Next Article in Journal
Complete Utilization of the Major Carbon Sources Present in Sugar Beet Pulp Hydrolysates by the Oleaginous Red Yeasts Rhodotorula toruloides and R. mucilaginosa
Next Article in Special Issue
Emergence of Candida auris in Brazil in a COVID-19 Intensive Care Unit
Previous Article in Journal
Culture-Dependent and Amplicon Sequencing Approaches Reveal Diversity and Distribution of Black Fungi in Antarctic Cryptoendolithic Communities
Previous Article in Special Issue
Comparison of Two Commercially Available qPCR Kits for the Detection of Candida auris
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Candida auris Whole-Genome Sequence Benchmark Dataset for Phylogenomic Pipelines

Mycotic Diseases Branch, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
*
Author to whom correspondence should be addressed.
J. Fungi 2021, 7(3), 214; https://doi.org/10.3390/jof7030214
Submission received: 10 February 2021 / Revised: 4 March 2021 / Accepted: 11 March 2021 / Published: 16 March 2021
(This article belongs to the Special Issue Candida auris 2.0)

Abstract

:
Candida auris is a multidrug-resistant pathogen that represents a serious public health threat due to its rapid global emergence, increasing incidence of healthcare-associated outbreaks, and high rates of antifungal resistance. Whole-genome sequencing and genomic surveillance have the potential to bolster C. auris surveillance networks moving forward. Laboratories conducting genomic surveillance need to be able to compare analyses from various national and international surveillance partners to ensure that results are mutually trusted and understood. Therefore, we established an empirical outbreak benchmark dataset consisting of 23 C. auris genomes to help validate comparisons of genomic analyses and facilitate communication among surveillance networks. Our outbreak benchmark dataset represents a polyclonal phylogeny with three subclades. The genomes in this dataset are from well-vetted studies that are supported by multiple lines of evidence, which demonstrate that the whole-genome sequencing data, phylogenetic tree, and epidemiological data are all in agreement. This C. auris benchmark set allows for standardized comparisons of phylogenomic pipelines, ultimately promoting effective C. auris collaborations.

1. Introduction

The emerging multidrug-resistant pathogenic yeast Candida auris has been reported in over 40 countries and represents a threat to global health [1,2]. Patient-to-patient spread has been documented in multiple countries [3]. Infection prevention guided by rapid detection in healthcare settings is essential because C. auris is easily spread in healthcare settings as it colonizes patients’ skin and can survive and persist in the clinical environment for weeks [4]. C. auris is capable of causing severe bloodstream infections and has become the leading cause of invasive candidiasis in some hospitals [5]. In addition, C. auris is difficult to treat as it is commonly resistant to multiple antifungal drug classes.
Whole-genome sequencing (WGS) based methods have increasingly been used to detect and characterize outbreaks for this emerging pathogen [2,6,7]. Population analysis of C. auris genomic data has revealed that most of the detected cases are stratified into four major clades (Clades I, II, III, and IV) except for a single representative case describing a potential fifth clade (Clade V) [8,9]. Currently, WGS is used as part of the public health response to halt the spread of C. auris by helping to characterize introductions, phylogeographic mixing, and transmission dynamics [2]. The potential for genomic surveillance to serve as a powerful tool to support epidemiological investigation in public health is clear, but genomic analysis methods need to be validated and documented as a prerequisite for public health authorities to trust genomic data in routine investigations [10].
As access to genomic sequence data has grown to help meet the challenge of investigating transmission chains and large-scale pathogen populations, the capacity to analyze the ever-increasing amount of sequence data has struggled to keep up. One common bottleneck of scaling analysis capacity often occurs when onboarding and validating new and constantly changing phylogenomic pipelines and methods to identify variants and distinguish related genomes [11]. Previous studies have utilized single nucleotide polymorphisms (SNPs), short tandem repeat, and multilocus sequence typing (MLST) strategies for molecular typing of C. auris outbreaks [12,13]. Outbreak datasets with publicly available raw reads have been published for multiple outbreak events in North America and globally [6,14]. However, a standardized dataset to allow for comparisons of these molecular typing and phylogenomic pipelines has not been established for C. auris. Therefore, we set out to establish a genomic benchmark dataset to serve as a resource to facilitate global efforts to collaborate and rapidly validate sequence analysis tools.
Here, we present an empirically and epidemiologically validated outbreak dataset of Clade I isolates from North America in a standardized format that enables easy access and automated analysis. To ensure computational feasibility on a wide range of computing infrastructures, we included a small subset of these well-vetted genome sequences in the C. auris benchmark dataset. This resource provides an important first step towards a collaborative infrastructure for academic and public health authorities to document and validate variant calling and resulting phylogenetic tree topologies to aid communication, outbreak genomic surveillance, and containment efforts.

