals known as echinocandins, is reported. Several species of non-albicans Candida, such as C. tropicalis, C. glabrata, and C. parapsilosis, are well-recognized pathogens in BSIs in different geographic locations. More recently, Candida auris, a multidrug-resistant (MDR) yeast that exhibits resistance to FLU and markedly variable susceptibility to other azoles, amphotericin B (AMB), and echinocandins, has globally emerged as a nosocomial pathogen (Fig 1) [2–20]. Alarmingly, in a span of only 7 years, this yeast, which is difficult to treat and displays clonal inter- and intra-hospital transmission, has become widespread across several countries, causing a broad range of healthcare-associated invasive infections [4, 5, 10, 12, 21, 22]. 10.1371/journal.ppat.1006290.g001 Fig 1 A global map depicting rapid emergence of multidrug-resistant clinical Candida auris strains in 5 continents. The value in parentheses denotes the year of report of C. auris from the respective country or state. Why is C. auris often misidentified in the routine microbiology laboratory? In 2009, a novel Candida species, C. auris, in the C. haemulonii complex (Metchnikowiaceae), was first described from a patient in Japan after its isolation from the external ear canal [23]. The species exhibits a close phylogenetic relationship to C. haemulonii and is differentiated based on sequence analysis of the D1/D2 domain of the large ribosomal subunit (LSU) of 26S rRNA gene and the internal transcribed spacer (ITS) regions of the nuclear rRNA gene operon [23]. The first 3 cases of nosocomial fungemia due to C. auris reported in 2011 from South Korea highlighted the fact that this yeast is commonly misidentified as C. haemulonii and Rhodotorula glutinis by the commercial identification systems VITEK (BioMérieux, Marcy l’Etoile, France) and API-20C AUX (BioMérieux), respectively [3]. These systems involve precast panels of assimilation/growth tests using sets of carbon and nitrogen compounds and are still widely used for routine identification of yeasts. A comprehensive study from India investigated C. auris prevalence among 102 clinical isolates previously identified as C. haemulonii or C. famata with the VITEK system and found that 88.2% of the isolates were C. auris, as confirmed by ITS sequencing [9]. It is evident from several studies published recently that C. auris in routine microbiology laboratories remains an unnoticed pathogen, as 90% of the isolates characterized by commercial biochemical identification systems are misidentified primarily because of a lack of the yeast in their databases [3–9, 12, 16–19, 24, 25]. Different biochemical systems are used in microbiology laboratories, and the majority of them listed in Table 1 misidentify C. auris. A recent study on validating the identification of C. auris with 4 biochemical identification platforms found that all C. auris isolates were misidentified as R. glutinis by API-20C AUX, as C. haemulonii (except 1, as C. catenulata) by Phoenix (BD-Diagnostics, Sparks, MD), as C. haemulonii by VITEK, and as C. famata, C. lusitaniae, C. guilliermondii, or C. parapsilosis by MicroScan (Beckman Coulter, Pasadena, CA) [25] (Table 1). However, Matrix-assisted laser desorption ionization–time of flight mass spectrometry (MALDI-TOF MS) is considered a more rapid and robust diagnostic technique for C. auris identification [9, 10, 13, 16]. Currently, the MALDI-TOF MS approach is commercialized by mainly 2 manufacturers, namely MALDI Biotyper (Bruker-Daltonics, Bremen, Germany) and VITEK MS (BioMérieux). The MALDI Biotyper (Bruker-Daltonics) has a database library that contains spectra of 3 strains of C. auris: 2 from Korea and 1 from Japan. Although both the Bruker-Biotyper and VITEK-MS MALDI-TOF systems lack C. auris entries in the FDA-approved libraries, the research-use-only libraries contain the C. auris database in both MALDI-TOF MS systems [25]. Due to the fact that this yeast is MDR, it is important to identify these species correctly in order to provide optimal patient care. 10.1371/journal.ppat.1006290.t001 Table 1 Worldwide reports of Candida auris along with their misidentification using commercial systems and frequency of antifungal resistance. Country Number of Candida auris isolates Sample (number) Biochemical misidentification (System) Molecular/MALDI-TOF MS