Introduction
Fungal pathogens cause infectious disease in many organisms [1, 2] among both animals [3] and plants [4]. Fungal pathogens of agriculture such as Puccina [5], Botrytis [6], and others [7] can be devastating to yields of many different crops [8] and endanger food security among a growing population [9]. In addition, the production of mycotoxins by fungi such as Fusarium graminearum [10] can contaminate agricultural products, [11] and threaten human and animal health if consumed [12, 13]. Fungal pathogens in humans cause comparable mortality to tuberculosis and malaria [14]. The World Health Organization (WHO) recently published a list of the most concerning fungal pathogens, [15] highlighting the global concern over fungal infection. The repeated use of antifungals with limited number of modes of action results in an increase in resistant strains within fungal populations that are pathogens of humans or agriculture [16, 17].
Fungal infections in humans can range from mild subcutaneous and mucosal infections [18] to life-threatening systemic disease. In particular, the rise of sophisticated medical devices, interventions, and therapeutics has increased vulnerable and immunocompromised patient populations that tend to be more susceptible to mycoses [19, 20]. Populations that are especially vulnerable to fungal infections include prematurely born children [21] and AIDS patients, [22] leukemia, [23] and organ transplant patients [24]. Fungal infections can be a heavy cost for the health care system. For example, in 2018 there were 666,235 fungal infections were diagnosed in the United States alone [25–27]. Specifically, Candida yeasts are a leading cause of nosocomial fungal infections, particularly Candida albicans, which is a common fungal species associated with human mucosal surfaces [28]. Non-life-threatening infections include mucosal infections by Candida yeasts. Specifically, vulvovaginal candidiasis affects 75% of women at least once in their lifetime and is a major cause of morbidity, especially due to recurrent infections caused by drug-resistant species of yeasts [29]. Invasive fungal diseases can have high mortality rates, depending on the organisms. Recent outbreaks of mucomycoses associated with the COVID-19 pandemic have also highlighted the lack of effective fungal treatments, with mortality rates > 40% [30].
Agriculture incurs substantial losses from many different fungal pathogens in all regions of the world [31]. Infection by pathogens such such as Pyricularia oryzae and Fusarium species, that can cause devastating economic losses [10] costing farmers billions of dollars annually [32] in antifungal, i.e., fungicide, treatments. For example infection by Pyricularia oryzae, the causal organism of rice blast, can reduce rice productivity by 10-35%. Ustilago maydis, causing corn smut [33], can reduce corn yield by 2-20% [34]. Fungal plant pathogens can have a wide host range, such as Botrytis cinerea and Colletotrichum truncatum [35], or a narrow host range, such as Blumeria graminis [36]. Globalization can also introduce new fungal pathogens to agriculture from different regions of the world, potentially introducing new fungi for which plants and farmers have no experience [37], such as the wheat pathogen Mycosphaerella graminicola, which spread to the Americas and Australia over the last 500 years [38]. Similarly, wheat rust Puccinia graminis tritici strain Ug99 presents a major threat to food security, spreading from east Africa into Asia [39]. In addition to acting as direct pathogens, some fungi, such as saprophytes, can spoil food stores [40]. Repeated use of fungicides in agriculture also selects for resistance in Aspergillus to azoles [41]; this is problematic because azoles are used for both agricultural and human medicinal applications [42].
It is widely understood that there is a critical need for new classes of antifungal compounds [43, 44]. Developing antifungal compounds with novel modes of action will provide more treatment options, reducing the need for repeated applications and selection for resistance. One key challenge in the development of new antifungal drugs is the lack of novel protein targets. An ideal compound should maximize harm against the pathogen and minimize off-target effects for the host organism, which can be accomplished by designing compounds to bind targets that are absent in the host. This can be challenging due to the shared eukaryotic ancestry and similar but not identical biochemical pathways [45] between fungi, plants, and humans. For example, within fungal systems, ergosterol serves many of the same cellular functions that cholesterol serves in humans, but is not present in animals, hence proteins in the ergosterol synthesis pathway have been targeted by many different antifungals (S1 Table). Similarly, azoles targeting 14-demethylase [46, 47] and morpholines targeting Erg2 and Erg24 in the ergosterol synthesis pathway are commonly used antifungals [48] (S1 Table). Likewise, the echinocandins target the pathway of cell wall -(1,3) glucan synthesis [49], but are exclusively used against human and animal infections [50, 51]. Because the cell wall is a unique structure to fungi, this pathway is absent in humans and leads to fungal osmotic instability and death [52]. Unfortunately, many of these antifungal compounds also have undesirable properties (S1 Table). Azoles are used against both human and agricultural pathogens [53]. Despite their potent activity against fungal pathways of ergosterol synthesis, they have been associated with hepatotoxicity and numerous hormone-related effects [54]. In addition, polyenes such as amphotericin B have severe renal toxicity [55, 56]. Proliferation of resistance to current antifungal compounds further highlights the need for new modes of action [53]. Examples include Candida’s resistance to echinocandins [57] and flucytosine [58], azole resistance in Aspergillus [59] and Candida [60–62] and fungal species that are resistant to multiple types of antifungal drugs [63]. Some other minor classes of antifungals exist in addition to those mentioned above [64], and others used exclusively in agriculture [65].
Identification of target proteins can be done with subtractive genomics [66], which identifies potentially targetable molecules based on differences in alignment. “Subtractive genomics” can refer to protein sequences or to genomic pathways, or to genomic pathways where orthologs are removed [67], and will be referred to in the protein sense. This approach has been used to identify many potential protein targets in Mycobacterium tuberculosis [68], Mycoplasma pneumoniae [69], and Mycoplasma genitalium [70]. The only study that used subtractive genomics to identify anti-fungal targets has been on Histoplasma capsulatum, using the similar process called “reverse vaccinology” to identify drug and vaccine candidates [71]. However, most fungal pathogens have never been analyzed with subtractive genomics.
Computer-aided drug design, or CADD [72] has been used in the design and development of antifungals to treat human disease and it is significantly cheaper and faster for screening large libraries of compounds. Virtual screening is performed by using docking strategies to simulate the binding of a ligand with a given protein target [73], then applying various scoring functions to estimate the protein-ligand binding strength. A common method for developing antifungal compounds involves high-throughput empirical screening of large chemical libraries to identify possible antifungal candidates [74]. This process is expensive and time-consuming, with the added challenge that the cellular target of the compound is unknown [75, 76]. CADD has been used to identify further inhibitors of known drug targets, but has yet to produce a viable antifungal drug [77]. At least 70 human drugs have been approved that used virtual screening as part of the drug discovery process, including Captopril, Norfloxacin, and Imatinib, but antifungals were not among them [78]. Recent advances in computational determination and approximation [79] of protein structure are also accelerating the pace of drug discovery due to the increased availability of protein structures.
In this study, a bioinformatics pipeline, HitList, was developed and used to identify possible antimicrobial targets for fungi. The purpose was to discover novel antifungal targets for the World Health Organization critical fungal pathogen list [15] and the top 10 agricultural fungal pathogens [10]. The current study started with a list of essential genes from the model fungus Saccharomyces cerevisiae. Essential genes with known human and/or plant orthologs were removed. Subtractive genomics [66] was then used to identify protein regions that could serve as targets. HitList surveyed more than a thousand proteins. HitList was validated by identifying proteins that already have known antifungal inhibitors, but we also found eight proteins that had not been previously considered as antifungal targets.
Methods
Source data and resources
Protein sequences encoded by essential genes from the Database for Essential Genes (DEG) [80] for S. cerevisiae (yeast) were downloaded from http://essentialgene.org. All hosts and pathogens are listed in Table 1. The pathogen targets were identified by utilizing the World Health Organization critical pathogen list [81] (S4 Table) and the top 10 agricultural fungal pathogen list (S5 Table) [10]). Proteome data was downloaded for common agricultural organisms that can be hosts for fungi Homo sapiens (human), Glycine max (soy), Oryza sativa (rice), Solanum tuberosum (potato), and Zea mays (corn) from https://ftp.ncbi.nlm.nih.gov/genomes/refseq/ (S3 Table). Pathogen proteomes were downloaded from from https://ftp.ncbi.nlm.nih.gov (S4 and S5 Tables).
[Figure omitted. See PDF.]
Bioinformatics pipeline
Our HitList pipeline starts with all genes from S. cerevisiae as obtained from the DEG [82]. Of the 1,110 that were considered essential, 909 genes that have human orthologs according to the Alliance of Genome Resources [83] were eliminated from consideration (Fig 1). The remaining 201 genes were used as queries in a BLASTP analysis using default parameters [84] (version 2.14) against all hosts and pathogens proteomes. Proteins with homology to query proteins having an expectation value above 0.1 were excluded. For each query protein, the top resulting protein from each pathogen and each host BLAST analysis was placed together and aligned with Clustal 1.2.4 [85] to obtain multiple sequence alignments (MSAs). MSA regions that show alignment with pathogens and not with hosts are considered potentially targetable, using an approach that has previously been called “subtractive genomics” [66]. This approach identifies amino acid sequences that are likely only present in the pathogens, and absent in the hosts, avoiding effects on the host. Analgous to a study showing that amino acid sequence could determine whether or not an immunoglobulin E protein would bind to casein for milk-allergic patients [86], we sought to identify amino acid regions of proteins that would be susceptible to any sort of potential inhibitor or antibody.
[Figure omitted. See PDF.]
Considering only genes that have no human ortholog according to the Saccharomyces Genome Database (SGD) [88], the field of essential genes was narrowed to 201. These genes are then used as BLAST [89] queries against all pathogen and host proteomes (S1 Section). Clustal was then used to create MSAs to identify targetable regions via subtractive genomics [66].
Fig 2 shows an example comparing two proteins, one with a poor targetability, and another with desirable targetability. The targetability of a region is quantified by the number of pathogens present at each residue in the MSA, the more pathogen proteins are present within a given region, the better that region is for a target. While it is beyond the scope of this study, proteins with known experimental, e.g., NMR or X-ray, structures are preferred, as eventual structure-based drug design [87] would have a more reliable starting point for design of antifungal compounds. Additionally, protein location within the cell is also important, as drugs must be able to bind the target protein wherever it is found in the cell.
[Figure omitted. See PDF.]
An example of two essential S. cerevisiae proteins analyzed for target suitability among the top 10 agricultural fungal pathogens. The figure shows results from BLAST searches performed for each essential gene with alignment statistics shown. (A) Acc1 is an example of a poor target that shows high similarity between homologous proteins of hosts and pathogens, while on the right Fcy21 is an example of a potentially good target that shows neither desirable expectation values nor long alignment lengths with host proteins. (B) The desirable alignment between host and pathogen proteins for Acc1, and the lack of host proteins for Fcy21 is visible in multiple sequence alignments, which are visualized using a modified version of CIAlign [90].
The similarity of the amino acids within each MSA was quantified by Sneath’s similarity index [91] (S2 Table). Sneath’s was chosen because of its intuitive nature, in that it ranks within (0,1], and an amino acid compared with itself can be exactly 1. The Sneath index is calculated at each position within an MSA by comparing every amino acid in a group, against every other amino acid in that group:
(1)
where M values for two amino acids i and j are given by the Sneath similarity in S2 Table, and Nseq is the number of sequences at that position. For example, consider two peptides, each containing nine alanine residues. In this case, all Sneath similarity values would be 1.0. Comparing a 9-mer of alanine against a 9-mer of cysteine would give a Sneath similarity of 0.87 across the 9 residues. Uncertain amino acids, e.g. “X” are set to the minimum Sneath value of 0.003 to avoid type II errors. If a known amino acid is compared with an ambiguous amino acid, e.g. A with B, where B could be either D or N, then the Sneath index of A and B is set at the mean of MA/D and MA/N.
Results & Discussion
In this study, our newly developed bioinformatics pipeline HitList was used to identify antifungal protein targets against two groups of pathogens: WHO fungal pathogens (S4 Table), and agricultural pathogens (S5 Table). Desirable qualities in protein targets include (e.g., “Fcy21” in Fig 2): (1) High bit scores and longer alignment lengths to an essential protein for S. cerevisiae and not to any host protein. (2) Homogeneity as measured by approximately within the range . That is, proteins in the top right corners of Fig 3 represent the best preliminary targets. Proteins in the top left will have shorter targetable regions, but could be potential targets. Conversely, proteins that have good alignment between both host and pathogen proteins and to the S. cerevisiae essential protein, especially over the entire length, are considered less desirable targets (e.g., “Acc1” in Fig 2).
[Figure omitted. See PDF.]
The output of the pipeline is visualized by a scatterplot of alignment length, mean pathogen column occupancy, and (Eq 1). Labeled proteins are those with long targetable regions, and large and mean column occupancy values.
Table 2 summarizes our list of most promising potential protein targets. Of the 16 proteins in the table, seven are in common between WHO and agricultural fungal pathogens and thus could be targets that lead to the development of broad-spectrum antifungal compounds. Detailed figures and tables are shown for each protein in Table 2, sorted alphabetically in S2 Section, containing S1-S197 Figs and S6-S39 Tables. While the potential target list is greater than these 16 proteins, these are the targets that met our criteria listed above in Fig 3 and hence further analysis will focus on the proteins listed in Table 2. A larger number of possible protein targets were found for the WHO pathogens than for agricultural pathogens, which is expected because agricultural hosts include not only human but also a wide phylogenetic range of plants, as compared to WHO pathogens that only include human as a host.
[Figure omitted. See PDF.]