2. Materials and Methods

The C. auris isolates used in this study were from a subset of outbreak cases from the United States where the sequence data and epidemiological data, such as the facility where the patient isolates were collected, are all in agreement as previously described by [2,6]. The isolates included in the study belong to the two Clade I lineages involved in outbreaks (subclade 1b and 1c), the complete reference genome is from subclade 1c, and the subclade designations were previously described (Muñoz et al. 2021). The isolates were obtained from clinical and colonization cases, as defined by The Council of State and Territorial Epidemiologists (CSTE). For quality assurance, sequences with greater than 20× sequencing coverage were selected for the dataset, which is the quality threshold average coverage guideline for the National Center for Biotechnology Information (NCBI) Pathogen Detection. Strain identifiers for each isolate, accession numbers to the genomes, subclade, PubMed study identifiers, and outbreak and facility anonymized codes are listed in Table 1. The sequencing metrics (sequence quality, total reads, assembly length, and contigs) for each genome were obtained from NCBI [15]. Additionally, we used FastQC to estimate the sequence read quality and percent GC content [16]. We identified SNPs using MycoSNP GeneFlow workflows (https://git.biotech.cdc.gov/geneflow-workflows/mycosnp/), which performs read mapping using Burrows-Wheeler Aligner (BWA) v0.7.17, SNP calling using Genome Analysis Toolkit (GATK) v4.1.4.1, and generates a multi-FASTA file of the informative variants. To ensure that multiple phylogenetic analyses produce similar results, maximum parsimony and maximum likelihood phylogenetic analyses with 1000 bootstraps using MEGA version X were generated [17] as previously described [6] and visualized on Microreact [18]. All of sequence data and tree materials are publicly available for download at GitHub: https://github.com/globalmicrobialidentifier-WG3/datasets (accessed on 18 February 2021).

3. Results

Twenty-four isolates from 21 clinical or colonization C. auris cases were included in this dataset. These cases were reported during 2016–2017 United States regional spread where ongoing transmission was previously described for healthcare facilities in New York, New Jersey, and Massachusetts [6]. Specifically, three Clade I outbreaks (Outbreak 1–3) that each form their own separate monophyletic branch and comprise eight healthcare facilities (Facility A–G) are represented in this dataset (Figure 1). Clade I includes isolates representing three major lineages; two lineages are routinely involved in outbreaks and are diverged from a small subclade that includes the commonly used B8441 reference genome (Muñoz et al. 2021). These were previously designated subclade 1a, 1b, and 1c and representatives of each subclade are contained in the dataset (Table 1). From previous descriptions, subclade 1a includes the B8441 reference, subclade 1b contains isolates from cases in India, Pakistan, Kenya, France, Germany, China, United Kingdom, Saudi Arabia, and the United States (California and Connecticut); and subclade 1c contains isolates from cases in India, Pakistan, Saudi Arabia, and the United States (New York, New Jersey, and Massachusetts).
Phylogenetic analysis showed that all 23 genomes clustered with Clade I, with 22 (92%) belonging to subclade 1c, which represents three distinct New York, New Jersey, and Massachusetts outbreak branches (Figure 1). Of the subclade 1c isolates, 13 (59%) clustered with the New York outbreak 1 branch node, 5 (27%) form the New Jersey outbreak 2 branch node, and 3 (14%) with the Massachusetts outbreak 3 branch node (Figure 1). Outbreak 1 describes 13 genomes collected from 11 New York cases originating from three healthcare facilities (Facility A, B, and C) (Table 1). Outbreak 2 describes six genomes collected from five New Jersey cases in two separate healthcare facilities (Facility D and E). Outbreak 3 describes three genomes from three Massachusetts cases from a single facility (Facility F). The mean SNP difference among isolates obtained from outbreak codes 1, 2, and 3 were 16 (range 0–34), 15 (range 3–20), and 1 (range 0–2), respectively.
The single outgroup subclade 1b representative genome from a Connecticut case is clearly separated by >100 SNPs from the subclade 1c lineage genomes. Multiple independent phylogenetic analyses support the observed phylogenetic relationships, and Supplementary Figure S1 shows a maximum likelihood phylogeny with 1000 bootstrap replications produced similar results. The quality metrics for each genome are reported in Table 2. The scripts for downloading and accessing the associated benchmark files are available on GitHub: https://github.com/globalmicrobialidentifier-WG3/datasets (accessed on 18 February 2021).