identification Number of isolates Year of publication [References] FLU(≥32 μg/ml) ITC(≥1 μg/ml) VRC(≥2 μg/ml) Echinocandins(≥8 μg/ml) AMB(>1 μg/ml) Japan 1 Ear discharge ND ITS, D1D2 ND ND ND ND ND 2009 [23] Korea 15 Ear discharge ND ITS, D1D2 8 8 2 none 9 2009 [2] South Korea 6 Blood C. haemulonii (VITEK), Rhodotorula glutinis (API20C-AUX) ITS, D1D2 4 2 none none 3 2011 [3] India 12 Blood C. haemulonii, C. famata (VITEK); C. sake (API20C-AUX) ITS, D1D2 10 none none none none 2013 [4] 15 Blood (7), CVC tip (3), Excised tissue (3), BAL (1), pus (1) C. haemulonii (VITEK) ITS 15 none 7 none none 2014 [5] 4 Blood (1), urine (1), pericardial fluid (1), BAL (1) C. haemulonii (VITEK); C. sake (API20C-AUX) ITS, D1D2 4 none none none none 2014 [6] 102 Blood (78), tissue (4), pleural fluid (6), peritoneal fluid (7), urine (4), sputum (3) C. haemulonii/C. famata (VITEK) ITS, MALDI-TOF MS 80 none 32 none 14 2015 [9] 51 Blood Not mentioned ITS, D1D2 49 3 9 none 10 2017 [19] India, South Africa, Korea, Japan, Brazil 104: 90 India (I), 6 South Africa (SA), 5 Brazil (B), 2 Korea (K), 1 Japan (J) Blood (n = 89; 78 I, 6 SA, 5 B), peritoneal and pleural fluid (5), invasive infections (4), urine (1), sputum (2) C. haemulonii (VITEK) ITS, D1D2, MALDI-TOF MS 5 (SA); 5 (B); 1 (K); none (J) None (SA); none (B); 1 (K); none (J) 1 (SA); 5 (B); 1 (K); none (J) none (SA); none (B); none (K); none (J) none (SA); 3 (B); none (K); none (J) 2016 [10] a Kuwait 1 Blood C. haemulonii (VITEK) ITS, D1D2 1 ND none none none 2015 [8] Israel 6 Blood (5), urine (1) C. haemulonii (VITEK) ITS, D1D2 6 none none none 6 2017 [18] Spain 8 Blood (4), catheter tip (4) Saccharomyces cerevisiae (AuxaColor 2); C. sake (API20C-AUX); C. lusitaniae, C. haemulonii (VITEK) ITS 8 none 8 none none 2017 [17] UK 12 Blood, sputum, CSF, pleural fluid, arterial line, pustule swab, wound swab, femoral line ITS, D1D2, MALDI-TOF MS 5 ND 1 none none 2016 [11] 50 Blood (16), wound (3), urinary catheter (1), unknown site with invasive candidiasis (2), colonization (28) b MALDI-TOF MS 50 ND ND none Range 0.5–2 μg/ml 2016 [13] Kenya 21 Blood C. haemulonii (VITEK) ITS - - - - - 2014 [24] South Africa 4 Blood C. haemulonii (VITEK) and R. glutinis (API20C-AUX) ITS, D1D2 4 none 1 none none 2014 [7] US 7 Blood (5), urine (1), external ear canal (1) Whole genome sequencing 5 c ND ND 1 c 1 c 2016 [14] CDC Collaborative Project [Pakistan (n = 18), India (n = 19), South Africa, (n = 10), Venezuela (n = 5), Japan (n = 1)] 54 Blood (27), urine (10), soft tissue (5), other sites (12) D1D2, Whole genome sequencing 50 Range 0.125–2 μg/ml 29 4 19 2017 [15] US 10 NA R. glutinis (API20C-AUX); C. haemulonii, C. catenulata (BD Phoenix); C. haemulonii (VITEK); C. famata, C. lusitaniae, C. guilliermondii, or C. parapsilosis (MicroScan) ITS, D1D2, MALDI-TOF MS ND ND ND ND ND 2017 [25] US, tested strains from Germany (n = 2), India (n = 11), Korea (n = 2), Japan (n = 1) 16 Blood (15), ear (1) Unidentified (API20C-AUX) ITS 8 5 5 d none 12 e , 16 d 2017 [20] Venezuela 18 Blood C. haemulonii (VITEK) ITS 18 ND 18 none Range 1–2 μg/ml 2016 [12] Colombia 17 Blood (13) peritoneal fluid (1), CSF (1), bone (1), urine (1) C. haemulonii (VITEK, Phoenix); C. tropicalis (MicroScan Walkaway); C. famata (API Candida); C. albicans (MicroScanautoSCAN); C. tropicalis (MicroScan Walkaway)/ C. famata (API Candida); C. albicans (MicroScanAutoSCAN) MALDI-TOF MS 10 ND 4 none 11 2017 [16] Abbreviations: -, not clear in the abstract; AMB, amphotericin B; BAL, bronchoalveolar lavage; CDC, US Centers for Disease Control and Prevention; CSF, cerebral spinal fluid; CVC tip, central venous catheter tip; FLU, fluconazole; ITC, itraconazole; ITS, internal transcribed spacer; MALDI-TOF MS, Matrix- assisted laser desorption ionization–time of flight mass spectrometry; MIC, minimum inhibitory concentration; ND, not done; VRC, voriconazole. aAntifungal susceptibility testing data of Indian isolates is same as reported by Kathuria et al., 2015. b Colonization with C. auris was defined as culture-positive skin, oropharynx, vascular line exit site, respiratory, and urinary tract without clinical signs of Candida infection. cMIC value not given. dMICs read after 48 hours. eMICs read after 24 hours. Does genetic predisposition make C. auris virulent? A recently published draft genome of C. auris shows that