Identification of previously identified targets validates the pipeline
Fig 3 shows that HitList identifies previously known targets that already have known inhibitors, detailed information of which is included in S3 Section, containing S198-S205 Figs and S40-S45 Tables. HitList selected Fas1 and Fas2 (S2.5 and S2.6 Sections); these subunits of fatty acid synthase [108] have been previously identified as viable targets against fungi, and inhibitors have already been found [97–99], serving as a validation of HitList. Fas1 is the specific target of the antifungal compound NPD6433 [98, 109]. Our analysis suggests Fas2 is a better target for WHO pathogens compared to agricultural pathogens, because host plant proteins show weakly aligned sequences, and fungal sequences also align more weakly (S58 and S61 Figs). Indeed, the natural products CT2108A and CT2108B are effective antifungals against the FAS complex in Candida, S. cerevisiae, and Cryptococcus, yet both are made by the fungus Penicillium solitum, so broad antifungal activity is less likely. Fas1 BLAST hits for the plant pathogens P. graminis, P. triticina, and P. striiformis are much weaker than for other agricultural pathogens, as demonstrated by the lower value in Figs 3, as well as weaker alignments in S50 and S51 Figs. BLAST searches against the non-redundant protein database show that Fas1 is broadly conserved across many different genera of fungi from Asomycota, Basidomycota, and Zoopagomycota (S57 Fig) and Fas2 (S69 Fig). Table 2 also shows Chitin synthase 2 (Chs2) as a possible agricultural target, and this protein already has known inhibitors including nikkomycins and polyoxins [94–96]. However, Chs2 may not be a good choice for drug targeting due to strong homology to the human protein hyaluronan synthase. Nonetheless, chitin synthase (see Section S2.3) has no equivalent in humans or plants according to the SGD, agreeing with a search against the NCBI’s non-redundant database [110] (NR) (see S29 & S34 Figs). Even if Chs2 were successfully targeted, a Chs2-knockout strain of the human pathogen C. albicans did not have attenuated virulence [111], so targeting Chs2 with potential therapeutics may not be effective in treatment of infections. Finally, the plasma membrane transporter Alr1 [112] was identified in the pipeline, which has been shown to be a potential antifungal target and is known to be inhibited by Bovine pancreatic trypsin inhibitor [92]. Folate synthesis (Fol1) is an effective target of antibacterials, such as trimethoprim and sulfamethoxazole [113], and has been effective in fungi too [114].
HitList also identified some proteins that have been previously suggested as potential antifungal targets in the literature but have no known inhibitors. Proteins that are part of chemical pathways that are absent in hosts are especially attractive, as the chance of side effects is significantly lower. For example, ceramide phosphoinositol transferase, part of sphingolipid synthesis (Aur1), [115] and fructose 1,6-bisphosphate aldolase (Fba1) [100] were identified as potential antifungal targets by this analysis and others, and do not have mammalian equivalents [93, 115].
The pipeline identified Trl1, which is responsible for the splicing of introns from nascent tRNA as a potential target. Since this is done very differently in fungi compared to metazoa, Trl1 is a possible antifungal target [102–104, 116]. Yeast Elongation factor 3 (Yef3 or eEF3) is very well conserved among fungi [105, 117] and identified as a potential target using this pipeline. Because non-fungi use only two elongation factors when translating mRNA, as opposed to three for fungi, Yef3 has potential as a target [106, 118] Indeed, S192 & S197 Figs both show strong conservation of Yef3 across the fungal kingdom, however, this study also reveals 960 metazoan species with strong hits (S196 Fig). This makes intuitive sense because Yef3 contains ATP-binding cassette domains [119] that are very common throughout many domains of life (S39 Table), and inhibitors must be carefully designed to avoid unintentional effects on host organisms. Further, results suggest that Trl1 and Yef3 are less likely to be broad spectrum targets as can be seen with the relatively low and weaker MSA (Figs 3, S176, S186), compared to other proteins identified in the pipeline.
Identification of eight novel antifungal protein targets
Most importantly, our study has also uncovered a rich variety of novel possible protein targets that have not been previously identified to our knowledge (Table 2): Erg8, Fcy21, Ilv3, Ilv5, Rib3, Rib5, Ssy1, and Ste12. As will be detailed below, some of these targets are particularly well-suited for WHO pathogens and some for agricultural pathogens, while others appear to be candidates for antifungal development against both pathogen lists.
HitList identified potential broad-spectrum antifungal target proteins that are particularly well-suited for treating pathogens in the WHO pathogen list. The protein pair Ilv3 and Ilv5, [120] involved in branched chain amino acid synthesis, [121, 122] is an especially attractive target because 1) no human proteins have strong alignments via BLAST, 2) there is a high value between pathogen hit sequences, and 3) branched chain amino acid synthesis does not occur in metazoa [123]. The last point is critical since inhibition of a protein pathway that is present in a pathogen, but not in a host, is unlikely to cause harm to the host. The dihydroxyacid dehydratase enzyme performs the same chemistry as Ilv3 in cyanobacteria and is inhibited by aspterric acid [124]. Further experimentation is necessary to determine whether aspterric acid or related derivatives have potential to function as a broad-spectrum antifungals. The pipeline also identified Rib3 and Rib5 as attractive protein targets for the WHO pathogen list, with high values. These proteins function in riboflavin synthesis, which does not occur in metazoa. Our study suggests that Ilv3, Ilv5, Rib3 and Rib5 are not as suitable for developing agricultural antifungals as for humans, because riboflavin [125] and branched chain amino acid synthesis [122] also occurs in plants and could potentially lead to phytotoxicity (see S136 and S151 Figs).
Ssy1 is an amino acid sensor protein that has potential for broad-spectrum use against both agriculture and WHO pathogens, but appears to be better suited for agricultural application. This apparently novel antifungal target shows much stronger alignment with Candida pathogens than Cryptococcus (S154 Fig), which reduces the value within WHO pathogens. Furthermore, Nakaseomyces shows great differences with Candida and Cryptococcus sequence hits with Ssy1 (S153 and S154 Figs). BLAST hits against the Non-Redundant protein database animal and plant proteins are few and weak (S159 and S164 Figs), so any well-designed drug against Ssy1 would be unlikely to affect animals and plants. Furthermore, as a membrane protein, drug design need not be concerned with membrane penetration.
Fcy21, for “flucytosine resistance,” is another potential novel protein target for antifungal compounds, which has potential to be an antifungal target for WHO human pathogens. Fcy21 is a putative purine-cytosine permease [126, 127]. This protein is related to the target of flucytosine (Table 2) Fcy2p, but cannot substitute for its function [126] No human protein showed significant alignment with Fcy21 (S82 Fig), but strong conservation is seen among pathogenic fungi, apart from the first 100 residues in the N-terminal domain. No non-fungal orthologs exist according to the SGD, and searches against the non-redundant database indicate strong conservation among WHO pathogenic fungi, and no hits among metazoa and insignificant hits among plants (S92 Fig). Interestingly, Fcy21 does not have homology with proteins from three of the agricultural fungal pathogens of agricultural products, namely Puccina and P. oryzae (S85 Fig and S21 Table), hence Fcy21 is not likely to be as useful for developing agricultural antifungals. Ste12 was identified by HitList for agricultural pathogens, but not for human pathogens, and was not labeled due to weak alignment with the model S. cerevisiae (Fig 3). Ste12, a transcription factor that is important for the virulence of mycoparasites, including Trichoderma atroviride which is a parasite of plant pathogens, [128, 129] shows good antifungal potential for the top 10 agricultural fungal pathogens (S2.15 Section). Ste12 is one of the two proteins that is a good target against agricultural pathogens, but not against human pathogens (S165 and S168 Figs).
Our study shows that agricultural host proteomes have much stronger hits to Erg8 (S38 Fig) than human (S35 Fig), so any antifungal drug targeting of Erg8 would likely be more effective against human pathogens. Erg8 is part of the ergosterol synthesis pathway, enabling phosphomevalonate kinase activity. While the ergosterol synthesis pathway is well-studied and targeted by azole [47, 130] and morpholine antifungals [131], Erg8 appears to be a novel potential target. The difference in primary sequence of the Erg8 homolog from Cryptococcus neoformans compared to the homologs from other pathogen species is significant, with large gaps in the multiple sequence alignment because of C. neoformans (S35 Fig).
Limitations of the study
The primary limitation of the pipeline is that it relies on the list of essential genes. Here, essential genes for S. cerevisiae from DEG that do not have human orthologs according to the SGD are used. If a gene is not on this list, this gene will not be able to identified as a target. For example, consider Fks1, the target of echinocandin antifungals. Fks1 does not appear on our list because the -1,3-glucan synthase proteins (Fks1, Fks2, Fks3) are not considered essential genes according to the DEG [80]. Nonetheless, BLAST analysis of Fks1 against the non-redundant database identifies protein homologs in 1,149 fungal species (S203 Fig), and a multiple sequence alignment of these demonstrates that Fks1 is an ideal target (S202 Fig), as there are very large regions of the protein in the MSA that only have alignment to pathogen protein domains. This shows that Fks1 would have appeared on our final list of target proteins if it was in the initial pool of genes considered. Such results should be expected, as fungal cell wall synthesis is unique to fungi [132]. Furthermore, if a gene is essential in the model, but isn’t essential in the target pathogen, for example Chs2, HitList will identify a false positive.
Another potential weakness of HitList is our elimination of human orthologs. While this step reduces the chances of side effects in a resulting therapeutic, it could also obscure potential targets. For example, the antimalarial drug methylene blue targets the glutathione reductase protein in Plasmodium falciparum [133], in spite of a very similar protein being present in the human host. However, HitList is designed with caution in mind, and avoiding potential side effects to increase the chance of developing a successful therapeutic.
HitList also requires a well-characterized organism closely related to target pathogens such as S. cerevisiae, with an annotated genome and/or proteome, and a list of essential genes. For this study, NCBI had all desired pathogens available. However, niche organisms may not necessarily be available with the necessary quality for analysis. This study was possible due to the very high quality genome annotation of S. cerevisiae, and the availability of the pathogen data from NCBI.
HitList is designed to avoid possible effects on the host, and does so at the possible risk of missing otherwise good targets. For example, Erg11, the target of azole antifungals (S1 Table), has a high homology to a human protein, which can be further seen (S198 Fig). Indeed, human lanosterol and non-targeted cytochrome P450 are also affected by azole antifungals [134]. Similarly, Erg24 is a target of the morpholine antifungals, and shows a desirable sequence similarity with delta(14)-sterol reductase, and has human orthologs [83].
Evaluation of HitList cannot be evaluated by usual measures such as false positives, true negatives, etc., as no quantifiable metric exists for what constitutes a targetable protein, and personal opinion may differ from one individual to another. Databases are not necessarily consistent with proteins that are considered human homologs and not, so which proteins should be fed into the pipeline is not always clear. Also, few organisms are as well annotated as humans and S. cerevisiae are. Potatoes for example, and do not necessarily have an exact equivalent in the database for each protein. Furthermore, the lengths that are found targetable for each protein may be usable for some proteins, but not others.
Collateral damage of targeting potential proteins should also be taken into account. For example, symbiotic bacteria and mycorrhizal fungi [135, 136] were not included in this study’s host-pathogen relationship. Indeed, the location of the symbiotes and which part of the plant is affected by the pathogen should be considered, i.e. whether applied to leaves, roots, etc. Additionally, neither the impact on insects that prey on herbivores [137], nor downstream environmental impacts were considered. Off-target damage can be minimized by considering proteins that are fungal-specific and have no equivalent in any animal or plant, for example Yef3. Chs2, for example, although not recommended for targeting here, has equivalents in insects and fish.
We note that our strategy for validating HitList is its ability to find previously identified targets such as Fas1 and others. Ideally, it would be possible to quantify HitList’s performance in terms of accuracy and precision, however, this is not possible for the following reasons: 1) Essential gene lists can differ depending on how many are available for the model in question, depending on methods used for determination of essentiality [138], so an exact determination of HitList’s accuracy, precision, etc. is not possible. 2) The particular model organism used may not be appropriate for the pathogens in question. For example, fungi have at least two well-characterized species, S. cerevisiae and S. pombe, and for some pathogenic fungi, one model or the other may be appropriate, adding uncertainty. 3) The exact amino acid length necessary to target a protein can vary depending on antibody or inhibitor design capability, further eroding any clarity between what constitutes a true and false positive. 4) What constitutes an “ortholog” can vary between databases, and if A is an ortholog of B in one database, B is not guaranteed to be an ortholog of A in another database. 5) Stronger genomic data in the future could lead to changes in what is considered targetable.
In order to use CADD techniques, the identified target proteins should also be well-characterized structurally, e.g. by NMR or X-ray crystallography. Proteins should have identifiable binding pockets to be used for computational chemistry docking programs, so that potential inhibitors can be screened [139]. Many proteins in Table 2, such as Ssy1 and Fcy21, do not have any experimental structures available as of publication, while proteins such as Yef3 does have an X-ray structure available [106]. The paucity of experimental structural data in the Protein Data Bank [140] for the proteins that are found to be attractive targets here also precludes 3-dimensional structural searches, such as Dali [141] and FoldSeek [142], and harms AlphaFold [79] predictions by having an inadequate training set. The inability to perform structural searches against potential targets means that we do not have a means to check whether similar binding pockets may exist in host proteins, leading to possible off target effects for the resulting treatment. While AlphaFold structures can provide good estimates, experimental structures are preferred. Future research of possible protein targets should consider the presence and quality of existing structures.