4. Discussion

The C. auris empirical outbreak benchmark dataset was established to help standardize comparisons of genomic analysis tools designed for the specific purpose of validating molecular typing methods to aid outbreak surveillance. Compared to simulated datasets, manually established empirical benchmark datasets are often slow and tedious to generate, but they are a powerful resource for validating phylogenomic analysis tools (Timme et al. 2019). We included only a subset of the total cases from the three state outbreaks in our analysis; therefore, this dataset is not representative of the current C. auris molecular epidemiology observed in the United States. This resource represents well-vetted sequence data that can be used to compare test results against known results to aid validation efforts.
Limitations to consider for comparing SNP analyses are that the input sequence quality and coverage can generate different SNP numbers, which highlights the importance of adhering to sequence quality levels. Different sequence assembly tools, read mapping tools, and SNP pipelines can generate different SNP counts, but the topology should remain consistent. In addition, the number of SNPs described within an outbreak or healthcare facility is context dependent and may not be directly comparable with other studies due to multiple factors. For example, C. auris can colonize patients for extended periods of time (months to years) and coupled with the length of time for the on-going outbreak [19], the number of clades, number of importation events, and mixing can all expand the breath of genetic variation in an outbreak population.
C. auris is an emerging pathogen and more of these types of benchmark datasets will be needed. Future datasets should expand to include new clades, isolates, and scenarios to better serve the community. Benchmark datasets assembled through empirical approaches from well-vetted studies that are supported by multiple lines of evidence have been traditionally used for validation efforts [11,20]. Simulated datasets can allow for manipulation of multiple parameters and often compliment and help overcome some of the slow manual processes required for generating empirical dataset [21]. These rigorous phylogenomic validation practices and resources will help ensure confidence in utilizing various genomic analysis tools to inform public health decision and action.

Supplementary Materials

The following are available online at https://www.mdpi.com/2309-608X/7/3/214/s1, Figure S1: Phylogeny of Candida auris benchmark dataset including cases from Clade I.

Author Contributions

Conceptualization, R.M.W., M.L. and N.A.C.; methodology, R.M.W., K.F., E.M. and N.A.C.; validation, R.M.W., K.F., E.M. and N.A.C.; formal analysis, R.M.W. and E.M.; investigation, R.M.W., K.F., E.M. and N.A.C.; data curation, R.M.W., K.F., E.M. and N.A.C.; writing—original draft preparation, R.M.W.; writing—review and editing, R.M.W., K.F., M.L. and N.A.C.; visualization, R.M.W. and E.M.; supervision, R.M.W., M.L. and N.A.C.; All authors have read and agreed to the published version of the manuscript.

Funding

Oak Ridge Institute for Science and Education (ORISE) for E.M. research fellowship.

Institutional Review Board Statement

Ethical review and approval were waived for this study, due to fact the genome sequences were previously published in PubMed ID 30293877, which stated, “the work was part of an ongoing public health response, it was determined to be non-research public health practice by CDC officials who are responsible for human participant protection, and this study was therefore not subject to review by institutional review boards”.

Informed Consent Statement

Not applicable.

Data Availability Statement

All of materials for the benchmark dataset are publicly available for download at GitHub: https://github.com/globalmicrobialidentifier-WG3/datasets (accessed on 10 February 2021).