it has a genome size of approximately 12.3 Mb [26, 27]. A significant percentage of genes in C. auris are devoted to central metabolism, a property that is common to pathogenic Candida and crucial for adaptation to divergent environments. In addition, C. auris shares numerous virulence attributes with C. albicans, including genes and pathways involved in cell wall modelling and nutrient acquisition, histidine kinase-2 component systems, iron acquisition, tissue invasion, enzyme secretion, and multidrug efflux [21, 26, 27]. However, in vitro results in a single study that tested the production of phospholipase and secreted proteinase in multiple isolates of C. auris from different geographical regions showed that both secreted proteinase and phospholipase production was strain dependent. The phospholipase activity and secreted proteinase were detected in 37.5% and 64% of the tested isolates, respectively [20]. In general, the tested C. auris strains tended to have weak phospholipase activity, with the majority of isolates being non-phospholipase producers [20]. Furthermore, a significant portion of the C. auris genome encodes the ATP-binding cassette (ABC) and major facilitator superfamily (MFS) transporter families along with drug transporters that may explain the exceptional multidrug resistance in this pathogen [21, 27]. ABC-type efflux activity by Rhodamine 6G transport was significantly greater among C. auris than C. glabrata isolates, suggesting the intrinsic resistance of C. auris to azoles [18]. Interestingly, comparison of whole genome sequencing (WGS) data shows C. auris to be a close phylogenetic relative of C. lusitaniae, a species recognized for intrinsic antifungal resistance [21, 27]. C. auris also demonstrates thermotolerance, growing optimally at 37°C and maintaining viability at up to 42°C, salt tolerance, and cell aggregation into large, difficult-to-disperse clusters, which may help some strains to persist in the hospital environment [11, 23]. In a Galleria mellonella model, the aggregate-forming isolates exhibit significantly less pathogenicity than their non-aggregating counterparts [11]. Importantly, the non-aggregating isolates exhibited pathogenicity comparable to that of C. albicans, which is the most pathogenic member of the genus [11]. However, it is important to mention here that the observations made in this study are yet to be correlated with clinical cases and thus, assuming the same results in patients, need further experimentation. Furthermore, the virulence of C. auris tested in a mouse model of hematogenous-disseminated candidiasis showed distinct yeast cell aggregates in the kidneys of mice, with lethal C. auris infection suggesting that aggregation might be a mode of immune evasion and persistence in tissue [18]. Another significant factor involved in C. auris virulence is its ability to differentially adhere to polymeric surfaces, form biofilms, and resist antifungal agents that are active against its planktonic counterparts [28]. However, a more recent study reported that C. auris biofilms were significantly thinner, i.e., exhibited 50% thickness compared to C. albicans biofilm [20]. Also, C. auris exhibits minimal ability to adhere to silicone elastomer (a representative catheter material) relative to C. albicans [20]. C. auris’s weak adherence ability suggests that it is likely to play some role in catheter-associated candidiasis but not a large one, in contrast to C. albicans and C. parapsilosis, which are known to cause such infections [20]. Although, C. auris expresses several virulence factors, albeit to a lesser extent than C. albicans and in a strain-dependent manner [20]. The past and present of C. auris: Is the emergence of C. auris a menace to public health? In 2009, 15 isolates of C. auris were recovered from the ear canals of patients suffering from chronic otitis media in South Korea [2]. Most of these isolates showed a reduced susceptibility to AMB and azole antifungals. This report was followed by the first 3 cases of nosocomial fungemia caused by C. auris from South Korea [3]. The latter study reported that the earliest isolate of C. auris was found in 1996 in the Korean isolate collection [3]. All 3 patients had persistent fungemia for 10 to 31 days, and 2 patients who received FLU therapy followed by AMB showed