Diversity of pathogens and hosts hinders target selection
The phylogenetic diversity of the ascomycetes is very wide [143], complicating identification of targets. Consider the large evolutionary distance between the two ascomycete genera Saccharomyces and Schizosaccharomyces, which is on the order of 350 million years of divergence [144]. Indeed, Cryptococcus and Puccina are both genera within basidomycota, which is in an entirely different phylum than S. cerevisiae’s ascomycota, and at an even greater evolutionary distance than between Schizosaccharomyces and Saccharomyces. No basidomycete has the quality of annotation that S. cerevisiae has and a known essential gene list, so an ascomycete pathogen is the best choice at present. Nonetheless, potential protein targets were found that included the basidomycetes, indicating conservation of genes over very long periods in distantly related phyla [145].
The number of hosts/pathogens and their identities affects the possible protein targets and the length of the targetable regions identified by the pipeline. Consider Ssy1, which has a much longer targetable section for the WHO pathogens than for the top 10 agricultural pathogens (Fig 3 and S2.14 Section). An MSA of the Ssy1 homologs from the top 10 agricultural pathogen and respective host proteomes shows large gaps, reducing the potential target length (S156 Fig). Gaps also exist for Ssy1 homologs from the WHO pathogen proteomes (S156 Fig) in approximately the same regions as for the Ssy1 homologs from the top 10 agricultural pathogens, but the homologs from the Candida species and N. glabratus proteomes exhibit homology across these gaps, lengthening the potential region that could be targeted with antifungal compounds. For the agricultural pathogens, the plant proteins within the MSA shorten the targetable region. The larger the number of hosts and pathogens that are included in the analysis, and the more diversity within each group, the fewer and shorter the targets will be for antifungal compounds.
The length of targetable proteins is reduced by increasing diversity of pathogens and hosts, which is a consequence of diversity in primary sequence. Consider, for example, using only Candida pathogens in the WHO list for HitList. When this analysis is performed, Ccc1 shows as targetable, but does not show as targetable when the entire WHO list is considered. Ccc1 [146] (Section S4.1, S206-S212 Figs, S46-S47 Tables) is a / transporter that has previously been identified as a potential antifungal target [147]. Indeed, Ccc1 is particularly attractive because no metazoan equivalent exists, and Candida is a human pathogen. Cryptococcus proteins clearly cluster differently in the MSA from the other fungal genera (S206 and S207 Figs), which is intuitive as Cryptococcus is a basidomycete, while all other pathogens are ascomycetes. Candida pathogen proteins clearly show both desirable alignment to one another and to S. cerevisiae, while Cryptococcus shows its own cluster that doesn’t align strongly to S. cerevisiae (S207 Fig). Strong homology exists between the Candida proteins and S. cerevisiae, making more probable that this protein is essential in Candida as well, agreeing with S207 Fig. Inspection of S206 Fig implies that the broad-spectrum targeting region is approximately from residue 110-170 in MSA coordinates, a narrow region of the protein to target with a compound. Given that antibodies can target decapeptides [86], Ccc1 is a possible target for Cryptococcus, but development of an antifungal drug could be challenging. By contrast, using only Candida species as pathogens suggest that residues 100-340 are targetable (S206 Fig), which could simplify compound design. Thus, when using HitList, the number of pathogens should be kept to a minimum, and the pathogens that are used should be as closely related to one another as possible.
Conclusion
A new automated bioinformatics pipeline (HitList) that uses subtractive genomics to identify protein targets in parallel for any pathogen-host combination was developed, able to test thousands of proteins within a few hours on personal computers. HitList was applied to the discovery of novel antifungal targets for the WHO critical list of fungal pathogens and the top 10 agricultural fungal pathogens, using essential genes from S. cerevisiae. Current antifungal targets, such as Fas1 and Fas2, and previously hypothesized antifungal targets such as Trl1 and Yef3 were identified as potential targets by HitList. Both lists of pathogens and hosts showed established targets. Many previously identified targets that were identified do not yet have inhibitors, such as Alr1, Aur1, and Trl1. Most importantly, HitList identified novel protein targets that could be used to develop broad-spectrum antifungals that have not been previously mentioned in the literature, to our knowledge: Erg8, Fcy21, Ilv3, Ilv5, Rib3, Rib5, Ssy1, and Ste12. Based on our analysis, we anticipate that some of these targets will be more well-suited for developing antifungals against the WHO list (Erg8, Ilv3, Ilv5, Rib3, and Rib5), Ste12 against the agriculture list only, and others against both lists (Fcy21 and Ssy1). In addition, stronger potential targets are identified for the WHO/human pathogen list, since it has a single host, as compared to the agricultural top 10 list, that has five hosts across two different domains of life. While HitList identified proteins that could potentially be targets of compounds for antifungal purposes (Table 2), more research is clearly needed in order to validate these targets and develop possible inhibitors that could lead to new classes of antifungal drugs. Finally, we note that HitList could be used to investigate any list of pathogens and hosts for any kingdom of life, provided that a list of essential genes exists, and genomes and/or proteomes are available. The HitList software to run the bioinformatics pipeline and identify potential protein targets is freely available at https://github.com/hhg7/HitList.
Supporting information
Supporting Information. Supporting Information files
https://doi.org/10.1371/journal.pone.0323991.s001
(PDF).
Acknowledgments
Computational resources were provided in part by Research Computing and Data Services in the Institute for Interdisciplinary Data Science at University of Idaho.
References
1. 1. Dickman MB, de Figueiredo P. Comparative pathobiology of fungal pathogens of plants and animals. PLoS Pathog. 2011;7(12):e1002324. pmid:22194681
* View Article
* PubMed/NCBI
* Google Scholar
2. 2. Sexton AC, Howlett BJ. Parallels in fungal pathogenesis on plant and animal hosts. Eukaryot Cell. 2006;5(12):1941–9. pmid:17041185
* View Article
* PubMed/NCBI
* Google Scholar
3. 3. Heitman J. Microbial pathogens in the fungal kingdom. Fungal Biol Rev. 2011;25(1):48–60. pmid:21528015
* View Article
* PubMed/NCBI
* Google Scholar
4. 4. van Agtmaal M, Straathof A, Termorshuizen A, Teurlincx S, Hundscheid M, Ruyters S. Exploring the reservoir of potential fungal plant pathogens in agricultural soil. Appl Soil Ecol. 2017;121:152–60.
* View Article
* Google Scholar
5. 5. Ajala A, Asipita OH, Michael AT, Tajudeen MT, Abdulganiyyu IA, Ramu R. Therapeutic exploration potential of adenosine receptor antagonists through pharmacophore ligand-based modelling and pharmacokinetics studies against Parkinson disease. In Silico Pharmacol. 2025;13(1):17. pmid:39872470
* View Article
* PubMed/NCBI
* Google Scholar
6. 6. Hahn M. The rising threat of fungicide resistance in plant pathogenic fungi: Botrytis as a case study. J Chem Biol. 2014;7(4):133–41. pmid:25320647
* View Article
* PubMed/NCBI
* Google Scholar
7. 7. Sun S, Hoy MJ, Heitman J. Fungal pathogens. Curr Biol. 2020;30(19):R1163–9. pmid:33022261
* View Article
* PubMed/NCBI
* Google Scholar
8. 8. Strange RN, Scott PR. Plant disease: a threat to global food security. Annu Rev Phytopathol. 2005;43:83–116. pmid:16078878
* View Article
* PubMed/NCBI
* Google Scholar
9. 9. Bebber DP, Gurr SJ. Crop-destroying fungal and oomycete pathogens challenge food security. Fungal Genet Biol. 2015;74:62–4. pmid:25459533
* View Article
* PubMed/NCBI
* Google Scholar
10. 10. Dean R, Van Kan JAL, Pretorius ZA, Hammond-Kosack KE, Di Pietro A, Spanu PD, et al. The Top 10 fungal pathogens in molecular plant pathology. Mol Plant Pathol. 2012;13(4):414–30. pmid:22471698
* View Article
* PubMed/NCBI
* Google Scholar
11. 11. Luo S, Du H, Kebede H, Liu Y, Xing F. Contamination status of major mycotoxins in agricultural product and food stuff in Europe. Food Control. 2021;127:108120.
* View Article
* Google Scholar
12. 12. World Health Organization. Selected mycotoxins: ochratoxins, trichothecenes, ergot. World Health Organization; 1990.
13. 13. Richard JL. Some major mycotoxins and their mycotoxicoses—an overview. Int J Food Microbiol. 2007;119(1–2):3–10. pmid:17719115
* View Article
* PubMed/NCBI
* Google Scholar
14. 14. Kainz K, Bauer MA, Madeo F, Carmona-Gutierrez D. Fungal infections in humans: the silent crisis. Microb Cell. 2020;7(6):143–5. pmid:32548176
* View Article
* PubMed/NCBI
* Google Scholar
15. 15. World Health Organization. WHO fungal priority pathogens list to guide research, development and public health action. World Health Organization; 2022. Available from: https://www.who.int/publications/i/item/9789240060241
16. 16. Wiederhold NP. Antifungal resistance: current trends and future strategies to combat. Infect Drug Resist. 2017;10:249–59. pmid:28919789
* View Article
* PubMed/NCBI
* Google Scholar
17. 17. Lucas JA, Hawkins NJ, Fraaije BA. Chapter 2: The evolution of fungicide resistance. In: Advances in applied microbiology, vol. 90. Academic Press; 2015, pp. 29–92. Available from: https://www.sciencedirect.com/science/article/pii/S0065216414000021
18. 18. Kühbacher A, Burger-Kentischer A, Rupp S. Interaction of Candida species with the skin. Microorganisms. 2017;5(2):32. pmid:28590443
* View Article
* PubMed/NCBI
* Google Scholar
19. 19. Richardson M, Lass-Flörl C. Changing epidemiology of systemic fungal infections. Clin Microbiol Infect. 2008;14(Suppl 4):5–24. pmid:18430126
* View Article
* PubMed/NCBI
* Google Scholar
20. 20. Ashbee H, Gilleece M. Chapter 38: Pharmacogenomics of antifungal agents. In: Padmanabhan S, editor. Handbook of pharmacogenomics and stratified medicine. San Diego: Academic Press; 2014, pp. 879–900.
21. 21. Weimer KED, Smith PB, Puia-Dumitrescu M, Aleem S. Invasive fungal infections in neonates: a review. Pediatr Res. 2022;91(2):404–12. pmid:34880444
* View Article
* PubMed/NCBI
* Google Scholar
22. 22. Bitar D, Lortholary O, Le Strat Y, Nicolau J, Coignard B, Tattevin P, et al. Population-based analysis of invasive fungal infections, France, 2001–2010. Emerg Infect Dis. 2014;20(7):1149.