Acknowledgments

Special thanks to Anastasia P. Litvintseva, Lalitha Gade, the staff of the Mycotic Diseases Branch (MDB) at the Centers for Disease Control and Prevention (CDC).

Conflicts of Interest

The authors declare that there is no conflict of interest regarding the publication of this article.

Disclaimer

The findings and the conclusions in this report are those of the authors and do not necessarily represent the views of the Centers for Disease Control and Prevention. Use of trade names is for identification only and does not imply endorsement.

References

  1. Rhodes, J.; Fisher, M.C. Global epidemiology of emerging Candida auris. Curr. Opin. Microbiol. 2019, 52, 84–89. [Google Scholar] [CrossRef] [PubMed]
  2. Chow, N.A.; Muñoz, J.F.; Gade, L.; Berkow, E.L.; Li, X.; Welsh, R.M.; Forsberg, K.; Lockhart, S.R.; Adam, R.; Alanio, A.; et al. Tracing the Evolutionary History and Global Expansion of Candida auris Using Population Genomic Analyses. mBio 2020, 11, e03364-19. [Google Scholar] [CrossRef] [PubMed]
  3. Saris, K.; Meis, J.F.; Voss, A. Candida auris. Curr. Opin. Infect. Dis. 2018, 31, 334–340. [Google Scholar] [CrossRef] [PubMed]
  4. Welsh, R.M.; Bentz, M.L.; Shams, A.; Houston, H.; Lyons, A.; Rose, L.J.; Litvintseva, A.P. Survival, Persistence, and Isolation of the Emerging Multidrug-Resistant Pathogenic Yeast Candida auris on a Plastic Health Care Surface. J. Clin. Microbiol. 2017, 55, 2996–3005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Van Schalkwyk, E.; Mpembe, R.S.; Thomas, J.; Shuping, L.; Ismail, H.; Lowman, W.; Karstaedt, A.S.; Chibabhai, V.; Wadula, J.; Avenant, T.; et al. Epidemiologic Shift in Candidemia Driven by Candida auris, South Africa, 2016-2017(1). Emerg. Infect. Dis. 2019, 25, 1698–1707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Chow, N.A.; Gade, L.; Tsay, S.V.; Forsberg, K.; Greenko, J.A.; Southwick, K.L.; Barrett, P.M.; Kerins, J.L.; Lockhart, S.R.; Chiller, T.M.; et al. Multiple introductions and subsequent transmission of multidrug-resistant Candida auris in the USA: A molecular epidemiological survey. Lancet Infect. Dis. 2018, 18, 1377–1384. [Google Scholar] [CrossRef]
  7. Rhodes, J.; Abdolrasouli, A.; Farrer, R.A.; Cuomo, C.A.; Aanensen, D.M.; Armstrong-James, D.; Fisher, M.C.; Schelenz, S. Author Correction: Genomic epidemiology of the UK outbreak of the emerging human fungal pathogen Candida auris. Emerg. Microbes Infect. 2018, 7, 1–2. [Google Scholar] [CrossRef] [PubMed]
  8. Muñoz, J.F.; Gade, L.; Chow, N.A.; Loparev, V.N.; Juieng, P.; Berkow, E.L.; Farrer, R.A.; Litvintseva, A.P.; Cuomo, C.A. Genomic insights into multidrug-resistance, mating and virulence in Candida auris and related emerging species. Nat. Commun. 2018, 9, 5346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Chow, N.A.; De Groot, T.; Badali, H.; Abastabar, M.; Chiller, T.M.; Meis, J.F. Potential Fifth Clade of Candida auris, Iran, 2018. Emerg. Infect. Dis. 2019, 25, 1780–1781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Timme, R.E.; Strain, E.; Baugher, J.D.; Davis, S.; Gonzalez-Escalona, N.; Leon, M.S.; Allard, M.W.; Brown, E.W.; Tallent, S.; Rand, H. Phylogenomic Pipeline Validation for Foodborne Pathogen Disease Surveillance. J. Clin. Microbiol. 