therapeutic failure and had fatal outcome. Subsequently, 2 larger series of candidemia and deep-seated infections from India in 2013 and 2014 clearly showed that clonal strains of MDR C. auris had emerged in 3 hospitals [4, 5]. The isolates were resistant to FLU and 5-flucytosine (FC) and had elevated minimum inhibitory concentrations (MICs) of voriconazole (VRC) and caspofungin (CFG) [4, 5]. The most worrisome findings were persistent candidemia and high attributable mortality rates [4, 5]. C. auris accounted for >5% of candidemia in a national ICUs survey and up to 30% of candidemia at individual hospitals in India [4, 19]. In the subsequent 2 years, several reports of hospital-associated infections emerged from South Africa, United Kingdom, Venezuela, Colombia, United States, Pakistan, Israel, Kenya, and Spain [7, 11–18, 24]. Table 1 lists several countries reporting C. auris infection published so far across 5 continents. A collaborative project undertaken by the US Centers for Disease Control and Prevention (CDC) to understand the global emergence and epidemiology of C. auris reported that isolates from 54 patients with C. auris infection from Pakistan, India, South Africa, and Venezuela showed that 93% of isolates were resistant to FLU, 35% to AMB, and 7% to echinocandins; 41% were resistant to 2 antifungal classes, and 4% were resistant to 3 classes [15]. The fact that this yeast exhibits MDR clonal strains that are nosocomially transmitted is unusual in other Candida species [3, 5, 21]. Therefore, the possible threat of rapid spread in affected countries and its emergence in unaffected countries will not only challenge clinicians for its effective therapeutic management but will also bring high economic burden, especially to countries in resource-limited settings where modern identification facilities and access to antifungals other than FLU are limited. What are the drivers of clonal transmission and nosocomial outbreaks of C. auris? There is increasing evidence that suggests likely transmission of C. auris in healthcare settings. Recent reports highlight the persistent colonization by C. auris of hospital environments and multiple body-sites of patients, leading to high transmissibility and protracted outbreaks [13, 14]. A large outbreak of 50 C. auris cases in a London cardio-thoracic center between April 2015 and July 2016 showed persistent presence of the yeast around bed-space areas [13]. Genotyping with amplified fragment length polymorphism (AFLP) demonstrated that C. auris isolates clustered. Similarly, the investigation of the first 7 cases of C. auris infection identified in the US, which occurred between May 2013 and August 2016, showed colonization with C. auris on skin and other body sites weeks to months after their initial infection, which could possibly lead to contamination of the healthcare environment and pose a risk of continuous transmission [14]. Furthermore, C. auris was isolated from samples taken from the mattress, bedside table, bed rail, chair, and windowsill [14]. WGS results demonstrate that isolates from patients admitted to the same hospital in New Jersey were nearly identical, as were isolates from patients admitted to the same Illinois hospital [14]. Also, in the London outbreak, a healthcare worker caring for a heavily C. auris–colonized patient had a C. auris–positive nose swab [13]. Effective implementation of strict infection-prevention control measures are required to prevent transmission of C. auris. These include isolation of patients and their contacts, wearing of personal protective clothing by healthcare workers, screening of patients on affected wards, skin decontamination with chlorhexidine, environmental cleaning with chlorine-based reagents, and terminal decontamination with hydrogen peroxide vapor or ultraviolet (UV) light [13, 29]. Enhanced terminal cleaning with UV light has recently been shown to reduce infections with many nosocomial pathogens and might also be of use for preventing C. auris transmission [30]. Previously, several geographically related clusters have been reported from South Korea [2, 3], India [4, 5, 10], South Africa [10], Pakistan [15], and hospitals in Latin America [12, 16]. Clonality within C. auris has been shown using AFLP, multilocus sequence typing, and MALDI-TOF MS among strains in India, South