* View Article
* Google Scholar
23. 23. Leventakos K, Lewis RE, Kontoyiannis DP. Fungal infections in leukemia patients: how do we prevent and treat them?. Clin Infect Dis. 2010;50(3):405–15. pmid:20047485
* View Article
* PubMed/NCBI
* Google Scholar
24. 24. Pappas PG, Alexander BD, Andes DR, Hadley S, Kauffman CA, Freifeld A, et al. Invasive fungal infections among organ transplant recipients: results of the Transplant-Associated Infection Surveillance Network (TRANSNET). Clin Infect Dis. 2010;50(8):1101–11. pmid:20218876
* View Article
* PubMed/NCBI
* Google Scholar
25. 25. Rayens E, Norris KA. Prevalence and healthcare burden of fungal infections in the United States, 2018. Open Forum Infect Dis. 2022;9(1):ofab593. pmid:35036461
* View Article
* PubMed/NCBI
* Google Scholar
26. 26. Benedict K, Jackson BR, Chiller T, Beer KD. Estimation of direct healthcare costs of fungal diseases in the United States. Clin Infect Dis. 2019;68(11):1791–7. pmid:30204844
* View Article
* PubMed/NCBI
* Google Scholar
27. 27. Benedict K, Whitham HK, Jackson BR. Economic burden of fungal diseases in the United States. Open Forum Infect Dis. 2022;9(4):ofac097. pmid:35350173
* View Article
* PubMed/NCBI
* Google Scholar
28. 28. Barac A, Cevik M, Colovic N, Lekovic D, Stevanovic G, Micic J, et al. Investigation of a healthcare-associated Candida tropicalis candidiasis cluster in a haematology unit and a systematic review of nosocomial outbreaks. Mycoses. 2020;63(4):326–33. pmid:31930592
* View Article
* PubMed/NCBI
* Google Scholar
29. 29. Achkar JM, Fries BC. Candida infections of the genitourinary tract. Clin Microbiol Rev. 2010;23(2):253–73. pmid:20375352
* View Article
* PubMed/NCBI
* Google Scholar
30. 30. Hoenigl M, Seidel D, Sprute R, Cunha C, Oliverio M, Goldman GH, et al. COVID-19-associated fungal infections. Nat Microbiol. 2022;7(8):1127–40. pmid:35918423
* View Article
* PubMed/NCBI
* Google Scholar
31. 31. Savary S, Willocquet L, Pethybridge SJ, Esker P, McRoberts N, Nelson A. The global burden of pathogens and pests on major food crops. Nat Ecol Evol. 2019;3(3):430–9. pmid:30718852
* View Article
* PubMed/NCBI
* Google Scholar
32. 32. Jørgensen LN, van den Bosch F, Oliver RP, Heick TM, Paveley ND. Targeting fungicide inputs according to need. Annu Rev Phytopathol. 2017;55:181–203. pmid:28525306
* View Article
* PubMed/NCBI
* Google Scholar
33. 33. Brefort T, Doehlemann G, Mendoza-Mendoza A, Reissmann S, Djamei A, Kahmann R. Ustilago maydis as a pathogen. Annu Rev Phytopathol. 2009;47:423–45. pmid:19400641
* View Article
* PubMed/NCBI
* Google Scholar
34. 34. Fisher MC, Henk DA, Briggs CJ, Brownstein JS, Madoff LC, McCraw SL, et al. Emerging fungal threats to animal, plant and ecosystem health. Nature. 2012;484(7393):186–94. pmid:22498624
* View Article
* PubMed/NCBI
* Google Scholar
35. 35. Diao YZ, Zhang C, Lin D, Liu XL. First report of Colletotrichum truncatum causing anthracnose of tomato in China. Plant Dis. 2014;98(5):687. pmid:30708530
* View Article
* PubMed/NCBI
* Google Scholar
36. 36. Wyand RA, Brown JKM. Genetic and forma specialis diversity in Blumeria graminis of cereals and its implications for host-pathogen co-evolution. Mol Plant Pathol. 2003;4(3):187–98. pmid:20569378
* View Article
* PubMed/NCBI
* Google Scholar
37. 37. Santini A, Liebhold A, Migliorini D, Woodward S. Tracing the role of human civilization in the globalization of plant pathogens. ISME J. 2018;12(3):647–52. pmid:29330537
* View Article
* PubMed/NCBI
* Google Scholar
38. 38. Stukenbrock EH, Banke S, McDonald BA. Global migration patterns in the fungal wheat pathogen Phaeosphaeria nodorum. Mol Ecol. 2006;15(10):2895–904. pmid:16911209
* View Article
* PubMed/NCBI
* Google Scholar
39. 39. Singh RP, Hodson DP, Huerta-Espino J, Jin Y, Bhavani S, Njau P, et al. The emergence of Ug99 races of the stem rust fungus is a threat to world wheat production. Annu Rev Phytopathol. 2011;49:465–81. pmid:21568701
* View Article
* PubMed/NCBI
* Google Scholar
40. 40. Lacey J. Pre- and post-harvest ecology of fungi causing spoilage of foods and other stored products. Soc Appl Bacteriol Symp Ser. 1989;18:11S–25S. pmid:2508232
* View Article
* PubMed/NCBI
* Google Scholar
41. 41. Azevedo M-M, Faria-Ramos I, Cruz LC, Pina-Vaz C, Rodrigues AG. Genesis of azole antifungal resistance from agriculture to clinical settings. J Agric Food Chem. 2015;63(34):7463–8. pmid:26289797
* View Article
* PubMed/NCBI
* Google Scholar
42. 42. Kang SE, Sumabat LG, Melie T, Mangum B, Momany M, Brewer MT. Evidence for the agricultural origin of resistance to multiple antimicrobials in Aspergillus fumigatus, a fungal pathogen of humans. G3 (Bethesda). 2022;12(2):jkab427. pmid:34897421
* View Article
* PubMed/NCBI
* Google Scholar
43. 43. Nicola AM, Albuquerque P, Paes HC, Fernandes L, Costa FF, Kioshima ES, et al. Antifungal drugs: new insights in research & development. Pharmacol Ther. 2019;195:21–38. pmid:30347212
* View Article
* PubMed/NCBI
* Google Scholar
44. 44. Fernandes CM, Dasilva D, Haranahalli K, McCarthy JB, Mallamo J, Ojima I, et al. The future of antifungal drug therapy: novel compounds and targets. Antimicrob Agents Chemother. 2021;65(2):e01719-20. pmid:33229427
* View Article
* PubMed/NCBI
* Google Scholar
45. 45. Feofilova EP. The kingdom fungi: heterogeneity of physiological and biochemical properties and relationships with plants, animals, and prokaryotes (Review). Appl Biochem Microbiol. 2001;37:124–37.
* View Article
* Google Scholar
46. 46. Fromtling RA. Overview of medically important antifungal azole derivatives. Clin Microbiol Rev. 1988;1(2):187–217. pmid:3069196
* View Article
* PubMed/NCBI
* Google Scholar
47. 47. Sheehan DJ, Hitchcock CA, Sibley CM. Current and emerging azole antifungal agents. Clin Microbiol Rev. 1999;12(1):40–79. pmid:9880474
* View Article
* PubMed/NCBI
* Google Scholar
48. 48. Mercer EI. Morpholine antifungals and their mode of action. Biochem Soc Trans. 1991;19(3):788–93. pmid:1783217
* View Article
* PubMed/NCBI
* Google Scholar
49. 49. Denning DW. Echinocandin antifungal drugs. Lancet. 2003;362(9390):1142–51. pmid:14550704
* View Article
* PubMed/NCBI
* Google Scholar
50. 50. Szymański M, Chmielewska S, Czyżewska U, Malinowska M, Tylicki A. Echinocandins—structure, mechanism of action and use in antifungal therapy. J Enzyme Inhib Med Chem. 2022;37(1):876–94. pmid:35296203
* View Article
* PubMed/NCBI
* Google Scholar
51. 51. Kaya Y, Dokuzeylül B, Bakırel U, Or ME. Antifungal resistance and clinical significance in small animals. Ger J Vet Res. 2022;2:28–36.
* View Article
* Google Scholar
52. 52. Eschenauer G, Depestel DD, Carver PL. Comparison of echinocandin antifungals. Ther Clin Risk Manag. 2007;3(1):71–97. pmid:18360617
* View Article
* PubMed/NCBI
* Google Scholar
53. 53. Fisher MC, Alastruey-Izquierdo A, Berman J, Bicanic T, Bignell EM, Bowyer P, et al. Tackling the emerging threat of antifungal resistance to human health. Nat Rev Microbiol. 2022;20(9):557–71. pmid:35352028
* View Article
* PubMed/NCBI
* Google Scholar
54. 54. Benitez LL, Carver PL. Adverse effects associated with long-term administration of azole antifungal agents. Drugs. 2019;79(8):833–53. pmid:31093949
* View Article
* PubMed/NCBI
* Google Scholar
55. 55. Mohr J, Johnson M, Cooper T, Lewis JS, Ostrosky-Zeichner L. Current options in antifungal pharmacotherapy. Pharmacotherapy. 2008;28(5):614–45. pmid:18447660
* View Article
* PubMed/NCBI
* Google Scholar
56. 56. Zotchev SB. Polyene macrolide antibiotics and their applications in human therapy. Curr Med Chem. 2003;10(3):211–23. pmid:12570708
* View Article
* PubMed/NCBI
* Google Scholar
57. 57. Perlin DS. Mechanisms of echinocandin antifungal drug resistance. Ann N Y Acad Sci. 2015;1354(1):1–11. pmid:26190298
* View Article
* PubMed/NCBI
* Google Scholar
58. 58. Florent M, Noël T, Ruprich-Robert G, Da Silva B, Fitton-Ouhabi V, Chastin C, et al. Nonsense and missense mutations in FCY2 and FCY1 genes are responsible for flucytosine resistance and flucytosine-fluconazole cross-resistance in clinical isolates of Candida lusitaniae. Antimicrob Agents Chemother. 2009;53(7):2982–90. pmid:19414575
* View Article
* PubMed/NCBI
* Google Scholar
59. 59. Meis JF, Chowdhary A, Rhodes JL, Fisher MC, Verweij PE. Clinical implications of globally emerging azole resistance in Aspergillus fumigatus. Philos Trans R Soc Lond B Biol Sci. 2016;371(1709):20150460. pmid:28080986
* View Article
* PubMed/NCBI
* Google Scholar
60. 60. Pristov KE, Ghannoum MA. Resistance of Candida to azoles and echinocandins worldwide. Clin Microbiol Infect. 2019;25(7):792–8. pmid:30965100
* View Article
* PubMed/NCBI
* Google Scholar
61. 61. Whaley SG, Berkow EL, Rybak JM, Nishimoto AT, Barker KS, Rogers PD. Azole antifungal resistance in Candida albicans and emerging non-albicans Candida species. Front Microbiol. 2017;7:2173. pmid:28127295
* View Article
* PubMed/NCBI
* Google Scholar
62. 62. Berkow EL, Lockhart SR. Fluconazole resistance in Candida species: a current perspective. Infect Drug Resist. 2017;10:237–45. pmid:28814889
* View Article
* PubMed/NCBI
* Google Scholar
63. 63. Forsberg K, Woodworth K, Walters M, Berkow EL, Jackson B, Chiller T, et al. Candida auris: the recent emergence of a multidrug-resistant fungal pathogen. Med Mycol. 2019;57(1):1–12. pmid:30085270
* View Article
* PubMed/NCBI
* Google Scholar
64. 64. Gintjee TJ, Donnelley MA, Thompson GR III. Aspiring antifungals: review of current antifungal pipeline developments. J Fungi (Basel). 2020;6(1):28. pmid:32106450
* View Article
* PubMed/NCBI
* Google Scholar
65. 65. CropLife International. FRAC code list 2024. 2024. Available from: https://www.frac.info/docs/default-source/publications/frac-code-list/frac-code-list-2024.pdf.
66. 66. Barh D, Tiwari S, Jain N, Ali A, Santos A, Misra A. In silico subtractive genomics for target identification in human bacterial pathogens. Drug Dev Res. 2011;72(2):162–77.
* View Article
* Google Scholar
67. 67. Ashraf B, Atiq N, Khan K, Wadood A, Uddin R. Subtractive genomics profiling for potential drug targets identification against Moraxella catarrhalis. PLoS One. 2022;17(8):e0273252. pmid:36006987
* View Article
* PubMed/NCBI
* Google Scholar
68. 68. Hosen MI, Tanmoy AM, Mahbuba D-A, Salma U, Nazim M, Islam MT, et al. Application of a subtractive genomics approach for in silico identification and characterization of novel drug targets in Mycobacterium tuberculosis F11. Interdiscip Sci. 2014;6(1):48–56. pmid:24464704
* View Article
* PubMed/NCBI
* Google Scholar
69. 69. Vilela Rodrigues TC, Jaiswal AK, de Sarom A, de Castro Oliveira L, Freire Oliveira CJ, Ghosh P, et al. Reverse vaccinology and subtractive genomics reveal new therapeutic targets against Mycoplasma pneumoniae: a causative agent of pneumonia. R Soc Open Sci. 2019;6(7):190907. pmid:31417766
* View Article
* PubMed/NCBI
* Google Scholar
70. 70. Fatoba AJ, Okpeku M, Adeleke MA. Subtractive genomics approach for identification of novel therapeutic drug targets in Mycoplasma genitalium. Pathogens. 2021;10(8):921. pmid:34451385
* View Article
* PubMed/NCBI
* Google Scholar
71. 71. Almeida PCS, Roque BS, Felice AG, Jaiswal AK, Tiwari S, Azevedo V, et al. Comparative genomics of Histoplasma capsulatum and prediction of new vaccines and drug targets. J Fungi (Basel). 2023;9(2):193. pmid:36836308
* View Article
* PubMed/NCBI
* Google Scholar
72. 72. Sliwoski G, Kothiwale S, Meiler J, Lowe EW Jr. Computational methods in drug discovery. Pharmacol Rev. 2013;66(1):334–95. pmid:24381236
* View Article
* PubMed/NCBI
* Google Scholar
73. 73. Doman TN, McGovern SL, Witherbee BJ, Kasten TP, Kurumbail R, Stallings WC, et al. Molecular docking and high-throughput screening for novel inhibitors of protein tyrosine phosphatase-1B. J Med Chem. 2002;45(11):2213–21. pmid:12014959
* View Article
* PubMed/NCBI
* Google Scholar
74. 74. Beattie SR, Krysan DJ. A unique dual-readout high-throughput screening assay to identify antifungal compounds with Aspergillus fumigatus. mSphere. 2021;6(4):e0053921. pmid:34406854
* View Article
* PubMed/NCBI
* Google Scholar
75. 75. Farha MA, Brown ED. Strategies for target identification of antimicrobial natural products. Nat Prod Rep. 2016;33(5):668–80. pmid:26806527
* View Article
* PubMed/NCBI
* Google Scholar
76. 76. Wang R, Lu Y, Wang S. Comparative evaluation of 11 scoring functions for molecular docking. J Med Chem. 2003;46(12):2287–303. pmid:12773034
* View Article
* PubMed/NCBI
* Google Scholar
77. 77. Kumari A, Singh A, Raghava M, Singh M, Tiwari P, Prakash S. An approach of computer-aided drug design (CADD) tools for in silico assessment of various inhibitors of lanosterol-14α demethylase. Mater Today Proc. https://doi.org/10.1016/j.matpr.2023.09.075
78. 78. Sabe VT, Ntombela T, Jhamba LA, Maguire GEM, Govender T, Naicker T, et al. Current trends in computer aided drug design and a highlight of drugs discovered via computational techniques: a review. Eur J Med Chem. 2021;224:113705. pmid:34303871
* View Article
* PubMed/NCBI
* Google Scholar
79. 79. Jumper J, Evans R, Pritzel A, Green T, Figurnov M, Ronneberger O, et al. Highly accurate protein structure prediction with AlphaFold. Nature. 2021;596(7873):583–9. pmid:34265844
* View Article
* PubMed/NCBI
* Google Scholar
80. 80. Zhang R, Ou H-Y, Zhang C-T. DEG: a database of essential genes. Nucleic Acids Res. 2004;32(Database issue):D271-2. pmid:14681410
* View Article
* PubMed/NCBI
* Google Scholar
81. 81. Parums DV. Editorial: The World Health Organization (WHO) fungal priority pathogens list in response to emerging fungal pathogens during the COVID-19 pandemic. Med Sci Monit. 2022;28:e939088. pmid:36453055
* View Article
* PubMed/NCBI
* Google Scholar
82. 82. Giaever G, Chu AM, Ni L, Connelly C, Riles L, Véronneau S, et al. Functional profiling of the Saccharomyces cerevisiae genome. Nature. 2002;418(6896):387–91. pmid:12140549
* View Article
* PubMed/NCBI
* Google Scholar
83. 83. Alliance of Genome Resources Consortium. Updates to the alliance of genome resources central infrastructure. Genetics. 2024;227(1):iyae049. pmid:38552170
* View Article
* PubMed/NCBI
* Google Scholar
84. 84. Altschul SF, Madden TL, Schäffer AA, Zhang J, Zhang Z, Miller W, et al. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res. 1997;25(17):3389–402. pmid:9254694
* View Article
* PubMed/NCBI
* Google Scholar
85. 85. Sievers F, Higgins DG. Clustal omega. Curr Protoc Bioinformatics. 2014;48:3.13.1-3.13.16. pmid:25501942
* View Article
* PubMed/NCBI
* Google Scholar
86. 86. Järvinen K-M, Beyer K, Vila L, Chatchatee P, Busse PJ, Sampson HA. B-cell epitopes as a screening instrument for persistent cow’s milk allergy. J Allergy Clin Immunol. 2002;110(2):293–7. pmid:12170271
* View Article
* PubMed/NCBI
* Google Scholar
87. 87. Anderson AC. The process of structure-based drug design. Chem Biol. 2003;10(9):787–97. pmid:14522049
* View Article
* PubMed/NCBI
* Google Scholar
88. 88. Wong ED, Miyasato SR, Aleksander S, Karra K, Nash RS, Skrzypek MS, et al. Saccharomyces genome database update: server architecture, pan-genome nomenclature, and external resources. Genetics. 2023;224(1):iyac191. pmid:36607068
* View Article
* PubMed/NCBI
* Google Scholar
89. 89. Arora T, Malik A. An introduction to BLAST: applications for computer-aided drug design and development. In: Sharma N, Ojha H, Raghav P, Goyal R, editors. Chemoinformatics and bioinformatics in the pharmaceutical sciences. San Diego: Academic Press; 2021, pp. 423–53.