2019, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Timme, R.E.; Crandall, K.; Shumway, M.; Trees, E.K.; Simmons, M.; Agarwala, R.; Davis, S.; Tillman, G.E.; Defibaugh-Chavez, S.; Carleton, H.A.; et al. Benchmark datasets for phylogenomic pipeline validation, applications for foodborne pathogen surveillance. PeerJ 2017, 5, e3893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. De Groot, T.; Puts, Y.; Berrio, I.; Chowdhary, A.; Meis, J.F. Development of Candida auris Short Tandem Repeat Typing and Its Application to a Global Collection of Isolates. mBio 2020, 11, e02971-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kwon, Y.J.; Shin, J.H.; Byun, S.A.; Choi, M.J.; Won, E.J.; Lee, D.; Lee, S.Y.; Chun, S.; Lee, J.H.; Choi, H.J.; et al. Candida auris Clinical Isolates from South Korea: Identification, Antifungal Susceptibility, and Genotyping. J. Clin. Microbiol. 2019, 57, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Mohsin, J.; Hagen, F.; Al-Balushi, Z.A.M.; De Hoog, G.S.; Chowdhary, A.; Meis, J.F.; Al-Hatmi, A.M.S. The first cases of Candida auris candidaemia in Oman. Mycoses 2017, 60, 569–575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. NCBI Pathogen Detection System. 2017. Available online: https://www.ncbi.nlm.nih.gov/pathogens/ (accessed on 18 February 2021).
  16. Ldquo; FastQC. Available online: http://www.bioinformatics.babraham.ac.uk/projects/fastqc/ (accessed on 18 February 2021).
  17. Kumar, S.; Stecher, G.; Li, M.; Knyaz, C.; Tamura, K. MEGA X: Molecular evolutionary genetics analysis across computing platforms. Mol. Biol. Evol. 2018, 35, 1547–1549. [Google Scholar] [CrossRef] [PubMed]
  18. Argimón, S.; AbuDahab, K.; Goater, R.J.E.; Fedosejev, A.; Bhai, J.; Glasner, C.; Feil, E.J.; Holden, M.T.G.; Yeats, C.A.; Grundmann, H.; et al. Microreact: Visualizing and sharing data for genomic epidemiology and phylogeography. Microb. Genom. 2016, 2, e000093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Bergeron, G.; Bloch, D.; Murray, K.; Kratz, M.; Parton, H.; Ackelsberg, J.; Antwi, M.; Del Rosso, P.; Dorsinville, M.; Kubinson, H.; et al. Candida auris Colonization After Discharge to a Community Setting: New York City, 2017–2019. Open Forum Infect. Dis. 2021, 8, ofaa620. [Google Scholar] [CrossRef] [PubMed]
  20. Taylor, T.L.; Volkening, J.D.; DeJesus, E.; Simmons, M.; Dimitrov, K.M.; Tillman, G.E.; Suarez, D.L.; Afonso, C.L. Rapid, multiplexed, whole genome and plasmid sequencing of foodborne pathogens using long-read nanopore technology. Sci. Rep. 2019, 9, 16350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. McTavish, E.J.; Pettengill, J.; Davis, S.; Rand, H.; Strain, E.; Allard, M.; Timme, R.E. TreeToReads—A pipeline for simulating raw reads from phylogenies. BMC Bioinform. 2017, 18, 178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Phylogeny of Candida auris benchmark dataset including cases from Clade I. Numbers above the branch indicate the number of changes that have occurred in that branch length, and the internal solid circle nodes indicate separations with a bootstrap value of at least 90%. Each leaf node represents a unique isolate; the shape of the node refers to the outbreak number code and the node color refers to the facility.
Figure 1. Phylogeny of Candida auris benchmark dataset including cases from Clade I. Numbers above the branch indicate the number of changes that have occurred in that branch length, and the internal solid circle nodes indicate separations with a bootstrap value of at least 90%. Each leaf node represents a unique isolate; the shape of the node refers to the outbreak number code and the node color refers to the facility.
Jof 07 00214 g001