Africa, and Brazil [10]. A recent study applying WGS demonstrated highly related C. auris isolates in 4 unrelated and geographically separated Indian hospitals, suggesting that this pathogen exhibits a low diversity [21]. A large-scale application of WGS analysis suggests recent independent and nearly simultaneous emergence of different clonal populations on 3 continents, demonstrating highly related C. auris isolates in the same geographic areas [15]. So far, no reservoir of C. auris has been identified, although future studies on its isolation from animals, plants, and water sources are warranted. Is antifungal resistance in C. auris a therapeutic challenge? Patients with C. auris infections have risk factors similar to those of other Candida spp. infections, including abdominal surgery (25%–77%), broad-spectrum antibiotics (25%–100%), ICU admission (58%), diabetes mellitus (18%), presence of central venous catheters (25%–94%), and malignancies (11%–43%) [3–5, 7, 12, 14–16]. The overall crude in-hospital mortality rate of C. auris candidemia ranges from 30% to 60%, and infections typically occur several weeks (10‒50 days) after admission [4, 5, 10, 12, 13]. C. auris invasive infections represent a therapeutic challenge, and no consensus exists for optimal treatment. A few studies report breakthrough fungemia while on FLU, and this correlates with commonly reported high MICs (>32 μg/ml), suggesting intrinsic resistance against this drug [3–5]. Although epidemiological cutoff values (ECVs) or clinical breakpoints are not yet defined for C. auris, newer azoles such as posaconazole (range, 0.06–1 μg/ml) and isavuconazole (range, 2 μg/ml) MICs [9, 15]. Up till now, echinocandin resistance is noted in fewer isolates (2%–8%) [9, 14, 15], but almost half of isolates are MDR (resistant to ≥2 antifungal classes), and a low number (4%) exhibit resistance to all classes of antifungals [2, 9, 12, 15, 16, 19]. Echinocandins remain the first-line therapy for C. auris infections, provided that specific susceptibility testing is undertaken at the earliest opportunity. Although CFG is normally highly effective against Candida biofilms, a recent report demonstrated that CFG was predominately inactive against C. auris biofilms [29]. FC (MIC50, 0.125–1 μg/ml) is a treatment option in renal tract or urinary tract infections, as the echinocandins fail to achieve therapeutic concentrations in urine [4, 5, 7, 9, 11–13, 15, 18]. Also, a novel drug, SCY-078, which is the first orally bioavailable 1, 3-β-D-glucan synthesis inhibitor, has been shown to possess potent activity against various Candida spp. and exhibit potent antifungal activity against C. auris isolates [20]. Furthermore, SCY-078 showed growth-inhibition and anti-biofilm activity and could be an important antifungal to treat this MDR species [20]. At present, the mechanism of antifungal resistance in C. auris is unclear. The recently published draft genome of C. auris revealed the presence of single copies of ERG3, ERG11, FKS1, FKS2, and FKS3 genes [21]. Detection of azole-resistant mutations by comparing ERG11 amino acid sequences between C. albicans and C. auris showed that alterations at azole-resistance codons in C. albicans were present in C. auris isolates [15]. These substitutions were strongly associated with country-wise–specific geographic clades [15]. Resistance is probably inducible under antifungal pressure, resulting in rapid mutational changes. However, future studies with emphasis on several molecular mechanisms, including efflux and transporters, could provide insight on C. auris resistance. What are the important things that we still need to learn about C. auris? We are just beginning to know the epidemiology and behavior of C. auris, but at the present, far more gaps exist in our knowledge. The earliest findings of C. auris are from 1996. The pertinent question remains whether this pathogen existed far earlier than 1996, and we were just unable to identify it. The latter is less plausible because many centers have reviewed archived isolate collections that have not shown any isolates of C. auris before 1996. We also do not know why C. auris is independently, almost simultaneously, emerging in so many places worldwide. It has been shown that there is a profound phylo-geographic structure with large genetic differences