90. 90. Tumescheit C, Firth AE, Brown K. CIAlign: a highly customisable command line tool to clean, interpret and visualise multiple sequence alignments. PeerJ. 2022;10:e12983. pmid:35310163
* View Article
* PubMed/NCBI
* Google Scholar
91. 91. Sneath PH. Relations between chemical structure and biological activity in peptides. J Theor Biol. 1966;12(2):157–95. pmid:4291386
* View Article
* PubMed/NCBI
* Google Scholar
92. 92. Bleackley MR, Hayes BM, Parisi K, Saiyed T, Traven A, Potter ID, et al. Bovine pancreatic trypsin inhibitor is a new antifungal peptide that inhibits cellular magnesium uptake. Mol Microbiol. 2014;92(6):1188–97. pmid:24750237
* View Article
* PubMed/NCBI
* Google Scholar
93. 93. Denny PW, Shams-Eldin H, Price HP, Smith DF, Schwarz RT. The protozoan inositol phosphorylceramide synthase: a novel drug target that defines a new class of sphingolipid synthase. J Biol Chem. 2006;281(38):28200–9. pmid:16861742
* View Article
* PubMed/NCBI
* Google Scholar
94. 94. Lenardon MD, Munro CA, Gow NAR. Chitin synthesis and fungal pathogenesis. Curr Opin Microbiol. 2010;13(4):416–23. pmid:20561815
* View Article
* PubMed/NCBI
* Google Scholar
95. 95. Gaughran JP, Lai MH, Kirsch DR, Silverman SJ. Nikkomycin Z is a specific inhibitor of Saccharomyces cerevisiae chitin synthase isozyme Chs3 in vitro and in vivo. J Bacteriol. 1994;176(18):5857–60. pmid:8083179
* View Article
* PubMed/NCBI
* Google Scholar
96. 96. Munro CA, Gow NAR. Chitin biosynthesis as a target for antifungals. In: Dixon GK, Copping LG, Hollomon DW, editors. Antifungal agents: discovery and mode of action. Oxford: Bios Scientific; 1995, pp. 161–71.
97. 97. Laakso JA, Raulli R, McElhaney-Feser GE, Actor P, Underiner TL, Hotovec BJ, et al. CT2108A and B: new fatty acid synthase inhibitors as antifungal agents. J Nat Prod. 2003;66(8):1041–6. pmid:12932120
* View Article
* PubMed/NCBI
* Google Scholar
98. 98. Iyer KR, Li SC, Revie NM, Lou JW, Duncan D, Fallah S, et al. Identification of triazenyl indoles as inhibitors of fungal fatty acid biosynthesis with broad-spectrum activity. Cell Chem Biol. 2023;30(7):795-810.e8. pmid:37369212
* View Article
* PubMed/NCBI
* Google Scholar
99. 99. Johansson P, Wiltschi B, Kumari P, Kessler B, Vonrhein C, Vonck J, et al. Inhibition of the fungal fatty acid synthase type I multienzyme complex. Proc Natl Acad Sci U S A. 2008;105(35):12803–8. pmid:18725634
* View Article
* PubMed/NCBI
* Google Scholar
100. 100. Rodaki A, Young T, Brown AJP. Effects of depleting the essential central metabolic enzyme fructose-1,6-bisphosphate aldolase on the growth and viability of Candida albicans: implications for antifungal drug target discovery. Eukaryot Cell. 2006;5(8):1371–7. pmid:16896220
* View Article
* PubMed/NCBI
* Google Scholar
101. 101. Viana R, Couceiro D, Carreiro T, Dias O, Rocha I, Teixeira MC. A genome-scale metabolic model for the human pathogen Candida parapsilosis and early identification of putative novel antifungal drug targets. Genes (Basel). 2022;13(2):303. pmid:35205348
* View Article
* PubMed/NCBI
* Google Scholar
102. 102. Bates S. Trl1 as an antifungal target. 2007.
103. 103. Banerjee A, Ghosh S, Goldgur Y, Shuman S. Structure and two-metal mechanism of fungal tRNA ligase. Nucleic Acids Res. 2019;47(3):1428–39. pmid:30590734
* View Article
* PubMed/NCBI
* Google Scholar
104. 104. Ahammed KS, van Hoof A. Fungi of the order mucorales express a “sealing-only” tRNA ligase. RNA. 2024;30(4):354–66. pmid:38307611
* View Article
* PubMed/NCBI
* Google Scholar
105. 105. Sturtevant J. Translation elongation-3-like factors: are they rational antifungal targets? Expert Opin Ther Targets. 2002;6(5):545–53. pmid:12387678
* View Article
* PubMed/NCBI
* Google Scholar
106. 106. Andersen CBF, Becker T, Blau M, Anand M, Halic M, Balar B, et al. Structure of eEF3 and the mechanism of transfer RNA release from the E-site. Nature. 2006;443(7112):663–8. pmid:16929303
* View Article
* PubMed/NCBI
* Google Scholar
107. 107. UniProt Consortium. UniProt: the Universal Protein Knowledgebase in 2023. Nucleic Acids Res. 2023;51(D1):D523–31. pmid:36408920
* View Article
* PubMed/NCBI
* Google Scholar
108. 108. Fischer M, Joppe M, Mulinacci B, Vollrath R, Konstantinidis K, Kötter P, et al. Analysis of the co-translational assembly of the fungal fatty acid synthase (FAS). Sci Rep. 2020;10(1):895. pmid:31964902
* View Article
* PubMed/NCBI
* Google Scholar
109. 109. Schweizer M, Roberts LM, Höltke HJ, Takabayashi K, Höllerer E, Hoffmann B, et al. The pentafunctional FAS1 gene of yeast: its nucleotide sequence and order of the catalytic domains. Mol Gen Genet. 1986;203(3):479–86. pmid:3528750
* View Article
* PubMed/NCBI
* Google Scholar
110. 110. Pruitt KD, Tatusova T, Maglott DR. NCBI reference sequences (RefSeq): a curated non-redundant sequence database of genomes, transcripts and proteins. Nucleic Acids Res. 2007;35(Database issue):D61-5. pmid:17130148
* View Article
* PubMed/NCBI
* Google Scholar
111. 111. Gow NA, Robbins PW, Lester JW, Brown AJ, Fonzi WA, Chapman T, et al. A hyphal-specific chitin synthase gene (CHS2) is not essential for growth, dimorphism, or virulence of Candida albicans. Proc Natl Acad Sci U S A. 1994;91(13):6216–20. pmid:8016141
* View Article
* PubMed/NCBI
* Google Scholar
112. 112. Reza MH, Shah H, Manjrekar J, Chattoo BB. Magnesium uptake by CorA transporters is essential for growth, development and infection in the rice blast fungus Magnaporthe oryzae. PLoS One. 2016;11(7):e0159244. pmid:27416318
* View Article
* PubMed/NCBI
* Google Scholar
113. 113. Estrada A, Wright DL, Anderson AC. Antibacterial antifolates: from development through resistance to the next generation. Cold Spring Harb Perspect Med. 2016;6(8):a028324. pmid:27352799
* View Article
* PubMed/NCBI
* Google Scholar
114. 114. Koshy R, Chen T. Combination therapy with trimethoprim-sulfamethoxazole and caspofungin in a case of severe pneumocystis pneumonia. IDCases. 2019;15:e00496. pmid:30828545
* View Article
* PubMed/NCBI
* Google Scholar
115. 115. Nagiec MM, Nagiec EE, Baltisberger JA, Wells GB, Lester RL, Dickson RC. Sphingolipid synthesis as a target for antifungal drugs. Complementation of the inositol phosphorylceramide synthase defect in a mutant strain of Saccharomyces cerevisiae by the AUR1 gene. J Biol Chem. 1997;272(15):9809–17. pmid:9092515
* View Article
* PubMed/NCBI
* Google Scholar
116. 116. Remus BS, Schwer B, Shuman S. Characterization of the tRNA ligases of pathogenic fungi Aspergillus fumigatus and Coccidioides immitis. RNA. 2016;22(10):1500–9. pmid:27492257
* View Article
* PubMed/NCBI
* Google Scholar
117. 117. Maldonado G, García A, Herrero S, Castaño I, Altmann M, Fischer R, et al. The gene YEF3 function encoding translation elongation factor eEF3 is partially conserved across fungi. Front Microbiol. 2024;15:1438900. pmid:39247690
* View Article
* PubMed/NCBI
* Google Scholar
118. 118. Triana-Alonso FJ, Chakraburtty K, Nierhaus KH. The elongation factor 3 unique in higher fungi and essential for protein biosynthesis is an E site factor. J Biol Chem. 1995;270(35):20473–8. pmid:7657623
* View Article
* PubMed/NCBI
* Google Scholar
119. 119. Schneider E, Hunke S. ATP-binding-cassette (ABC) transport systems: functional and structural aspects of the ATP-hydrolyzing subunits/domains. FEMS Microbiol Rev. 1998;22(1):1–20. pmid:9640644
* View Article
* PubMed/NCBI
* Google Scholar
120. 120. Su H, Han L, Huang X. Potential targets for the development of new antifungal drugs. J Antibiot (Tokyo). 2018;71(12):978–91. pmid:30242283
* View Article
* PubMed/NCBI
* Google Scholar
121. 121. Amorim Franco TM, Blanchard JS. Bacterial branched-chain amino acid biosynthesis: structures, mechanisms, and drugability. Biochemistry. 2017;56(44):5849–65. pmid:28977745
* View Article
* PubMed/NCBI
* Google Scholar
122. 122. Steyer JT, Todd RB. Branched-chain amino acid biosynthesis in fungi. Essays Biochem. 2023;67(5):865–76. pmid:37455545
* View Article
* PubMed/NCBI
* Google Scholar
123. 123. Neinast M, Murashige D, Arany Z. Branched chain amino acids. Annu Rev Physiol. 2019;81:139–64. pmid:30485760
* View Article
* PubMed/NCBI
* Google Scholar
124. 124. Zhang P, MacTavish BS, Yang G, Chen M, Roh J, Newsome KR, et al. Cyanobacterial dihydroxyacid dehydratases are a promising growth inhibition target. ACS Chem Biol. 2020;15(8):2281–8. pmid:32786290
* View Article
* PubMed/NCBI
* Google Scholar
125. 125. Demuyser L, Palmans I, Vandecruys P, Van Dijck P. Molecular elucidation of riboflavin production and regulation in Candida albicans, toward a novel antifungal drug target. mSphere. 2020;5(4):e00714-20. pmid:32759338
* View Article
* PubMed/NCBI
* Google Scholar
126. 126. Wagner R, Straub ML, Souciet JL, Potier S, de Montigny J. New plasmid system to select for Saccharomyces cerevisiae purine-cytosine permease affinity mutants. J Bacteriol. 2001;183(14):4386–8. pmid:11418581
* View Article
* PubMed/NCBI
* Google Scholar
127. 127. Paluszynski JP, Klassen R, Rohe M, Meinhardt F. Various cytosine/adenine permease homologues are involved in the toxicity of 5-fluorocytosine in Saccharomyces cerevisiae. Yeast. 2006;23(9):707–15. pmid:16845689
* View Article
* PubMed/NCBI
* Google Scholar
128. 128. Gruber S, Zeilinger S. The transcription factor Ste12 mediates the regulatory role of the Tmk1 MAP kinase in mycoparasitism and vegetative hyphal fusion in the filamentous fungus Trichoderma atroviride. PLoS One. 2014;9(10):e111636. pmid:25356841
* View Article
* PubMed/NCBI
* Google Scholar