Table 1. List of sequences included in the Candida auris whole-genome sequence benchmark dataset.
Table 1. List of sequences included in the Candida auris whole-genome sequence benchmark dataset.
BioSampleIsolate IDSRA RunOutbreak IDFacility IDSubcladeDate CollectedStudy *
SAMN10139509B12352SRR79092821BIc201630293877
SAMN10139317B12681SRR79092141BIc201730293877
SAMN10139320B12677SRR79091351BIc201730293877
SAMN10139376B12490SRR79092101BIc201630293877
SAMN10139493B12427SRR79091841BIc201630293877
SAMN10139490B12430SRR79092331BIc201630293877
SAMN10139465B12461SRR79092341AIc201630293877
SAMN10139461B12467SRR79092971AIc201630293877
SAMN10139268B12824SRR79093131CIc201730293877
SAMN10139267B12825SRR79093851CIc201730293877
SAMN10139529B12044SRR79091471CIc201630293877
SAMN10139470B12456SRR79091561CIc201630293877
SAMN10139462B12466SRR79094131CIc201630293877
SAMN10139189B13613SRR79092843FIc201730293877
SAMN10139194B13520SRR79093943FIc201730293877
SAMN10139195B13519SRR79094083FIc201730293877
SAMN10139295B12749SRR79091662EIc201730293877
SAMN10139364B12552SRR79091922EIc201730293877
SAMN10139361B12555SRR79091832EIc201730293877
SAMN10139289B12776SRR79093082DIc201730293877
SAMN10139190B13558SRR79092462DIc201730293877
SAMN10142006B13343SRR7909249NAGIb201730293877
* PubMed Study Identifier.
Table 2. Candida auris whole-genome sequence benchmark dataset sequence metrics.
Table 2. Candida auris whole-genome sequence benchmark dataset sequence metrics.
BioSampleIsolate IDSRA RunTotal Reads (Millions)Avg. GC Content (%)Assembly LengthContigsN50
SAMN10139509B12352SRR79092823.445%12,219,46399720,257
SAMN10139317B12681SRR79092146.247%11,914,250103019,891
SAMN10139320B12677SRR79091356.246%12,125,777100620,610
SAMN10139376B12490SRR79092103.643%12,215,65198921,267
SAMN10139493B12427SRR79091842.842%12,209,708103020,335
SAMN10139490B12430SRR79092333.243%12,212,62699020,510
SAMN10139465B12461SRR79092345.243%12,223,488103320,401
SAMN10139461B12467SRR79092975.444%12,227,902104920,090
SAMN10139268B12824SRR7909313543%12,217,316107319,453
SAMN10139267B12825SRR79093856.444%12,235,136101121,091
SAMN10139529B12044SRR79091473.243%12,219,48797921,004
SAMN10139470B12456SRR79091564.443%12,218,763104919,233
SAMN10139462B12466SRR7909413543%12,223,757103819,885
SAMN10139189B13613SRR79092844.843%12,225,91397621,224
SAMN10139194B13520SRR79093944.243%12,215,973101620,348
SAMN10139195B13519SRR79094082.844%12,161,014103920,040
SAMN10139295B12749SRR79091667.644%12,234,23999321,085
SAMN10139364B12552SRR79091924.643%12,217,67998321,519
SAMN10139361B12555SRR79091833.643%12,215,60998221,427
SAMN10139289B12776SRR79093086.842%12,228,29398921,510
SAMN10139190B13558SRR79092465.244%12,222,52098221,174
SAMN10142006B13343SRR79092494.241%12,213,89099721,547
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Welsh, R.M.; Misas, E.; Forsberg, K.; Lyman, M.; Chow, N.A. Candida auris Whole-Genome Sequence Benchmark Dataset for Phylogenomic Pipelines. J. Fungi 2021, 7, 214. https://doi.org/10.3390/jof7030214

AMA Style

Welsh RM, Misas E, Forsberg K, Lyman M, Chow NA. Candida auris Whole-Genome Sequence Benchmark Dataset for Phylogenomic Pipelines. Journal of Fungi. 2021; 7(3):214. https://doi.org/10.3390/jof7030214

Chicago/Turabian Style

Welsh, Rory M., Elizabeth Misas, Kaitlin Forsberg, Meghan Lyman, and Nancy A. Chow. 2021. "Candida auris Whole-Genome Sequence Benchmark Dataset for Phylogenomic Pipelines" Journal of Fungi 7, no. 3: 214. https://doi.org/10.3390/jof7030214

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