among geographic clades and high clonality within the geographic clades. However, a common characteristic is the high level of antifungal resistance, which is rare in other Candida spp. C. auris is the only species in which several isolates have been identified with resistance to all 4 classes of human antifungal drugs. It seems reasonable to opine that changes or misuse of antifungal drugs is one of the factors, although no specific risk factors for acquiring C. auris seem to exist. What we do know is that environmental factors probably play a role in outbreaks in healthcare settings that include prolonged survival in healthcare environments, probably due to skin colonization of patients and asymptomatic carriers. It is obvious that future research is warranted on multiple aspects of C. auris, which seems to have the typical characteristics of well-known, healthcare-associated pathogens such as carbapenemase-producing gram-negatives, Clostridium difficile, vancomycin-resistant Enterococcus (VRE), and methicillin-resistant Staphylococcus aureus (MRSA). Given the behavior of the latter 4, a further spread of C. auris in healthcare settings on a worldwide scale is expected. C. auris worldwide emergence has prompted the CDC, (http://www.cdc.gov/fungal/diseases/candidiasis/candida-auris-alert.html [last accessed February 2017]), Public Health England (PHE), London (https://www.gov.uk/government/uploads/system/uploads/attachment_data/file/534174/Guidance_Candida__auris.pdf [last accessed February 2017]), and the European Centre for Disease Prevention and Control (ECDC), Europe (http://ecdc.europa.eu/en/publications/Publications/Candida-in-healthcare-settings_19-Dec-2016.pdf) to issue health alerts for strict vigilance of C. auris cases. International collaborative consortia and timely efforts by the medical community are indispensable in controlling this super bug before it adapts in our healthcare facilities. Furthermore, more intensive efforts are required, and one such crucial step is the support from funding agencies to initiate multidisciplinary research to better understand its ecology, evolution, and resistance mechanisms, which will go a long way for its treatment and prevention.
                Bookmark

                Author and article information

                Contributors
                huanggh@im.ac.cn
                Journal
                Emerg Microbes Infect
                Emerg Microbes Infect
                Emerging Microbes & Infections
                Nature Publishing Group UK (London )
                2222-1751
                28 November 2018
                28 November 2018
                2018
                : 7
                : 188
                Affiliations
                [1 ]ISNI 0000000119573309, GRID grid.9227.e, State Key Laboratory of Mycology, Institute of Microbiology, , Chinese Academy of Sciences, ; Beijing, 100101 China
                [2 ]ISNI 0000 0004 1797 8419, GRID grid.410726.6, University of Chinese Academy of Sciences, ; Beijing, 100049 China
                [3 ]ISNI 0000 0001 0125 2443, GRID grid.8547.e, State Key Laboratory of Genetic Engineering, , School of Life Sciences, Fudan University, ; Shanghai, 200438 China
                [4 ]ISNI 0000 0001 0125 2443, GRID grid.8547.e, Institutes of Biomedical Sciences, , Fudan University, ; Shanghai, 200032 China
                [5 ]ISNI 0000 0004 0632 4559, GRID grid.411634.5, Department of Clinical Laboratory, , Peking University People’s Hospital, ; Beijing, 100044 China
                Article
                187
                10.1038/s41426-018-0187-x
                6258701
                30482894
                25dc32b2-64da-4a48-96ca-91604320d2a5
                © The Author(s) 2018

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 23 July 2018
                : 22 October 2018
                : 25 October 2018
                Funding
                Funded by: FundRef https://doi.org/10.13039/501100001809, National Natural Science Foundation of China (National Science Foundation of China);
                Award ID: 31625002 and 31370175
                Award Recipient :
                Categories
                Article
                Custom metadata
                © The Author(s) 2018

                Comments

                Comment on this article