129. 129. Vandermeulen MD, Cullen PJ. Ecological inducers of the yeast filamentous growth pathway reveal environment-dependent roles for pathway components. mSphere. 2023;8(5):e0028423. pmid:37732804
* View Article
* PubMed/NCBI
* Google Scholar
130. 130. Benhamou RI, Bibi M, Steinbuch KB, Engel H, Levin M, Roichman Y, et al. Real-Time imaging of the azole class of antifungal drugs in live Candida cells. ACS Chem Biol. 2017;12(7):1769–77. pmid:28472585
* View Article
* PubMed/NCBI
* Google Scholar
131. 131. Hay RJ. Antifungal drugs on the horizon. J Am Acad Dermatol. 1994;31(3 Pt 2):S82-6. pmid:8077515
* View Article
* PubMed/NCBI
* Google Scholar
132. 132. Bowman SM, Free SJ. The structure and synthesis of the fungal cell wall. Bioessays. 2006;28(8):799–808. pmid:16927300
* View Article
* PubMed/NCBI
* Google Scholar
133. 133. Färber PM, Arscott LD, Williams CH Jr, Becker K, Schirmer RH. Recombinant Plasmodium falciparum glutathione reductase is inhibited by the antimalarial dye methylene blue. FEBS Lett. 1998;422(3):311–4. pmid:9498806
* View Article
* PubMed/NCBI
* Google Scholar
134. 134. Monk BC, Sagatova AA, Hosseini P, Ruma YN, Wilson RK, Keniya MV. Fungal Lanosterol 14α-demethylase: a target for next-generation antifungal design. Biochim Biophys Acta Proteins Proteom. 2020;1868(3):140206. pmid:30851431
* View Article
* PubMed/NCBI
* Google Scholar
135. 135. Martin FM, Uroz S, Barker DG. Ancestral alliances: plant mutualistic symbioses with fungi and bacteria. Science. 2017;356(6340):eaad4501. pmid:28546156
* View Article
* PubMed/NCBI
* Google Scholar
136. 136. Bastías DA, Johnson LJ, Card SD. Symbiotic bacteria of plant-associated fungi: friends or foes? Curr Opin Plant Biol. 2020;56:1–8. pmid:31786411
* View Article
* PubMed/NCBI
* Google Scholar
137. 137. van den Bosch TJM, Welte CU. Detoxifying symbionts in agriculturally important pest insects. Microb Biotechnol. 2017;10(3):531–40. pmid:27943632
* View Article
* PubMed/NCBI
* Google Scholar
138. 138. Goodall ECA, Robinson A, Johnston IG, Jabbari S, Turner KA, Cunningham AF, et al. The essential genome of Escherichia coli K-12. mBio. 2018;9(1):e02096-17. pmid:29463657
* View Article
* PubMed/NCBI
* Google Scholar
139. 139. Stanzione F, Giangreco I, Cole JC. Use of molecular docking computational tools in drug discovery. Prog Med Chem. 2021;60:273–343. pmid:34147204
* View Article
* PubMed/NCBI
* Google Scholar
140. 140. Berman HM, Westbrook J, Feng Z, Gilliland G, Bhat TN, Weissig H, et al. The protein data bank. Nucleic Acids Res. 2000;28(1):235–42. pmid:10592235
* View Article
* PubMed/NCBI
* Google Scholar
141. 141. Holm L. Dali server: structural unification of protein families. Nucleic Acids Res. 2022;50(W1):W210–5. pmid:35610055
* View Article
* PubMed/NCBI
* Google Scholar
142. 142. van Kempen M, Kim SS, Tumescheit C, Mirdita M, Lee J, Gilchrist CLM, et al. Fast and accurate protein structure search with Foldseek. Nat Biotechnol. 2024;42(2):243–6. pmid:37156916
* View Article
* PubMed/NCBI
* Google Scholar
143. 143. Liu YJ, Hall BD. Body plan evolution of ascomycetes, as inferred from an RNA polymerase II phylogeny. Proc Natl Acad Sci U S A. 2004;101(13):4507–12. pmid:15070748
* View Article
* PubMed/NCBI
* Google Scholar
144. 144. Hoffman CS, Wood V, Fantes PA. An ancient yeast for young geneticists: a primer on the Schizosaccharomyces pombe model system. Genetics. 2015;201(2):403–23. pmid:26447128
* View Article
* PubMed/NCBI
* Google Scholar
145. 145. Kachroo AH, Laurent JM, Yellman CM, Meyer AG, Wilke CO, Marcotte EM. Evolution. Systematic humanization of yeast genes reveals conserved functions and genetic modularity. Science. 2015;348(6237):921–5. pmid:25999509
* View Article
* PubMed/NCBI
* Google Scholar
146. 146. Cockrell A, McCormick SP, Moore MJ, Chakrabarti M, Lindahl PA. Mössbauer, EPR, and modeling study of iron trafficking and regulation in Δccc1 and CCC1-up Saccharomyces cerevisiae. Biochemistry. 2014;53(18):2926–40. pmid:24785783
* View Article
* PubMed/NCBI
* Google Scholar
147. 147. Sorribes-Dauden R, Peris D, Martínez-Pastor MT, Puig S. Structure and function of the vacuolar Ccc1/VIT1 family of iron transporters and its regulation in fungi. Comput Struct Biotechnol J. 2020;18:3712–22. pmid:33304466
* View Article
* PubMed/NCBI
* Google Scholar
Citation: Condon DE, Schroeder BK, Rowley PA, Ytreberg FM (2025) Discovery of novel targets for important human and plant fungal pathogens via an automated computational pipeline HitList. PLoS One 20(6): e0323991. https://doi.org/10.1371/journal.pone.0323991
About the Authors:
David E. Condon
Roles: Formal analysis, Methodology, Visualization, Writing – original draft, Writing – review & editing
Affiliation: Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, United States of America
Brenda K. Schroeder
Roles: Conceptualization, Writing – review & editing
Affiliations: Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, United States of America, Department of Software Engineering, College of Computer and Information Sciences, King Saud University, Riyadh 11543, Saudi Arabia
Paul A. Rowley
Roles: Conceptualization, Visualization, Writing – review & editing
Affiliations: Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, United States of America, Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America, Institute of Ocean and Earth Sciences, Universiti Malaya, C308, Institute of Advanced Studies Building, Kuala Lumpur 50603, Malaysia
F. Marty Ytreberg
Roles: Conceptualization, Funding acquisition, Investigation, Writing – review & editing
E-mail: [email protected]
Affiliations: Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, United States of America, Department of Physics, University of Idaho, Moscow, Idaho, United States of America
ORICD: https://orcid.org/0000-0001-7439-8224
[/RAW_REF_TEXT]
[/RAW_REF_TEXT]
[/RAW_REF_TEXT]
[/RAW_REF_TEXT]
[/RAW_REF_TEXT]
[/RAW_REF_TEXT]
[/RAW_REF_TEXT]
[/RAW_REF_TEXT]
[/RAW_REF_TEXT]
1. Dickman MB, de Figueiredo P. Comparative pathobiology of fungal pathogens of plants and animals. PLoS Pathog. 2011;7(12):e1002324. pmid:22194681
2. Sexton AC, Howlett BJ. Parallels in fungal pathogenesis on plant and animal hosts. Eukaryot Cell. 2006;5(12):1941–9. pmid:17041185
3. Heitman J. Microbial pathogens in the fungal kingdom. Fungal Biol Rev. 2011;25(1):48–60. pmid:21528015
4. van Agtmaal M, Straathof A, Termorshuizen A, Teurlincx S, Hundscheid M, Ruyters S. Exploring the reservoir of potential fungal plant pathogens in agricultural soil. Appl Soil Ecol. 2017;121:152–60.
5. Ajala A, Asipita OH, Michael AT, Tajudeen MT, Abdulganiyyu IA, Ramu R. Therapeutic exploration potential of adenosine receptor antagonists through pharmacophore ligand-based modelling and pharmacokinetics studies against Parkinson disease. In Silico Pharmacol. 2025;13(1):17. pmid:39872470
6. Hahn M. The rising threat of fungicide resistance in plant pathogenic fungi: Botrytis as a case study. J Chem Biol. 2014;7(4):133–41. pmid:25320647
7. Sun S, Hoy MJ, Heitman J. Fungal pathogens. Curr Biol. 2020;30(19):R1163–9. pmid:33022261
8. Strange RN, Scott PR. Plant disease: a threat to global food security. Annu Rev Phytopathol. 2005;43:83–116. pmid:16078878
9. Bebber DP, Gurr SJ. Crop-destroying fungal and oomycete pathogens challenge food security. Fungal Genet Biol. 2015;74:62–4. pmid:25459533
10. Dean R, Van Kan JAL, Pretorius ZA, Hammond-Kosack KE, Di Pietro A, Spanu PD, et al. The Top 10 fungal pathogens in molecular plant pathology. Mol Plant Pathol. 2012;13(4):414–30. pmid:22471698
11. Luo S, Du H, Kebede H, Liu Y, Xing F. Contamination status of major mycotoxins in agricultural product and food stuff in Europe. Food Control. 2021;127:108120.
12. World Health Organization. Selected mycotoxins: ochratoxins, trichothecenes, ergot. World Health Organization; 1990.
13. Richard JL. Some major mycotoxins and their mycotoxicoses—an overview. Int J Food Microbiol. 2007;119(1–2):3–10. pmid:17719115
14. Kainz K, Bauer MA, Madeo F, Carmona-Gutierrez D. Fungal infections in humans: the silent crisis. Microb Cell. 2020;7(6):143–5. pmid:32548176
15. World Health Organization. WHO fungal priority pathogens list to guide research, development and public health action. World Health Organization; 2022. Available from: https://www.who.int/publications/i/item/9789240060241
16. Wiederhold NP. Antifungal resistance: current trends and future strategies to combat. Infect Drug Resist. 2017;10:249–59. pmid:28919789
17. Lucas JA, Hawkins NJ, Fraaije BA. Chapter 2: The evolution of fungicide resistance. In: Advances in applied microbiology, vol. 90. Academic Press; 2015, pp. 29–92. Available from: https://www.sciencedirect.com/science/article/pii/S0065216414000021
18. Kühbacher A, Burger-Kentischer A, Rupp S. Interaction of Candida species with the skin. Microorganisms. 2017;5(2):32. pmid:28590443
19. Richardson M, Lass-Flörl C. Changing epidemiology of systemic fungal infections. Clin Microbiol Infect. 2008;14(Suppl 4):5–24. pmid:18430126
20. Ashbee H, Gilleece M. Chapter 38: Pharmacogenomics of antifungal agents. In: Padmanabhan S, editor. Handbook of pharmacogenomics and stratified medicine. San Diego: Academic Press; 2014, pp. 879–900.
21. Weimer KED, Smith PB, Puia-Dumitrescu M, Aleem S. Invasive fungal infections in neonates: a review. Pediatr Res. 2022;91(2):404–12. pmid:34880444
22. Bitar D, Lortholary O, Le Strat Y, Nicolau J, Coignard B, Tattevin P, et al. Population-based analysis of invasive fungal infections, France, 2001–2010. Emerg Infect Dis. 2014;20(7):1149.
23. Leventakos K, Lewis RE, Kontoyiannis DP. Fungal infections in leukemia patients: how do we prevent and treat them?. Clin Infect Dis. 2010;50(3):405–15. pmid:20047485
24. Pappas PG, Alexander BD, Andes DR, Hadley S, Kauffman CA, Freifeld A, et al. Invasive fungal infections among organ transplant recipients: results of the Transplant-Associated Infection Surveillance Network (TRANSNET). Clin Infect Dis. 2010;50(8):1101–11. pmid:20218876
25. Rayens E, Norris KA. Prevalence and healthcare burden of fungal infections in the United States, 2018. Open Forum Infect Dis. 2022;9(1):ofab593. pmid:35036461
26. Benedict K, Jackson BR, Chiller T, Beer KD. Estimation of direct healthcare costs of fungal diseases in the United States. Clin Infect Dis. 2019;68(11):1791–7. pmid:30204844
27. Benedict K, Whitham HK, Jackson BR. Economic burden of fungal diseases in the United States. Open Forum Infect Dis. 2022;9(4):ofac097. pmid:35350173
28. Barac A, Cevik M, Colovic N, Lekovic D, Stevanovic G, Micic J, et al. Investigation of a healthcare-associated Candida tropicalis candidiasis cluster in a haematology unit and a systematic review of nosocomial outbreaks. Mycoses. 2020;63(4):326–33. pmid:31930592
29. Achkar JM, Fries BC. Candida infections of the genitourinary tract. Clin Microbiol Rev. 2010;23(2):253–73. pmid:20375352
30. Hoenigl M, Seidel D, Sprute R, Cunha C, Oliverio M, Goldman GH, et al. COVID-19-associated fungal infections. Nat Microbiol. 2022;7(8):1127–40. pmid:35918423
31. Savary S, Willocquet L, Pethybridge SJ, Esker P, McRoberts N, Nelson A. The global burden of pathogens and pests on major food crops. Nat Ecol Evol. 2019;3(3):430–9. pmid:30718852
32. Jørgensen LN, van den Bosch F, Oliver RP, Heick TM, Paveley ND. Targeting fungicide inputs according to need. Annu Rev Phytopathol. 2017;55:181–203. pmid:28525306
33. Brefort T, Doehlemann G, Mendoza-Mendoza A, Reissmann S, Djamei A, Kahmann R. Ustilago maydis as a pathogen. Annu Rev Phytopathol. 2009;47:423–45. pmid:19400641
34. Fisher MC, Henk DA, Briggs CJ, Brownstein JS, Madoff LC, McCraw SL, et al. Emerging fungal threats to animal, plant and ecosystem health. Nature. 2012;484(7393):186–94. pmid:22498624
35. Diao YZ, Zhang C, Lin D, Liu XL. First report of Colletotrichum truncatum causing anthracnose of tomato in China. Plant Dis. 2014;98(5):687. pmid:30708530
36. Wyand RA, Brown JKM. Genetic and forma specialis diversity in Blumeria graminis of cereals and its implications for host-pathogen co-evolution. Mol Plant Pathol. 2003;4(3):187–98. pmid:20569378
37. Santini A, Liebhold A, Migliorini D, Woodward S. Tracing the role of human civilization in the globalization of plant pathogens. ISME J. 2018;12(3):647–52. pmid:29330537
38. Stukenbrock EH, Banke S, McDonald BA. Global migration patterns in the fungal wheat pathogen Phaeosphaeria nodorum. Mol Ecol. 2006;15(10):2895–904. pmid:16911209
39. Singh RP, Hodson DP, Huerta-Espino J, Jin Y, Bhavani S, Njau P, et al. The emergence of Ug99 races of the stem rust fungus is a threat to world wheat production. Annu Rev Phytopathol. 2011;49:465–81. pmid:21568701
40. Lacey J. Pre- and post-harvest ecology of fungi causing spoilage of foods and other stored products. Soc Appl Bacteriol Symp Ser. 1989;18:11S–25S. pmid:2508232
41. Azevedo M-M, Faria-Ramos I, Cruz LC, Pina-Vaz C, Rodrigues AG. Genesis of azole antifungal resistance from agriculture to clinical settings. J Agric Food Chem. 2015;63(34):7463–8. pmid:26289797
42. Kang SE, Sumabat LG, Melie T, Mangum B, Momany M, Brewer MT. Evidence for the agricultural origin of resistance to multiple antimicrobials in Aspergillus fumigatus, a fungal pathogen of humans. G3 (Bethesda). 2022;12(2):jkab427. pmid:34897421
43. Nicola AM, Albuquerque P, Paes HC, Fernandes L, Costa FF, Kioshima ES, et al. Antifungal drugs: new insights in research & development. Pharmacol Ther. 2019;195:21–38. pmid:30347212
44. Fernandes CM, Dasilva D, Haranahalli K, McCarthy JB, Mallamo J, Ojima I, et al. The future of antifungal drug therapy: novel compounds and targets. Antimicrob Agents Chemother. 2021;65(2):e01719-20. pmid:33229427
45. Feofilova EP. The kingdom fungi: heterogeneity of physiological and biochemical properties and relationships with plants, animals, and prokaryotes (Review). Appl Biochem Microbiol. 2001;37:124–37.
46. Fromtling RA. Overview of medically important antifungal azole derivatives. Clin Microbiol Rev. 1988;1(2):187–217. pmid:3069196
47. Sheehan DJ, Hitchcock CA, Sibley CM. Current and emerging azole antifungal agents. Clin Microbiol Rev. 1999;12(1):40–79. pmid:9880474
48. Mercer EI. Morpholine antifungals and their mode of action. Biochem Soc Trans. 1991;19(3):788–93. pmid:1783217
49. Denning DW. Echinocandin antifungal drugs. Lancet. 2003;362(9390):1142–51. pmid:14550704
50. Szymański M, Chmielewska S, Czyżewska U, Malinowska M, Tylicki A. Echinocandins—structure, mechanism of action and use in antifungal therapy. J Enzyme Inhib Med Chem. 2022;37(1):876–94. pmid:35296203
51. Kaya Y, Dokuzeylül B, Bakırel U, Or ME. Antifungal resistance and clinical significance in small animals. Ger J Vet Res. 2022;2:28–36.
52. Eschenauer G, Depestel DD, Carver PL. Comparison of echinocandin antifungals. Ther Clin Risk Manag. 2007;3(1):71–97. pmid:18360617
53. Fisher MC, Alastruey-Izquierdo A, Berman J, Bicanic T, Bignell EM, Bowyer P, et al. Tackling the emerging threat of antifungal resistance to human health. Nat Rev Microbiol. 2022;20(9):557–71. pmid:35352028
54. Benitez LL, Carver PL. Adverse effects associated with long-term administration of azole antifungal agents. Drugs. 2019;79(8):833–53. pmid:31093949
55. Mohr J, Johnson M, Cooper T, Lewis JS, Ostrosky-Zeichner L. Current options in antifungal pharmacotherapy. Pharmacotherapy. 2008;28(5):614–45. pmid:18447660
56. Zotchev SB. Polyene macrolide antibiotics and their applications in human therapy. Curr Med Chem. 2003;10(3):211–23. pmid:12570708
57. Perlin DS. Mechanisms of echinocandin antifungal drug resistance. Ann N Y Acad Sci. 2015;1354(1):1–11. pmid:26190298
58. Florent M, Noël T, Ruprich-Robert G, Da Silva B, Fitton-Ouhabi V, Chastin C, et al. Nonsense and missense mutations in FCY2 and FCY1 genes are responsible for flucytosine resistance and flucytosine-fluconazole cross-resistance in clinical isolates of Candida lusitaniae. Antimicrob Agents Chemother. 2009;53(7):2982–90. pmid:19414575
59. Meis JF, Chowdhary A, Rhodes JL, Fisher MC, Verweij PE. Clinical implications of globally emerging azole resistance in Aspergillus fumigatus. Philos Trans R Soc Lond B Biol Sci. 2016;371(1709):20150460. pmid:28080986
60. Pristov KE, Ghannoum MA. Resistance of Candida to azoles and echinocandins worldwide. Clin Microbiol Infect. 2019;25(7):792–8. pmid:30965100
61. Whaley SG, Berkow EL, Rybak JM, Nishimoto AT, Barker KS, Rogers PD. Azole antifungal resistance in Candida albicans and emerging non-albicans Candida species. Front Microbiol. 2017;7:2173. pmid:28127295
62. Berkow EL, Lockhart SR. Fluconazole resistance in Candida species: a current perspective. Infect Drug Resist. 2017;10:237–45. pmid:28814889
63. Forsberg K, Woodworth K, Walters M, Berkow EL, Jackson B, Chiller T, et al. Candida auris: the recent emergence of a multidrug-resistant fungal pathogen. Med Mycol. 2019;57(1):1–12. pmid:30085270
64. Gintjee TJ, Donnelley MA, Thompson GR III. Aspiring antifungals: review of current antifungal pipeline developments. J Fungi (Basel). 2020;6(1):28. pmid:32106450
65. CropLife International. FRAC code list 2024. 2024. Available from: https://www.frac.info/docs/default-source/publications/frac-code-list/frac-code-list-2024.pdf.
66. Barh D, Tiwari S, Jain N, Ali A, Santos A, Misra A. In silico subtractive genomics for target identification in human bacterial pathogens. Drug Dev Res. 2011;72(2):162–77.
67. Ashraf B, Atiq N, Khan K, Wadood A, Uddin R. Subtractive genomics profiling for potential drug targets identification against Moraxella catarrhalis. PLoS One. 2022;17(8):e0273252. pmid:36006987
68. Hosen MI, Tanmoy AM, Mahbuba D-A, Salma U, Nazim M, Islam MT, et al. Application of a subtractive genomics approach for in silico identification and characterization of novel drug targets in Mycobacterium tuberculosis F11. Interdiscip Sci. 2014;6(1):48–56. pmid:24464704
69. Vilela Rodrigues TC, Jaiswal AK, de Sarom A, de Castro Oliveira L, Freire Oliveira CJ, Ghosh P, et al. Reverse vaccinology and subtractive genomics reveal new therapeutic targets against Mycoplasma pneumoniae: a causative agent of pneumonia. R Soc Open Sci. 2019;6(7):190907. pmid:31417766
70. Fatoba AJ, Okpeku M, Adeleke MA. Subtractive genomics approach for identification of novel therapeutic drug targets in Mycoplasma genitalium. Pathogens. 2021;10(8):921. pmid:34451385
71. Almeida PCS, Roque BS, Felice AG, Jaiswal AK, Tiwari S, Azevedo V, et al. Comparative genomics of Histoplasma capsulatum and prediction of new vaccines and drug targets. J Fungi (Basel). 2023;9(2):193. pmid:36836308
72. Sliwoski G, Kothiwale S, Meiler J, Lowe EW Jr. Computational methods in drug discovery. Pharmacol Rev. 2013;66(1):334–95. pmid:24381236
73. Doman TN, McGovern SL, Witherbee BJ, Kasten TP, Kurumbail R, Stallings WC, et al. Molecular docking and high-throughput screening for novel inhibitors of protein tyrosine phosphatase-1B. J Med Chem. 2002;45(11):2213–21. pmid:12014959
74. Beattie SR, Krysan DJ. A unique dual-readout high-throughput screening assay to identify antifungal compounds with Aspergillus fumigatus. mSphere. 2021;6(4):e0053921. pmid:34406854
75. Farha MA, Brown ED. Strategies for target identification of antimicrobial natural products. Nat Prod Rep. 2016;33(5):668–80. pmid:26806527
76. Wang R, Lu Y, Wang S. Comparative evaluation of 11 scoring functions for molecular docking. J Med Chem. 2003;46(12):2287–303. pmid:12773034
77. Kumari A, Singh A, Raghava M, Singh M, Tiwari P, Prakash S. An approach of computer-aided drug design (CADD) tools for in silico assessment of various inhibitors of lanosterol-14α demethylase. Mater Today Proc. https://doi.org/10.1016/j.matpr.2023.09.075
78. Sabe VT, Ntombela T, Jhamba LA, Maguire GEM, Govender T, Naicker T, et al. Current trends in computer aided drug design and a highlight of drugs discovered via computational techniques: a review. Eur J Med Chem. 2021;224:113705. pmid:34303871
79. Jumper J, Evans R, Pritzel A, Green T, Figurnov M, Ronneberger O, et al. Highly accurate protein structure prediction with AlphaFold. Nature. 2021;596(7873):583–9. pmid:34265844
80. Zhang R, Ou H-Y, Zhang C-T. DEG: a database of essential genes. Nucleic Acids Res. 2004;32(Database issue):D271-2. pmid:14681410
81. Parums DV. Editorial: The World Health Organization (WHO) fungal priority pathogens list in response to emerging fungal pathogens during the COVID-19 pandemic. Med Sci Monit. 2022;28:e939088. pmid:36453055
82. Giaever G, Chu AM, Ni L, Connelly C, Riles L, Véronneau S, et al. Functional profiling of the Saccharomyces cerevisiae genome. Nature. 2002;418(6896):387–91. pmid:12140549
83. Alliance of Genome Resources Consortium. Updates to the alliance of genome resources central infrastructure. Genetics. 2024;227(1):iyae049. pmid:38552170
84. Altschul SF, Madden TL, Schäffer AA, Zhang J, Zhang Z, Miller W, et al. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res. 1997;25(17):3389–402. pmid:9254694
85. Sievers F, Higgins DG. Clustal omega. Curr Protoc Bioinformatics. 2014;48:3.13.1-3.13.16. pmid:25501942
86. Järvinen K-M, Beyer K, Vila L, Chatchatee P, Busse PJ, Sampson HA. B-cell epitopes as a screening instrument for persistent cow’s milk allergy. J Allergy Clin Immunol. 2002;110(2):293–7. pmid:12170271
87. Anderson AC. The process of structure-based drug design. Chem Biol. 2003;10(9):787–97. pmid:14522049
88. Wong ED, Miyasato SR, Aleksander S, Karra K, Nash RS, Skrzypek MS, et al. Saccharomyces genome database update: server architecture, pan-genome nomenclature, and external resources. Genetics. 2023;224(1):iyac191. pmid:36607068
89. Arora T, Malik A. An introduction to BLAST: applications for computer-aided drug design and development. In: Sharma N, Ojha H, Raghav P, Goyal R, editors. Chemoinformatics and bioinformatics in the pharmaceutical sciences. San Diego: Academic Press; 2021, pp. 423–53.
90. Tumescheit C, Firth AE, Brown K. CIAlign: a highly customisable command line tool to clean, interpret and visualise multiple sequence alignments. PeerJ. 2022;10:e12983. pmid:35310163
91. Sneath PH. Relations between chemical structure and biological activity in peptides. J Theor Biol. 1966;12(2):157–95. pmid:4291386
92. Bleackley MR, Hayes BM, Parisi K, Saiyed T, Traven A, Potter ID, et al. Bovine pancreatic trypsin inhibitor is a new antifungal peptide that inhibits cellular magnesium uptake. Mol Microbiol. 2014;92(6):1188–97. pmid:24750237
93. Denny PW, Shams-Eldin H, Price HP, Smith DF, Schwarz RT. The protozoan inositol phosphorylceramide synthase: a novel drug target that defines a new class of sphingolipid synthase. J Biol Chem. 2006;281(38):28200–9. pmid:16861742
94. Lenardon MD, Munro CA, Gow NAR. Chitin synthesis and fungal pathogenesis. Curr Opin Microbiol. 2010;13(4):416–23. pmid:20561815
95. Gaughran JP, Lai MH, Kirsch DR, Silverman SJ. Nikkomycin Z is a specific inhibitor of Saccharomyces cerevisiae chitin synthase isozyme Chs3 in vitro and in vivo. J Bacteriol. 1994;176(18):5857–60. pmid:8083179
96. Munro CA, Gow NAR. Chitin biosynthesis as a target for antifungals. In: Dixon GK, Copping LG, Hollomon DW, editors. Antifungal agents: discovery and mode of action. Oxford: Bios Scientific; 1995, pp. 161–71.
97. Laakso JA, Raulli R, McElhaney-Feser GE, Actor P, Underiner TL, Hotovec BJ, et al. CT2108A and B: new fatty acid synthase inhibitors as antifungal agents. J Nat Prod. 2003;66(8):1041–6. pmid:12932120
98. Iyer KR, Li SC, Revie NM, Lou JW, Duncan D, Fallah S, et al. Identification of triazenyl indoles as inhibitors of fungal fatty acid biosynthesis with broad-spectrum activity. Cell Chem Biol. 2023;30(7):795-810.e8. pmid:37369212
99. Johansson P, Wiltschi B, Kumari P, Kessler B, Vonrhein C, Vonck J, et al. Inhibition of the fungal fatty acid synthase type I multienzyme complex. Proc Natl Acad Sci U S A. 2008;105(35):12803–8. pmid:18725634
100. Rodaki A, Young T, Brown AJP. Effects of depleting the essential central metabolic enzyme fructose-1,6-bisphosphate aldolase on the growth and viability of Candida albicans: implications for antifungal drug target discovery. Eukaryot Cell. 2006;5(8):1371–7. pmid:16896220
101. Viana R, Couceiro D, Carreiro T, Dias O, Rocha I, Teixeira MC. A genome-scale metabolic model for the human pathogen Candida parapsilosis and early identification of putative novel antifungal drug targets. Genes (Basel). 2022;13(2):303. pmid:35205348
102. Bates S. Trl1 as an antifungal target. 2007.
103. Banerjee A, Ghosh S, Goldgur Y, Shuman S. Structure and two-metal mechanism of fungal tRNA ligase. Nucleic Acids Res. 2019;47(3):1428–39. pmid:30590734
104. Ahammed KS, van Hoof A. Fungi of the order mucorales express a “sealing-only” tRNA ligase. RNA. 2024;30(4):354–66. pmid:38307611
105. Sturtevant J. Translation elongation-3-like factors: are they rational antifungal targets? Expert Opin Ther Targets. 2002;6(5):545–53. pmid:12387678
106. Andersen CBF, Becker T, Blau M, Anand M, Halic M, Balar B, et al. Structure of eEF3 and the mechanism of transfer RNA release from the E-site. Nature. 2006;443(7112):663–8. pmid:16929303
107. UniProt Consortium. UniProt: the Universal Protein Knowledgebase in 2023. Nucleic Acids Res. 2023;51(D1):D523–31. pmid:36408920
108. Fischer M, Joppe M, Mulinacci B, Vollrath R, Konstantinidis K, Kötter P, et al. Analysis of the co-translational assembly of the fungal fatty acid synthase (FAS). Sci Rep. 2020;10(1):895. pmid:31964902
109. Schweizer M, Roberts LM, Höltke HJ, Takabayashi K, Höllerer E, Hoffmann B, et al. The pentafunctional FAS1 gene of yeast: its nucleotide sequence and order of the catalytic domains. Mol Gen Genet. 1986;203(3):479–86. pmid:3528750
110. Pruitt KD, Tatusova T, Maglott DR. NCBI reference sequences (RefSeq): a curated non-redundant sequence database of genomes, transcripts and proteins. Nucleic Acids Res. 2007;35(Database issue):D61-5. pmid:17130148
111. Gow NA, Robbins PW, Lester JW, Brown AJ, Fonzi WA, Chapman T, et al. A hyphal-specific chitin synthase gene (CHS2) is not essential for growth, dimorphism, or virulence of Candida albicans. Proc Natl Acad Sci U S A. 1994;91(13):6216–20. pmid:8016141
112. Reza MH, Shah H, Manjrekar J, Chattoo BB. Magnesium uptake by CorA transporters is essential for growth, development and infection in the rice blast fungus Magnaporthe oryzae. PLoS One. 2016;11(7):e0159244. pmid:27416318
113. Estrada A, Wright DL, Anderson AC. Antibacterial antifolates: from development through resistance to the next generation. Cold Spring Harb Perspect Med. 2016;6(8):a028324. pmid:27352799
114. Koshy R, Chen T. Combination therapy with trimethoprim-sulfamethoxazole and caspofungin in a case of severe pneumocystis pneumonia. IDCases. 2019;15:e00496. pmid:30828545
115. Nagiec MM, Nagiec EE, Baltisberger JA, Wells GB, Lester RL, Dickson RC. Sphingolipid synthesis as a target for antifungal drugs. Complementation of the inositol phosphorylceramide synthase defect in a mutant strain of Saccharomyces cerevisiae by the AUR1 gene. J Biol Chem. 1997;272(15):9809–17. pmid:9092515
116. Remus BS, Schwer B, Shuman S. Characterization of the tRNA ligases of pathogenic fungi Aspergillus fumigatus and Coccidioides immitis. RNA. 2016;22(10):1500–9. pmid:27492257
117. Maldonado G, García A, Herrero S, Castaño I, Altmann M, Fischer R, et al. The gene YEF3 function encoding translation elongation factor eEF3 is partially conserved across fungi. Front Microbiol. 2024;15:1438900. pmid:39247690
118. Triana-Alonso FJ, Chakraburtty K, Nierhaus KH. The elongation factor 3 unique in higher fungi and essential for protein biosynthesis is an E site factor. J Biol Chem. 1995;270(35):20473–8. pmid:7657623
119. Schneider E, Hunke S. ATP-binding-cassette (ABC) transport systems: functional and structural aspects of the ATP-hydrolyzing subunits/domains. FEMS Microbiol Rev. 1998;22(1):1–20. pmid:9640644
120. Su H, Han L, Huang X. Potential targets for the development of new antifungal drugs. J Antibiot (Tokyo). 2018;71(12):978–91. pmid:30242283
121. Amorim Franco TM, Blanchard JS. Bacterial branched-chain amino acid biosynthesis: structures, mechanisms, and drugability. Biochemistry. 2017;56(44):5849–65. pmid:28977745
122. Steyer JT, Todd RB. Branched-chain amino acid biosynthesis in fungi. Essays Biochem. 2023;67(5):865–76. pmid:37455545
123. Neinast M, Murashige D, Arany Z. Branched chain amino acids. Annu Rev Physiol. 2019;81:139–64. pmid:30485760
124. Zhang P, MacTavish BS, Yang G, Chen M, Roh J, Newsome KR, et al. Cyanobacterial dihydroxyacid dehydratases are a promising growth inhibition target. ACS Chem Biol. 2020;15(8):2281–8. pmid:32786290
125. Demuyser L, Palmans I, Vandecruys P, Van Dijck P. Molecular elucidation of riboflavin production and regulation in Candida albicans, toward a novel antifungal drug target. mSphere. 2020;5(4):e00714-20. pmid:32759338
126. Wagner R, Straub ML, Souciet JL, Potier S, de Montigny J. New plasmid system to select for Saccharomyces cerevisiae purine-cytosine permease affinity mutants. J Bacteriol. 2001;183(14):4386–8. pmid:11418581
127. Paluszynski JP, Klassen R, Rohe M, Meinhardt F. Various cytosine/adenine permease homologues are involved in the toxicity of 5-fluorocytosine in Saccharomyces cerevisiae. Yeast. 2006;23(9):707–15. pmid:16845689
128. Gruber S, Zeilinger S. The transcription factor Ste12 mediates the regulatory role of the Tmk1 MAP kinase in mycoparasitism and vegetative hyphal fusion in the filamentous fungus Trichoderma atroviride. PLoS One. 2014;9(10):e111636. pmid:25356841
129. Vandermeulen MD, Cullen PJ. Ecological inducers of the yeast filamentous growth pathway reveal environment-dependent roles for pathway components. mSphere. 2023;8(5):e0028423. pmid:37732804
130. Benhamou RI, Bibi M, Steinbuch KB, Engel H, Levin M, Roichman Y, et al. Real-Time imaging of the azole class of antifungal drugs in live Candida cells. ACS Chem Biol. 2017;12(7):1769–77. pmid:28472585
131. Hay RJ. Antifungal drugs on the horizon. J Am Acad Dermatol. 1994;31(3 Pt 2):S82-6. pmid:8077515
132. Bowman SM, Free SJ. The structure and synthesis of the fungal cell wall. Bioessays. 2006;28(8):799–808. pmid:16927300
133. Färber PM, Arscott LD, Williams CH Jr, Becker K, Schirmer RH. Recombinant Plasmodium falciparum glutathione reductase is inhibited by the antimalarial dye methylene blue. FEBS Lett. 1998;422(3):311–4. pmid:9498806
134. Monk BC, Sagatova AA, Hosseini P, Ruma YN, Wilson RK, Keniya MV. Fungal Lanosterol 14α-demethylase: a target for next-generation antifungal design. Biochim Biophys Acta Proteins Proteom. 2020;1868(3):140206. pmid:30851431
135. Martin FM, Uroz S, Barker DG. Ancestral alliances: plant mutualistic symbioses with fungi and bacteria. Science. 2017;356(6340):eaad4501. pmid:28546156
136. Bastías DA, Johnson LJ, Card SD. Symbiotic bacteria of plant-associated fungi: friends or foes? Curr Opin Plant Biol. 2020;56:1–8. pmid:31786411
137. van den Bosch TJM, Welte CU. Detoxifying symbionts in agriculturally important pest insects. Microb Biotechnol. 2017;10(3):531–40. pmid:27943632
138. Goodall ECA, Robinson A, Johnston IG, Jabbari S, Turner KA, Cunningham AF, et al. The essential genome of Escherichia coli K-12. mBio. 2018;9(1):e02096-17. pmid:29463657
139. Stanzione F, Giangreco I, Cole JC. Use of molecular docking computational tools in drug discovery. Prog Med Chem. 2021;60:273–343. pmid:34147204
140. Berman HM, Westbrook J, Feng Z, Gilliland G, Bhat TN, Weissig H, et al. The protein data bank. Nucleic Acids Res. 2000;28(1):235–42. pmid:10592235
141. Holm L. Dali server: structural unification of protein families. Nucleic Acids Res. 2022;50(W1):W210–5. pmid:35610055
142. van Kempen M, Kim SS, Tumescheit C, Mirdita M, Lee J, Gilchrist CLM, et al. Fast and accurate protein structure search with Foldseek. Nat Biotechnol. 2024;42(2):243–6. pmid:37156916
143. Liu YJ, Hall BD. Body plan evolution of ascomycetes, as inferred from an RNA polymerase II phylogeny. Proc Natl Acad Sci U S A. 2004;101(13):4507–12. pmid:15070748
144. Hoffman CS, Wood V, Fantes PA. An ancient yeast for young geneticists: a primer on the Schizosaccharomyces pombe model system. Genetics. 2015;201(2):403–23. pmid:26447128
145. Kachroo AH, Laurent JM, Yellman CM, Meyer AG, Wilke CO, Marcotte EM. Evolution. Systematic humanization of yeast genes reveals conserved functions and genetic modularity. Science. 2015;348(6237):921–5. pmid:25999509
146. Cockrell A, McCormick SP, Moore MJ, Chakrabarti M, Lindahl PA. Mössbauer, EPR, and modeling study of iron trafficking and regulation in Δccc1 and CCC1-up Saccharomyces cerevisiae. Biochemistry. 2014;53(18):2926–40. pmid:24785783
147. Sorribes-Dauden R, Peris D, Martínez-Pastor MT, Puig S. Structure and function of the vacuolar Ccc1/VIT1 family of iron transporters and its regulation in fungi. Comput Struct Biotechnol J. 2020;18:3712–22. pmid:33304466
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2025 E. Condon et al. This is an open access article distributed under the terms of the Creative Commons Attribution License: http://creativecommons.org/licenses/by/4.0/ (the “License”), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Fungi are a major threat to human health and agricultural productivity, causing 1.7 million human deaths and billions of dollars in crop losses and spoilage annually. While various antifungal compounds have been developed to combat these fungi in medical and agricultural settings, there are concerns that effectiveness is waning due to the emergence of acquired drug resistance and novel pathogens. Effectiveness is further hampered due to the limited number of modes of action for available antifungal compounds. To develop new strategies for the control and mitigation of fungal disease and spoilage, new antifungals are needed with novel fungal-specific protein targets that can overcome resistance, prevent host toxicity, and can target fungi that have no effective control measures. The increasing availability of complete genomes of pathogenic and spoilage fungi has enabled identification of novel protein targets essential for viability and not found in host plants or humans. In this study, an automated bioinformatics pipeline utilizing BLAST, Clustal , and subtractive genomics was created and used to identify potential new targets for any combination of hosts and pathogens with available genomic or proteomic data. This pipeline called HitList allows in silico screening of thousands of possible targets. HitList was then used to generate a list of potential antifungal targets for the World Health Organization fungal priority pathogens list and the top 10 agricultural fungal pathogens. Known antifungal targets were found, validating the approach, and an additional eight novel protein targets were discovered that could be used for the rational design of antifungal compounds.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer