Home Biology Ploidy evolution in a wild yeast is linked to an interplay between cell sort and metabolism

Ploidy evolution in a wild yeast is linked to an interplay between cell sort and metabolism

Ploidy evolution in a wild yeast is linked to an interplay between cell sort and metabolism

[ad_1]

Introduction

Ploidy is a elementary facet of the biology of all organisms, however it’s topic to placing range throughout the tree of life—between associated species, between people of the identical species, and inside people throughout cell varieties and life cycles [1]. The long-term impression of ploidy variation on eukaryotic evolution, significantly as a mechanism for producing uncooked materials for pure choice, has lengthy been acknowledged [26]. Latest work, primarily within the mannequin eukaryote Saccharomyces cerevisiae, has additional outlined short-term evolutionary penalties of various ploidy states [713]. It stays much less clear, nevertheless, what fast results on organismal health a ploidy transition can engender. In S. cerevisiae, ploidy variation is current each inside the pure life cycle [14] and amongst isolates from numerous environments [15,16]. Regardless of this pure variation, diploidy appears to be typically favored [15]. Certainly, diploids continuously come up and sweep to fixation in laboratory evolution experiments based with non-diploid strains [7,1722].

Within the restricted circumstances the place a direct health benefit of diploidy has been present in S. cerevisiae within the absence of confounding variation, the precise molecular bases have remained elusive. A big-scale survey of S. cerevisiae and its sister species Saccharomyces paradoxus instructed that particular ploidy-by-environment interactions have been obligatory to clarify noticed variations in health proxies between haploids and diploids, which argues towards generalizable predictions of health results of ploidies throughout environments [23]. Comparable experiments in Candida albicans discovered genetic background to affect health greater than ploidy in a number of circumstances that may be predicted to favor totally different ploidy states [24]. Against this, more moderen work capturing a large swath of genetic range in Saccharomyces eubayanus, which diverged from S. cerevisiae roughly 17 million years in the past [25], failed to search out significant variations in phenotypic traits between ploidies [26].

Including complexity to the interpretation and prediction of health variations between ploidy states in yeasts is the nuanced relationship between ploidy and cell sort throughout species. In wild-type Saccharomyces, for instance, ploidy not directly controls cell sort via the presence or absence of alleles at a single locus, the MATing sort locus [14,27,28]. The haploid cell varieties categorical a typical set of haploid-specific genes, in addition to mating type-specific genes depending on the allele current on the MAT locus, whereas diploids repress these gene units however are competent to induce the expression of a small variety of genes below particular circumstances (e.g., meiosis). Though investigations into ploidy-specific health results have primarily centered on the physiological variations between haploids and diploids which are impartial of cell sort, it stays believable that underappreciated points of cell-type specification might affect traits that in flip impression organismal health.

Maybe, essentially the most compelling proof for widespread results of choice on cell sort throughout fungi will be discovered amongst pathogenic species. Extremely skewed mating sort ratios have been described amongst isolates of Cryptococcus neoformans, Candida glabrata, Candida auris, Fusarium poae, and Fusarium verticillioides [2935]. Massive mating sort skews are additionally present in medical isolates of Aspergillus fumigatus however not in isolates from different sources, and mating sort has been proven to affect pathogenicity in vitro and virulence in vivo on this species [36,37]. Comparable hyperlinks between mating sort and virulence traits have been instructed in Cr. neoformans, C. auris, Mucor iregularis, and Fusarium graminearum [3846], suggesting that surprising hyperlinks between cell sort and traits experiencing intense choice could also be widespread amongst fungi.

Microbial traits and their underlying genotypes are of specific curiosity once they straight impression human well being, are essential for biotechnological processes, or function fashions of eukaryotic evolution. The latter 2 circumstances are exemplified within the rising mannequin yeast S. eubayanus, the wild mother or father of hybrid lager-brewing yeast. Since its isolation as a pure species [47], S. eubayanus has grow to be a mannequin for microbial inhabitants genomics and ecology [4853], in addition to a key goal for utilized biotechnological analysis [5459]. A focal ecological and industrial trait on this wild species is the flexibility to devour and metabolize the α-glucoside maltose, which is essentially the most considerable sugar within the wort used to brew beer [60,61]. This trait is almost ubiquitous amongst isolates of S. eubayanus and its sister species Saccharomyces uvarum [62], however it has been misplaced [63] or severely curtailed [48,49,64,65] within the Holarctic subpopulation of S. eubayanus, a low-genetic range lineage broadly distributed throughout the northern hemisphere that comprises the closest recognized family to the S. eubayanus subgenome of hybrid lager-brewing yeasts [51,53]. Paradoxically, the genomes of Holarctic S. eubayanus strains include practical structural maltose metabolism genes, which seem like inefficiently expressed within the presence of maltose [63,64]. As a result of the cis-regulatory logic of a minimum of a few of these structural genes seems to have been retained, it has been proposed that the trans-regulating proteins might have been rendered nonfunctional [63], a minimum of as regards to their homology-predicted actions.

In an effort to determine mechanisms by which maltose utilization may be refined or regained after secondary loss, we beforehand subjected a wild diploid S. eubayanus pressure from the Holarctic subpopulation to adaptive laboratory evolution (ALE) below choice for improved progress on maltose [64]. Right here, we map the genetic foundation of adaptation within the advanced clones. We discover that, surprisingly, haploids emerged and rose to excessive frequency in replicate ALE populations based with this diploid pressure, which is a extremely uncommon ploidy transition for Saccharomyces. We discover that haploidy confers a considerable health benefit within the ALE circumstances, however that haploids expertise a health tradeoff in wealthy circumstances, in keeping with earlier observations of diploid benefit in S. cerevisiae. We determine cell sort as the first driver of adaptive health, with a smaller however important contribution from absolute ploidy. Lastly, we display {that a} main fitness-modifying gene has elevated expression in advanced haploids, and that this impact is linked to surprising regulation by a haploid-specific transcription issue that regulates invasive progress in S. cerevisiae. Our outcomes counsel a mechanism underlying a ploidy-by-environment health impact and display how robust choice on traits linked to cell varieties can drive karyotypic evolution in fungi.

Outcomes and dialogue

Developed S. eubayanus isolates harbor mutations incongruous with ancestral ploidy

We beforehand experimentally advanced a wild pressure of S. eubayanus from the Holarctic subpopulation below choice for improved progress on the industrially related α-glucoside maltose [64]. We picked clones from 2 replicate populations of the ALE experiment that displayed considerably elevated progress (p = 0.002, Mann–Whitney U assessments) on maltose in comparison with the ancestral pressure (Fig 1A). To map the genetic foundation of improved progress on maltose, we sequenced the genomes of every clone to a closing common depth of 95-fold. We mapped these reads to a re-sequenced and annotated meeting of the ancestral pressure and recognized a complete of 4 single-nucleotide polymorphisms (SNPs) and three large-scale copy quantity variants (CNVs) within the type of aneuploidies throughout the advanced isolates (Fig 1B and S3 Desk). We didn’t determine single-nucleotide variants in or close to any genes with clear relationships to α-glucoside metabolism, though 1 SNP launched a untimely cease codon in IRA1, a typical goal of adaptive mutations in batch-style experimental evolution [7,22,6668]. One aneuploidy (ChrXV achieve) was shared between advanced isolates and encompassed a homolog of the S. cerevisiae generalist α-glucoside transporter AGT1/YGR289C, suggesting a possible mechanism for adaptation (Fig 1B); we didn’t detect additional copy quantity enlargement of this gene within the advanced isolates (S1 Fig).

thumbnail

Fig 1. Phenotypic and karyotypic evolution of S. eubayanus isolates.

(a) Progress of the wild S. eubayanus pressure (WT) and clonal isolates from 2 replicate experimental evolution populations (Evol. 1, Evol. 2) on maltose. A consultant progress curve is proven for every; bar plots present imply and customary error of complete progress (AUC) throughout 6 organic replicates for every genotype. (b) Relative copy variety of every chromosome within the WT and advanced strains, inferred from sequencing depth. The parallel ChrXV achieve features a homolog of an S. cerevisiae gene encoding an α-glucoside transporter (AGT1/YGR289C). (c) Smoothed histograms of mobile DNA content material within the WT and advanced strains as measured by circulation cytometry. Fluorescence depth is proportional to DNA content material; major peaks correspond to cells in G1 and G2. The information underlying this determine will be present in S1 Information. AUC, space below the curve; WT, wild-type.


https://doi.org/10.1371/journal.pbio.3001909.g001

Unexpectedly, all SNPs within the advanced isolates have been represented by a single, non-reference allele (S2 Fig). Though mitotic recombination can generate losses of heterozygosity at new or standing variation throughout adaptive evolution [6975], our outcomes differed considerably from 2 current large-scale experimental evolution research in S. cerevisiae, which discovered roughly 5% to 10% of mutations to be homozygous in diploid or autodiploid clones after 4,000 generations [7,9]. Compared, our noticed allele frequencies at mutated websites are extremely inconceivable below the null expectation of diploidy (binomial assessments: p = 5.3 × 10−6, p = 1 × 10−4, respectively). Thus, we reasoned that the noticed patterns in allele frequency would possibly greatest be defined by an surprising and atypical ploidy discount to haploidy throughout ALE.

Haploids emerged and rose to excessive frequency in diploid-founded populations

We straight decided the ploidy states of the advanced clones and the ancestral pressure utilizing circulation cytometry (Fig 1C) and confirmed that the pressure that was used to discovered the experimental populations was diploid (S3A Fig). In keeping with the outcomes of genome sequencing, we discovered that clones from each ALE replicates had grow to be haploid (Fig 1C). To check whether or not the clonal isolates we analyzed have been merely from a uncommon and nonrepresentative subpopulation, we assayed the ploidy states current on the inhabitants degree in each replicates of the ALE experiment (S3B Fig). Haploids have been clearly detectable in every replicate by roughly 100 and 250 generations, respectively. As an orthogonal strategy, we plated cells from the terminal time level of every inhabitants of the ALE experiment and used a PCR assay to genotype the MAT locus of single colonies. By this methodology, haploids constituted 74% to 100% of the cells we genotyped within the 2 ALE populations (S3C Fig). All haploids genotyped by PCR have been discovered to be MATa, as have been each sequenced isolates. Thus, though haploids might not have swept to fixation in each experimental populations, they repeatedly emerged and rose to excessive frequency over the period of the ALE experiment.

Haploids exhibit a direct condition-dependent health benefit

The abundance of haploids in our experimental populations may very well be defined by 2 various fashions: haploids might need a direct health benefit, or they could profit not directly from elevated adaptability in our ALE setting. Two well-documented traces of proof from earlier research appeared to strongly favor the latter speculation. First, S. cerevisiae haploids have repeatedly been proven to adapt extra quickly than diploids throughout experimental evolution, partly attributable to dominance results at adaptive targets and ploidy-specific mutation charges and spectra; even large-scale mutations, comparable to aneuploidies, can have totally different health results in numerous ploidies [712,76,77]. Second, S. cerevisiae shows a powerful development of converging on a diploid state throughout experimental evolution initiated with non-diploid strains [18]. Though concept predicts that haploids could also be higher in a position to meet their metabolic wants in nutrient-limiting circumstances attributable to elevated cell floor area-to-volume ratios, experimental proof in yeast has failed to search out widespread assist for such generalizable traits [23,7881], and our experimental evolution circumstances couldn’t strictly be thought of to be restricted in key vitamins. Given the relative simplicity of testing for variations in health between ploidies, we first sought to assist or refute the mannequin of direct haploid benefit.

We used a delicate competitors assay to measure the health of isogenic diploids and haploids within the wild-type pressure background following HO deletion, sporulation, and tetrad dissection. In keeping with observations in S. cerevisiae of direct or cryptic diploid benefit [7,15,17,18,20,22,82], haploids in our pressure background exhibited median health defects of 1.5% (p = 1.3 × 10−5) to 2.7% (p = 9.9 × 10−5, Mann–Whitney U assessments) relative to the isogenic diploid in wealthy medium (Fig 2A). Against this, within the ALE circumstances, haploids displayed median health benefits of 24.8% (p = 1.6 × 10−9) to twenty-eight.8% (p = 2.4 × 10−9, Mann–Whitney U assessments) per technology over diploids (Fig 2B). Curiously, we noticed a big health distinction between haploids of reverse mating varieties in each environments examined (Fig 2; wealthy medium p = 7.1 × 10−10, evolution circumstances p = 0.013, Mann–Whitney U assessments), suggesting a typical underlying mechanism linked to mating sort, reasonably than a particular mating type-by-environment interplay. Expression of the mating-type genes is expensive [83], making parts of this pathway widespread targets of adaptive loss-of-function mutations in haploids [7,67]. The noticed health defect of MATa haploids in our experiments might replicate an expression burden imposed by the higher variety of MATa-specific genes; a metabolic burden imposed by synthesizing the extra complicated, posttranslationally modified a-factor pheromone; or each. Whereas earlier large-scale research in S. cerevisiae, S. paradoxus, and S. eubayanus haven’t reported basic health variations between mating kinds of in any other case isogenic haploids [23,26], the delicate, however important, variations we noticed right here might have been beneath earlier limits of detection. Alternatively, the obvious defect of MATa cells could also be particular to the two circumstances we examined, so it stays to be decided whether or not our observations of mating-type health results are utterly generalizable on this pressure background or extra broadly. No matter mating sort, we discover that haploids have a big and surprising benefit over diploids below the ALE circumstances.

thumbnail

Fig 2. Haploids have a conditional health benefit.

Boxplots present health measurements of isogenic diploids (n = 12) and haploids from totally viable tetrads (n = 47) in wealthy medium (a) and ALE circumstances (b). *** p < 10−4 (Mann–Whitney U assessments) between diploids and every haploid group (black) or between haploid teams (teal). In ALE circumstances, the importance degree between haploid teams was 0.013 (*). A diploid outlier in (a) at −6.6% is truncated from the plot house. The information underlying this determine will be present in S1 Information. ALE, adaptive laboratory evolution.


https://doi.org/10.1371/journal.pbio.3001909.g002

Haploid health benefit is primarily attributable to cell-type specification

In Saccharomyces, ploidy is intrinsically linked with cell- and mating-type specification, that are decided by the allelic composition of the MAT locus (Fig 3A) [28]. Some variations in cell physiology and gene expression patterns between ploidies are attributable solely to complete mobile DNA content material, whereas lack of heterozygosity on the MAT locus establishes certainly one of 2 partially overlapping, cell type-specific gene expression applications [27,84,85]. The connection between DNA content material and cell-type specification can serve to confound inferences of the underlying foundation of health variations between ploidies, though in restricted circumstances, contributions of both absolute ploidy or MAT locus composition have been documented [7,23]. Right here, we consult with “cell-type specification” as the excellence between genotypes with a full complement of cell-type grasp regulators on the expressed MAT locus (e.g., wild-type diploids containing MATa1, MATα1, MATα2) and people with out. Cell varieties established by the absence of a number of cell-type regulators (e.g., wild-type haploids) impact the de-repression of a handful of genes, generally known as “haploid-specific,” however whose expression is technically impartial of ploidy and mating sort.

thumbnail

Fig 3. Cell-type specification is the first contributor to adaptive health.

(a) Simplified schematic of the cell-type specification circuit in Saccharomyces as decided by the MAT locus on ChrIII. Proteins encoded by every MAT idiomorph and their regulatory targets elsewhere within the genome are depicted. Haploids (high and center) categorical mating type-specific genes and a typical set of haploid-specific genes. Diploids (backside) repress all 3 units (clear gene symbols). (b) Schematic of strains in comparison with decide ploidy and cell-type results on health. The three courses of cell type-specific genes are depicted as coloured bars, with opacity indicating expression in a given genotype. Asterisks within the higher proper nook of fields point out the three wild-type genotypes. The engineered genotypes (no asterisks) have been created by deleting or including full MAT cassettes, except the “MAT-null” strains (marked ∅), which categorical haploid-specific, however not mating type-specific, genes as a result of they maintain solely MATα2. Dotted pink traces are comparisons that present the impact of absolute DNA content material, and stable traces are comparisons that present the impact of cell sort, with colours similar to (c). (c) Factors present variations in health in ALE circumstances between genotypes that differ in solely cell sort (left cluster, e.g., α vs. a/α) or ploidy (proper cluster). Grey shading reveals the density distribution of every group. In every case, the wild-type state is taken because the baseline for comparability (diploid; a/α cell sort). (d) Estimates and 95% confidence intervals for the impact of every variable on the distinction in health. The information underlying this determine will be present in S1 Information. ALE, adaptive laboratory evolution.


https://doi.org/10.1371/journal.pbio.3001909.g003

To dissect the contributions of DNA content material and cell sort to organismal health in our system, we generated a panel of 8 in any other case isogenic genotypes with distinctive combos of ploidy, mating sort, and cell type-specific gene expression (Fig 3B). We measured the health of those strains within the ALE situation (Fig 3C) and estimated the separable results of ploidy, mating-type specification, and cell type-specific gene expression patterns on health (Fig 3D). These 3 elements defined the vast majority of the variance in measured health throughout genotypes (a number of R2 = 0.96, df = 86, p < 2.2 × 10−16), with every having a big impact (p ≤ 2.56 × 10−7). Remarkably, cell-type specification had an impression on organismal health that was virtually an order of magnitude higher than both ploidy or mating sort (Fig 3D, health benefit estimate 18.8%, 95% CI: 17.7, 19.9), and defined way more of the variance (proportion sum squares: cell sort, 0.93; ploidy, 0.016; mating sort, 0.014). Absolute ploidy nonetheless impacted health throughout cell varieties, with haploids experiencing a 2.3% benefit relative to diploids within the ALE situation (Fig 3D, 95% CI: 1.5, 3.1). Paradoxically, expression of mating type-specific genes in these experiments appeared to modestly improve health between haploid-like cell varieties within the ALE situation (Fig 3D), in distinction to the documented price of their expression in different circumstances [83]. Whereas one potential interpretation is that each units of mating type-specific genes confer bona fide health benefits to cells rising in maltose medium, an alternate rationalization for this obvious discrepancy is that haploid-like, MAT-null cells expertise modest health defects because of their aberrant and synthetic cell sort. As such, our analyses might barely underestimate the health profit attributable to haploid-like cell sort within the ALE situation. We conclude that the cell sort specified by the MAT locus, reasonably than absolute ploidy per se, has the most important impact on health within the ALE situation.

Dynamics of different ploidy variants in adapting populations

We subsequent investigated the evolutionary dynamics and adaptive advantage of the opposite shared ploidy variant within the advanced clones: aneuploidy of ChrXV (Fig 1B). We carried out bulk whole-genome sequencing on the cryopreserved replicate ALE populations from the identical time factors at which we assayed ploidy states by circulation cytometry (S2B Fig) and quantified the obvious frequency of every chromosome in each populations as estimated by relative protection (S4 Fig). The one aneuploidies that rose to an considerable frequency have been these sampled in our clonal isolates: ChrXV (each populations) and ChrI (1 inhabitants). The dynamics of ploidy variants—together with each aneuploidy and haploidy—and the relative timing of their emergence differed between populations. Within the first inhabitants, the ChrXV aneuploidy rose to close fixation previous to the obvious emergence of haploids, whereas the rise in frequency of the ChrI aneuploidy was roughly coincident with that of haploids (S4B Fig). Within the second inhabitants, the ChrXV aneuploidy and the haploid state had remarkably comparable trajectories, with ChrXV aneuploidy showing to precede haploidy barely: each rose precipitously in frequency after roughly 50 generations, declined dramatically, and subsequently rebounded by the terminal time level (S4B Fig). These outcomes counsel {that a} haploid lineage with ChrXV aneuploidy was topic to clonal interference from 1 or extra extremely match genotypes on this replicate.

The change in frequency of the ChrXV aneuploidy over time in each populations instructed a powerful health profit, which we reasoned was doubtless attributable to the presence of AGT1 on this chromosome (Fig 1B). S. eubayanus Agt1p is a homolog of the well-characterized S. cerevisiae α-glucoside transporter, however in distinction to canonical MAL gene clusters that include structural and regulatory maltose metabolism genes, S. eubayanus AGT1 is remoted within the subtelomeric area of ChrXV. In our genome meeting, no predicted genes intersperse the AGT1 begin codon and the start of telomeric repeats some 6,770 bp upstream. We didn’t determine any homologs of genes encoding MAL regulators, transporters, α-glucosidases, or isomaltases on ChrXV, nor different robust candidates to clarify the adaptive potential of the aneuploidy. We thus examined whether or not copy quantity variation at AGT1 alone offered an adaptive profit within the ALE setting to clarify the sweep of ChrXV aneuploidy in each populations. We inserted an extra copy of AGT1 below its native promoter and terminator into the genomes of diploids and haploids at a separate web site, and we measured the health of the ensuing strains within the ALE circumstances. As predicted, elevated AGT1 copy quantity conferred a considerable and important health profit in each diploids and haploids (S5 Fig). Haploids acquired a extra modest improve in health than diploids upon the addition of AGT1 (S5A Fig), which we attribute to the consequences of diminishing returns epistasis; nonetheless, they have been considerably healthier general (S5B Fig). There was no interplay between the haploid mating sort and the health impact of elevated AGT1 copy quantity. Thus, the ChrXV aneuploidy we noticed in each clonal isolates and the ALE populations doubtless contributed to adaptation by growing copy variety of AGT1, and its emergence might have preceded that of haploids.

AGT1 expression is elevated in aneuploid haploids

The conditional health benefit of haploids and elevated health of haploid-like cell varieties instructed an surprising regulatory hyperlink between maltose metabolism and haploid-specific genes (i.e., these genes de-repressed within the absence of a heterozygous MAT locus). To determine potential targets of this interplay, we analyzed mRNA-seq knowledge collected from the wild-type diploid and advanced haploids grown in circumstances mimicking the evolution experiment (SC-maltose), in addition to a baseline for comparisons (SC-glucose). Though the haploid strains had discrete polymorphisms, they shared a typical cell sort and aneuploidy of chromosome XV (Fig 1B); thus, we reasoned that widespread variations in expression between these isolates relative to the wild-type pressure must be attributable to 1 (or each) of those shared genotypes. Transcriptomes of the advanced haploids have been extremely comparable, as anticipated (S6 Fig). Differentially expressed genes (DEGs) between the wild-type pressure and advanced haploids have been enriched for cell and mating type-specific transcripts and genes on aneuploid chromosomes; nevertheless, there was no clear practical enrichment amongst DEGs to clarify the maltose-specific haploid health benefit. The AGT1 transporter on ChrXV was the one maltose metabolism gene up-regulated in maltose in each advanced haploids when in comparison with the wild-type pressure, which was anticipated given its 2-fold relative copy quantity in these isolates (Fig 1B). Upon nearer examination, nevertheless, AGT1 expression was increased than the 2-fold improve anticipated commensurate with its relative copy quantity [86,87]. Certainly, AGT1 expression in haploids exceeded null expectations primarily based on 2 distinct fashions (Fig 4A): (1) we calculated the fold change for AGT1 within the ancestral pressure in maltose in comparison with glucose and utilized this multiplier to the glucose expression degree within the advanced haploids; and (2) we utilized a 2-fold multiplier to the gene expression ranges within the wild-type pressure in each glucose and maltose, which accounted for copy quantity variation within the advanced haploids. Whereas AGT1 expression in glucose within the advanced haploids was consistent with the naïve aneuploid expectation (p = 0.81, one-sided Mann–Whitney U check), its expression in maltose within the advanced haploids was a mean of 69% increased than may very well be modeled by accounting for copy quantity and native regulation (Fig 4A, p = 0.0005, one-sided Mann–Whitney U check).

thumbnail

Fig 4. Elevated expression of an α-glucoside transporter gene in haploids.

(a) Factors and bars present imply and customary error of AGT1 expression in advanced haploids, plotted towards the null expectation of expression primarily based on copy quantity variation and induction within the WT pressure. Expression in maltose is larger than the null expectations within the advanced haploids (p = 0.0005, one-sided Mann–Whitney U check). (b–d) Boxplots present LFC of gene expression in maltose in advanced haploids in comparison with the WT pressure. Whiskers lengthen to 1.5× the interquartile vary. Strains join the y-axis coordinates of the identical gene in every advanced isolate; axes are scaled such that an occasional outlier is truncated from the plot house for a single pressure. AGT1 expression is plotted as pink dots and features, and black dashed traces point out the null expectation for expression values. (b) Genes induced in maltose within the WT pressure (n = 544). (c) Genes on aneuploid ChrXV (n = 370). (d) Subtelomeric genes (n = 200). For all courses, expression in both advanced haploid just isn’t considerably higher than the null expectation (one-sided t assessments, min. p = 0.42). The information underlying this determine will be present in S1 Information. LFC, log2-transformed fold change; WT, wild-type.


https://doi.org/10.1371/journal.pbio.3001909.g004

We subsequent requested whether or not elevated AGT1 expression may very well be defined by delicate modifications in world gene expression ranges between the wild-type diploid and advanced haploids. We in contrast expression ranges of two related courses of genes below which AGT1 falls and which we reasoned may be topic to modest differential expression: maltose-induced genes and subtelomeric genes (Fig 4B and 4D). We additionally examined expression of genes on the aneuploid ChrXV to check whether or not these broadly exceeded the expectation of a 2-fold expression improve commensurate with copy quantity (Fig 4C). In every case, expression within the advanced haploids was not considerably higher than the null expectation (one-sided t assessments, p > 0.4), and AGT1 expression in maltose was within the higher tail of gene expression values for every class. Most notably, AGT1 expression in maltose relative to the euploid diploid ranked increased than 95.9% and 98.6% of different ChrXV genes in every advanced haploid, respectively.

In keeping with quite a few research in yeasts, we noticed common expression from the aneuploid ChrXV to be elevated, if not precisely 2-fold increased than within the euploid diploid [86,8893]. Importantly, we noticed this expression attenuation throughout circumstances, that means that the elevated expression noticed at AGT1 just isn’t more likely to be an artifact of condition-specific aneuploid gene expression variations. In comparison with the wild-type pressure, we noticed median fold modifications for ChrXV genes of 1.58 in maltose for each advanced haploids (Fig 4C) and 1.72 and 1.70 in glucose, respectively (S7A Fig). Certainly, the potential impact of cell sort on AGT1 expression turns into much more evident in mild of the median expression ranges of aneuploid genes in haploids: AGT1 is up-regulated a mean of 4-fold in maltose throughout the haploid strains (S4 Desk and S7B Fig), whereas median fold modifications for all ChrXV genes between maltose and glucose are 0.969 and 0.970 for every haploid, respectively (S7B Fig). In comparison with roughly 2.3-fold induction of AGT1 in maltose within the euploid diploid (S4 Desk), this elevated induction within the advanced isolates might replicate the combinatorial results of cell sort and sugar response. As elevated AGT1 copy quantity (which ought to end in a concomitant improve in expression) considerably will increase health in maltose (S5 Fig), the elevated expression noticed in haploids can also be more likely to contribute to adaptation—and will clarify the condition-specific health benefit of isogenic haploids.

Naturally, it stays a chance that the elevated expression of AGT1 that we noticed in aneuploid haploids is the results of an interplay (whether or not direct or oblique) between this locus and a number of genes elsewhere on the chromosome. As well as, aneuploidy itself can set off a transcriptional response [89,92,94], though to our information, this response doesn’t lengthen to maltose metabolism genes. As an entire dissection of aneuploidy response and its targets in S. eubayanus—in addition to how this response might differ between ploidies and cell varieties—is past the scope of the present work, we as an alternative investigated potential regulators of AGT1 that might clarify its cell type-linked improve in expression (detailed beneath). Thus, our knowledge can not unequivocally reject the speculation that whole-chromosome duplication itself might have an effect on AGT1 expression along with the function we’ve established for gene copy quantity and cell sort.

The AGT1 promoter integrates cell-type and sugar-responsive regulatory networks

We first investigated potential regulators of AGT1 by scanning its promoter for putative transcription factor-binding websites utilizing high-confidence S. cerevisiae motifs (S5 Desk). This evaluation recognized clustered binding motifs for the canonical constructive and destructive regulators of maltose metabolism genes, Mal63p and Mig1p (Fig 5A), in a corporation in keeping with the characterised regulatory module that controls the expression of maltose metabolism genes in S. cerevisiae [9597]. Though a causal relationship has not been straight established, the presence of Mal63p consensus sequences upstream of maltose metabolism genes is nicely correlated with their induction by maltose within the sort pressure of S. eubayanus [58]. Along with these anticipated regulators, we recognized putative binding websites for a number of transcription elements which are concerned in regulating filamentous progress (e.g., these encoded by ASH1, SIP4, STE12, FKH1, MIG1/MIG2, and NRG1). This class was significantly noteworthy as a result of filamentous progress will be induced in response to glucose depletion as a hunger response, and it requires a haploid-specific gene, TEC1 [27,98103]. Along with dimerizing with Ste12p, Tec1p can activate goal genes as a monomer in a dosage-dependent trend [104106], and it has been experimentally mapped to its consensus motif (TEA/ATTS consensus sequence or TCS) in vivo throughout the genus Saccharomyces [107]. We recognized a TCS within the promoter of AGT1 and hypothesized that Tec1p might mediate the cell type-specific improve in AGT1 expression we noticed in haploids. Supporting this notion, and in keeping with its characterization as a haploid-specific gene in S. cerevisiae [27], TEC1 was considerably up-regulated in each advanced isolates in our dataset (Fig 5B and S4 Desk).

thumbnail

Fig 5. AGT1 is regulated by cell sort.

(a) Schematic of the AGT1 promoter and reporter constructs. A clustered regulatory module containing Mal63p and Mig1p motifs (white containers) lies upstream of AGT1 in its native context (high), which is harking back to different maltose metabolism genes in Saccharomyces. One additional predicted binding web site for every regulator that lies nearer to the coding sequence is omitted for house. The promoter comprises a motif for Tec1p (TCS, pink field). We generated reporter constructs expressing GFP from the wild-type promoter (center, PAGT1) and a model with level mutations to the Tec1p motif (backside, PAGT1-tcs). (b) TEC1 expression is cell sort dependent in S. eubayanus. Factors and bars present imply and customary error of TEC1 expression (normalized counts) within the wild-type diploid and advanced haploids, averaged throughout circumstances. (c) Level mutations to the anticipated Tec1p-binding web site within the AGT1 promoter cut back reporter expression. Every level reveals the imply inhabitants fluorescence for a replicate experiment with a management untagged pressure (grey), in addition to strains expressing GFP from the wild-type AGT1 promoter (pink) or a promoter with a mutated Tec1p motif (teal). All engineered strains are considerably totally different from the untagged management (p ≤ 4.3 × 10−6, two-sided t assessments), and teams of promoter genotypes differ considerably (two-sided t check). The information underlying this determine will be present in S1 Information. TCS, TEA/ATTS consensus sequence.


https://doi.org/10.1371/journal.pbio.3001909.g005

To check this speculation, we cloned yEGFP below the management of the wild-type AGT1 promoter (PAGT1), in addition to a promoter variant with level mutations within the predicted Tec1p-binding web site (Pagt1-tcs), and launched a single copy of those reporters to the genome of euploid MATa haploids. We then measured single-cell fluorescence of the ensuing strains grown in maltose by circulation cytometry. Mutation of the Tec1p-binding web site considerably decreased fluorescence from the reporter assemble in comparison with the wild-type promoter (p < 2.2 × 10−16, two-sided t check), however it didn’t abolish expression utterly (Fig 5C). These outcomes are in keeping with the expression knowledge and collectively counsel that AGT1 receives regulatory enter from each cell-type and sugar-responsive networks, with separable activation by Tec1p and induction within the presence of maltose. We additionally measured expression of the PAGT1-GFP reporter in a number of media circumstances utilizing a much less delicate plate-based assay (S8 Fig). Throughout progress on glucose and galactose—each anticipated to be non-inducing—no fluorescence above baseline was detected. Progress on maltose induced expression considerably, as did progress on methyl-α-glucoside, one other substrate transported by AGT1 in S. cerevisiae [108,109]. Curiously, we additionally noticed modest reporter expression when cells have been pre-grown in glucose and switched to medium containing no sugar (S8 Fig), additional supporting the notion that AGT1 could also be expressed in response to suboptimal carbon circumstances on this background.

In synthesis, the proof for a direct health benefit by haploid-like cell varieties (Figs 2 and 3), elevated expression of fitness-modifying AGT1 in haploids (Fig 4), and the partial dependence of AGT1 expression on the motif for a haploid-specific transcription issue (Fig 5) suggests a relationship between ploidy evolution and adaptation in our system. Future experimentation might extra clearly outline the function of AGT1 and its regulation in definitively driving the ploidy evolution we noticed, comparable to by replaying the experimental evolution utilizing genotypes missing TEC1 or with totally different promoters driving AGT1. Though absolute ploidy does appear to impart a small health distinction in our experiments, the total impression on health requires cell sort (Fig 3C and 3D).

Conclusions

Resolving the genotype-to-phenotype map stays a central aim in genetics and evolutionary biology, however it has continuously confirmed difficult, even in microbes. Whereas gene content material is mostly correlated with metabolic traits throughout budding yeasts [25], regulatory nuances in organisms that aren’t conventional fashions can confound inferences of phenotypes from genome sequences [63,110,111]. Within the taxonomic sort pressure of S. eubayanus, structural maltose metabolism genes in canonical MAL clusters are exquisitely repressed or induced hundreds-fold in response to carbon supply [58], which has similarities to their S. cerevisiae homologs [112]. Against this, within the pressure from the Holarctic subpopulation studied right here, what seems to be the focal maltose transporter is partially decoupled from such stringent catabolite regulation: AGT1 is barely induced roughly 2.3-fold within the wild-type pressure in maltose (S4 Desk). We are able to envision 2 potential explanations for the apparently uncommon regulation of this gene.

First, AGT1 is more likely to encode a transporter with broad substrate affinity like its S. cerevisiae homolog [64,113116], whereas different phylogenetically distinct maltose transporters are inclined to have increased specificity [108,117]. It’s potential that choice favored putting management of this generalist transporter below a broader transcriptional response to hunger or glucose depletion as a part of a scavenging technique, which the transition to filamentous progress is assumed to symbolize [102]. Certainly, current work has instructed that maltose could also be an surprising inducer of filamentous progress in S. cerevisiae [118]. Decoupling various carbon metabolism genes from their stringent canonical regulation has additionally been proven to be adaptive amongst isolates of S. cerevisiae topic to particular ecologies [119] and ALE in fluctuating environments [120].

Second, the group—and doubtlessly regulation—of AGT1 in S. eubayanus could also be reflective of the ancestral state for Saccharomyces. In strains of Saccharomyces paradoxus, Saccharomyces mikatae, and S. eubayanus, AGT1 homologs are scattered in subtelomeric areas and never in canonical MAL loci, whereas homologs encoding high-affinity maltose transporters are inclined to happen in gene clusters with the everyday group [63,96]. Thus, the precise group of AGT1 within the MAL1 locus of mannequin S. cerevisiae strains—and its ensuing beautiful regulation by glucose and maltose—might itself symbolize a derived state that’s not reflective of untamed yeasts. Certainly, there may be clade-specific variation inside S. cerevisiae as as to whether the MAL1 locus is occupied by the generalist AGT1 or a gene encoding a high-specificity maltose transporter [121], suggesting that domestication might have formed the genetic structure of α-glucoside metabolism on this mannequin eukaryote [122]. Supporting this notion, AGT1 homologs will be readily detected in publicly obtainable Saccharomyces genomes, whereas progress on maltotriose—a sugar transported by AGT1 however not most different maltose transporters—is extraordinarily uncommon [62]. A notable exception is Saccharomyces jurei, the primary wild Saccharomyces reported to develop on maltotriose, which comprises a transparent homolog of AGT1 that requires intensive hunger or depletion of fermentable carbon sources for its induction [123]. Whether or not the group and regulation of AGT1 in Holarctic S. eubayanus symbolize a derived or ancestral state, it created a paradigm whereby a transition between ploidy states—and thereby cell varieties—was the adaptive step conferring the best improve in health amongst advanced genotypes we examined. The exact mutational occasion or occasions underlying this adaptive step stay unclear; nevertheless, with estimates of the speed of single-chromosome loss in S. cerevisiae starting from roughly 10−4 to 10−8 per technology [8,124127], sequential lack of 14 to fifteen chromosomes is extremely inconceivable in our system. A programmed transition from diploid to haploid throughout meiosis is an integral a part of the budding yeast life cycle; thus, we suspect that uncommon sporulation occasions—maybe triggered by the biking nutrient availability in our batch-style ALE—enabled haploids to come up.

Remarkably, there’s a robust parallel to the rewiring of carbon metabolism to cell sort management in sure domesticated strains of Saccharomyces cerevisiae. Diastatic strains (typically known as Saccharomyces cerevisiae var. diastaticus) are characterised by their hyperattenuation, which is attributable to the presence of a novel extracellular glucoamylase, encoded by STA1 [128]. STA1 is a chimeric gene, created by the fusion of the sporulation-specific intracellular glucoamylase gene SGA1 with the promoter and parts of the coding sequence of FLO11, which encodes a flocculin concerned in filamentous progress that’s topic to cell type-specific regulation [129134]. Resulting from this gene fusion, STA1 is expressed in a cell type-specific method, and its regulation integrates catabolite repression by glucose and direct activation by Tec1p [135137]. The cell sort dependence mediated by Tec1p on this case might have precipitated choice for haploidy amongst diastatic strains of the Beer 2 clade of S. cerevisiae [15,138], which lack the clade-specific AGT1 allele on the MAL1 locus [121] and due to this fact should hydrolyze higher-order maltodextrins extracellularly.

STA1 in diastatic brewing strains, AGT1 in our ALE strains, and genes associated to pathogenesis throughout fungi have undoubtedly skilled intense bouts of choice, and plainly ploidy and cell sort modifications could also be a typical technique of adapting, a minimum of in microbial eukaryotes which have this flexibility. Right here, we’ve proven that placing, fast, and strange ploidy evolution in a wild yeast is related to the mixing of regulatory inputs from metabolism and cell-type networks on the AGT1 promoter. Our outcomes thus present compelling perception into the premise of a ploidy health impact in fungi.

How generalizable would possibly these rules be? Given the evolutionary lability of ploidy, its hyperlink to cell sort, and proof for interactions between cell sort and conditionally adaptive traits in different fungal programs, we suggest that environment- and genotype-specific regulatory nuances would possibly play a broad function in shaping each the extant range of fungal ploidy states and the conflicting, and sometimes cryptic, ploidy and cell-type evolution seen in programs experiencing intense choice. This view argues that interactions between cell varieties, ploidy states, and conditionally adaptive traits could also be widespread throughout fungal evolution and will affect fungal life cycles greater than is presently appreciated.

Supplies and strategies

Strains, plasmids, and cultivation circumstances

Strains, oligonucleotides, and plasmids used on this work are listed in S1 and S2 Tables. Yeast strains have been propagated on wealthy YPD medium (10 g/L yeast extract, 20 g/L peptone, 20 g/L glucose, with 18 g/L agar added for plates), Artificial Full medium with maltose or glucose (5 g/L ammonium sulfate, 1.7 g/L Yeast Nitrogen base, 2 g/L drop out combine, 20 g/L maltose or glucose, pH = 5.8, with 18 g/L agar added for plates), or Minimal Medium (5 g/L ammonium sulfate, 1.7 g/L Yeast Nitrogen base, 10 g/L maltose or glucose, pH = 5.8) at room temperature. Yeast strains and ALE populations have been saved in 15% glycerol at −80° for long-term storage. For supplementation with medicine, 1 g/L glutamic acid was substituted for ammonium sulfate in SC media. G418, Hygromycin B, and Nourseothricin (CloNAT) have been added to media at closing concentrations of 400 mg/L, 300 mg/L, and 50 mg/L, respectively. Transformation of S. eubayanus was carried out through a modified PEG-LiAc methodology [139] as beforehand described [64]. Restore templates for homologous recombination have been generated by PCR utilizing Phusion polymerase (NEB) and purified genomic DNA as template or Taq polymerase (NEB) and purified plasmid as template per the producer’s directions, adopted by purification with QiaQuick or MinElute spin columns (Qiagen). For CRISPR-mediated transformations, pXIPHOS vectors [111] expressing Cas9 and a target-specific sgRNA have been co-transformed into strains with double-stranded restore templates. Multi-fragment restore templates have been assembled by overlap extension PCR with Phusion polymerase or co-transformed as a number of linear fragments with 80 bp overlapping homology for in vivo recombination. Following transformation, yeast cells have been plated to YPD for restoration and replica-plated to medium containing the suitable antibiotic for choice after 24 to 36 h. Gene deletions and knock-ins have been verified by colony PCR and Sanger sequencing.

Plasmids have been propagated in E. cloni 10G cells (Lucigen) and purified utilizing the ZR miniprep package (Zymo Analysis). sgRNAs for CRISPR/Cas9-mediated engineering have been designed utilizing CRISpy-pop [140], obtained as single-stranded 60-mers from Built-in DNA Applied sciences, inserted into NotI-digested pXIPHOS vectors utilizing HiFi meeting (NEB), and verified by Sanger sequencing.

MAT locus genotyping

We used a multiplex colony PCR with Taq polymerase (NEB) and oligos oHJC120, oHJC121, and oHJC122 to genotype the MAT locus of strains following tetrad dissection, mating sort engineering, and for estimating the frequency of haploids in ALE populations after plating. The multiplex response provides rise to MATa– and MATα-specific amplicons of differing dimension, which have been resolved on 2% agarose gels. All response circumstances have been per the producer’s directions and have been carried out alongside controls (diploid MATa/MATα; haploid MATa; haploid MATα; no enter DNA). We discarded any experiment the place the controls didn’t produce the anticipated amplicons (or lack thereof). To estimate the frequency of haploids in populations, we screened a complete of 55 to 56 single colonies throughout 4 impartial platings of every inhabitants. We notice that this strategy can not formally distinguish between cells of various ploidies with uncommon aberrant MAT locus composition (e.g., diploid MATa/MATa will generate the identical amplicon sample as haploid MATa; lack of MAT locus heterozygosity in diploid S. cerevisiae has been estimated to happen at a fee of two × 10−5 per cell per technology [20]). As well as, this S. eubayanus background is homothallic, that means that any diploid colony recovered following plating would possibly symbolize a haploid cell within the experimental inhabitants maintained in liquid medium. The speed of mating sort switching and clone-mate selfing on stable medium is probably going orders of magnitude increased than lack of MAT locus heterozygosity [14,143]; thus, our PCR-based estimates of haploid frequency could also be conservative.

DNA sequencing

To acquire excessive molecular weight genomic DNA from wild-type pressure yHRVM108, 2 single colonies have been every inoculated in 90 mL YPD and grown to mid-log section (OD600 = 0.5), harvested by centrifugation, washed with water, and resuspended in 5 mL DTT buffer (1 M sorbitol, 25 mM EDTA, 50 mM DTT). Cells have been DTT-treated for 15 min at 30° with light agitation, pelleted, washed with 1 M sorbitol, and resuspended in 1 mL 1 M sorbitol with 0.2 mg 100T Zymolyase. Cells have been spheroplasted for 30 min at 30° with light agitation, then pelleted. The pellet was gently resuspended in 450 μL EB (Qiagen) with out pipetting and handled with 50 μL RNAse A (10 μg/mL) for two h at 37°, and 55 μL 10% SDS was added, and the combination was incubated for an additional hour at 37° with light agitation to lyse spheroplasts. DNA was extracted by the phenol/chloroform methodology and precipitated by addition of 1 mL 100% ethanol and in a single day incubation at −80°. Precipitated DNA was pelleted, washed twice with 70% ethanol, dried briefly, and gently resuspended in 100 μL TE buffer at room temperature with out pipetting for two h. DNA was quantified utilizing the Qubit dsDNA BR package (Thermo Fisher Scientific), and purity was assessed by Nanodrop (Thermo Fisher Scientific).

DNA focus was adjusted to 50 ng/μL, and seven.5 μg genomic DNA was subjected to SPRI dimension choice with Agencort AMPure XP beads in customized buffer following the beneficial protocol from Oxford Nanopore Applied sciences; 1 μg size-selected DNA was ready for sequencing utilizing the SQK-LSK109 ligation package (Oxford Nanopore Applied sciences), and roughly 40 fmol library was loaded on a single FLO-FLG001 flowcell. Basecalling was carried out with Guppy v3.2.1. ONT sequencing yielded 885.1 Mb of base-called reads passing high quality filtering, for roughly 74-fold genomic protection.

We ready genomic DNA for Illumina sequencing from the wild-type pressure and advanced isolates as described beforehand [25]. Strains have been streaked to single colonies, and colonies have been inoculated to three mL YPD medium and grown to saturation earlier than assortment for DNA extraction. Purified DNA was quantified by Qubit dsDNA BR assay (Thermo Fisher Scientific), and purity and high quality have been assessed by Nanodrop (Thermo Fisher Scientific) and agarose gel electrophoresis. Library preparation and Illumina sequencing of the wild-type pressure and clonal advanced isolates have been carried out by the DOE Joint Genome Institute. Paired-end libraries have been sequenced on a NovaSeq S4 with 150 bp reads, yielding a mean of 8.47 million reads per pattern. For the wild-type pressure yHRVM108, we additionally built-in publicly obtainable reads (SRA: SRX1317977) from a earlier research [49].

To trace the frequency of aneuploidies within the ALE populations, whole populations cryopreserved at −80°C in 15% glycerol have been gently thawed, and 20 μL was inoculated on to 2 mL SC-2% Maltose and grown to saturation. DNA was extracted as described above, and libraries have been ready utilizing the NEBNext Extremely II FS package (New England Biolabs) per the producer’s directions. Libraries have been sequenced on an Illumina NovaSeq on the College of Wisconsin-Madison Biotechnology Heart with paired-end 150 bp reads, yielding a mean of two.23 million reads per pattern. All uncooked reads have been processed utilizing Trimmomatic v0.3 [144] to take away adapter sequences and low-quality bases.

Genome meeting, annotation, and evaluation

Canu v1.9 [145] was used to generate a genome meeting with Nanopore sequencing reads from the wild-type pressure, which was subsequently polished with Illumina reads utilizing 3 rounds of Pilon v1.23 [146]. The genome meeting was annotated utilizing the Yeast Genome Annotation Pipeline [147]. We mapped every predicted gene to its S. cerevisiae homolog utilizing BLASTp v2.9 [148]. QUAST v5.0.2 [149] and BUSCO v3.1.0 [150] have been used to evaluate genome completeness, and chromosomes within the meeting have been assigned numbers similar to the S. eubayanus sort pressure reference genome [58,151] utilizing MUMmer v3.2.3 [152] and BLASTn v2.9. BWA v0.7.12 [153] and samtools v1.9 [154] have been used to map brief reads from all sequenced strains and inhabitants samples to the meeting, and BEDtools v2.27 [155] was used to name sequencing depth. Protection throughout the genome of every pressure or inhabitants was analyzed in R and assessed by manually inspecting protection plots of every chromosome. Closing genome-wide Illumina-sequencing depths for every pressure have been 200.7-fold (wild-type), 106.1-fold (advanced clone 1), and 84.2-fold (advanced clone 2); sequencing depths for inhabitants samples ranged from 14.3- to 109.3-fold (median: 34.3). We used FreeBayes v1.3.1 [156] to name variants in every pressure, requiring a minimal protection depth of 10 to report a place, and manually inspected putative variants in IGV [157]. To annotate predicted transcription issue binding websites within the promoter of AGT1, we used the 700 bp upstream of the beginning codon as a question for YEASTRACT+ [158].

RNA sequencing and evaluation

mRNA enrichment, library preparation, and Illumina sequencing have been carried out by the DOE Joint Genome Institute for organic triplicate samples for every pressure and situation. Paired-end libraries have been sequenced on a NovaSeq S4 with 150 bp reads, yielding a mean of 23.23 million reads per pattern. Uncooked mRNA-seq reads have been processed with BBduk (https://sourceforge.web/tasks/bbmap/) to take away adapters and low-quality sequences, leading to a mean of twenty-two.89 million surviving reads per library. Filtered reads have been mapped to the wild-type pressure meeting with HISAT2 v2.1 [159] with common mapping charges of 98.5% per pattern.

HTSeq-count v0.11.1 [160] was used to generate counts at annotated genes, which have been handed to DESeq2 v1.30.1 [161,162] for additional evaluation. We faraway from evaluation a single library from an advanced isolate grown in maltose, as guide inspection of normalized gene expression values revealed that this pattern had stochastically misplaced the ChrXV aneuploidy. This diminished our energy to detect statistically important variations in expression for that particular advanced isolate. All different samples from advanced isolates remained aneuploid in each circumstances. We thought of DEGs between circumstances and genotypes with expression modifications of higher than or equal to 2-fold in both course and Benjamini–Hochberg adjusted p-values of lower than or equal to 0.01 (false discovery fee of 1%). Full differential expression evaluation outcomes will be present in S4 Desk. To check expression ranges of single genes, we used size-normalized counts from DESeq2, that are extra strong for this objective than different normalization strategies [163165]. We outlined subtelomeric genes as these falling inside 20 kb of the tip of a contig, which represented whole chromosomes in our meeting (except the two-contig ChrXII, for which we thought of genes inside 20 kb of the telomeric contig ends, not the ends containing rDNA repeats). This classification is corresponding to or extra conservative than these used beforehand [108,166]. GOrilla [167] was used to determine enriched gene ontology (GO) phrases in gene units of curiosity; we used S. cerevisiae GO annotations and specified all predicted genes in our annotation because the background set towards which to check for enrichment. Statistics and knowledge visualization have been carried out in R.

RT-qPCR

For the experiment proven in S9 Fig, 100 ng complete RNA was used as enter for the Luna Common One-Step RT-qPCR package (New England Biolabs), with biking and knowledge acquisition carried out on an Utilized Biosystems 7500 Actual-Time System (Thermo Fisher Scientific). Relative expression of AGT1 was analyzed utilizing the ΔΔCT methodology with normalization to ACT1 and ARP2 [168].

Ploidy willpower

Stream cytometry-based ploidy willpower was carried out as described beforehand [7], besides that we sampled asynchronous cultures. Briefly, we fastened mid-log cultures of every question, handled fastened cells with RNAse A and Proteinase Okay, and stained DNA with Sytox Inexperienced (Thermo Fisher Scientific). Haploid and diploid S. cerevisiae strains have been included in all experiments as controls. For clonal strains (S. cerevisiae controls, ancestral S. eubayanus, and advanced isolates), queries have been streaked to single colonies, and impartial colonies have been picked for ploidy evaluation. For inhabitants samples, whole populations cryopreserved at −80°C in 15% glycerol have been gently thawed, and roughly 50 μL was inoculated on to 1 mL SC-2% maltose. Cells have been harvested and glued in early log section after a minimal of two doublings, and 10,000 cells have been sampled for every question on an Attune NxT circulation cytometer (Thermo Fisher Scientific). Evaluation was carried out in FlowJo v10.

Health assays

Aside from experiments in wealthy medium proven in Fig 2A, the circumstances for health assays have been designed to imitate the unique ALE circumstances [64]. Briefly, this regime consisted of culturing in 1 mL SC medium with 2% maltose and 0.1% glucose (hereafter, “competitors medium”) with semiweekly 1:10 dilutions into new competitors medium. Question genotypes have been straight competed towards a typical competitor in co-culture. The competitor was a haploid within the ancestral S. eubayanus pressure background except a constitutively expressed GFP utilizing a TEF1 promoter and ADH1 terminator from S. cerevisiae and ste12 deletion (MATa hoΔ::PScTEF1-yEGFP-TScADH1-kanMX ste12Δ::natMX). We selected a ste12 deletion to forestall any interplay with rivals expressing MATα. Strains have been streaked to single colonies on YPD containing antibiotic as applicable, precultured in competitors medium for 3 days, blended in roughly equal query-to-competitor ratios (besides the place we diminished the competitor ratio towards less-fit question genotypes), sampled into chilly 1× PBST for circulation cytometry of time level 0, and inoculated into 1 mL competitors medium at an preliminary OD600 of roughly 0.1. At every switch, competitions have been sampled into chilly 1× PBST for circulation cytometry, and the optical density of every replicate was measured to calculate the variety of generations. Competitions in wealthy medium have been carried out in the identical method, albeit that preculturing and propagation have been in sterile-filtered YPD in 2 mL quantity with day by day dilutions of 1:100. For each competitors regimes, we sampled 13,000 cells per replicate and time level on an Attune NxT circulation cytometer (Thermo Fisher Scientific) to quantify the abundance of competitor (fluorescent) and question (non-fluorescent) cells, which all the time clearly fashioned distinct populations. Evaluation was carried out in FlowJo v10. Health was calculated as the choice coefficient, obtained by regressing the pure log ratio of question to competitor towards the variety of generations. To investigate the consequences of ploidy, mating sort, and cell sort (diploid-like and haploid-like) on the panel of strains proven in Fig 3, we used a number of linear regression with measured health because the response and ploidy, mating sort, and cell sort as categorical predictors with 2 ranges every (for mating sort, we grouped by whether or not genotypes expressed any mating type-specific genes, or none). All statistical analyses and visualization have been carried out in R.

Supporting data

S3 Fig. Ploidy variation throughout the adaptive evolution experiment.

(a) Smoothed histograms of mobile DNA content material for asynchronous haploid (high panel) and diploid (center panel) S. cerevisiae (Sc) controls and the wild-type S. eubayanus (Se Anc.) pressure (backside panel, reproduced from the identical knowledge as in Fig 1). (b) Histograms for population-level samples from each ALE replicates (grey) and clonal isolates from every inhabitants (pink shades). For clones, the two histograms symbolize outcomes from impartial experiments; the underside panel for every (darkish pink) is identical knowledge displayed in Fig 1. For inhabitants samples, panels are organized from high to backside with growing time and variety of ALE generations, representing roughly 50 technology intervals from 50–350. The underside panel for every inhabitants represents the terminal time level from which the tailored clones have been remoted and from which we quantitatively assessed haploid frequency. (c) Fraction of haploids within the terminal time level of every ALE inhabitants assayed by MAT locus PCR genotyping. Factors and bars present the imply and customary error of 4 experiments. The information underlying this determine will be present in S1 Information.

https://doi.org/10.1371/journal.pbio.3001909.s003

(TIF)

S4 Fig. Temporal dynamics of ploidy variants in evolving populations.

(a) Aneuploidy frequency throughout the adaptive evolution experiment. The relative copy variety of every chromosome, inferred from sequencing depth, is plotted for whole-population samples from the ALE experiment from roughly 50 technology intervals. The trajectories of aneuploidies that reached excessive frequencies are coloured; all different chromosomes are black. The time factors are the identical as these sampled to assay ploidy states (S3B Fig). (b) Aneuploidies in whole-population samples are plotted towards generations as in (a), however they’re rescaled to frequency per haploid genome. The obvious frequency of haploids in every inhabitants from the identical time factors is plotted as inexperienced traces and was calculated from the circulation cytometry knowledge proven in S3B Fig. The information underlying this determine will be present in S1 Information.

https://doi.org/10.1371/journal.pbio.3001909.s004

(TIF)

S7 Fig. Further gene expression comparisons.

(a) Boxplots present log2-transformed fold modifications (LFC) of gene expression on glucose (as an alternative of maltose, as in Fig 4) in advanced haploids in comparison with the wild-type pressure for genes on aneuploid ChrXV (n = 370) and subtelomeric genes (n = 200). (b) Boxplots present LFCs of gene expression in maltose in comparison with glucose for ChrXV genes in every advanced haploid. Whiskers lengthen to 1.5× the interquartile vary. Strains join the y-axis coordinates of the identical gene in every advanced isolate; axes are scaled such that an occasional outlier is truncated from the plot house for a single pressure. AGT1 expression is plotted as pink dots and features, and black dashed traces point out the null expectation for expression values between strains (a) or equal expression between circumstances (b). The information underlying this determine will be present in S1 Information.

https://doi.org/10.1371/journal.pbio.3001909.s007

(TIF)

References

  1. 1.
    Parfrey LW, Lahr DJG, Katz LA. The Dynamic Nature of Eukaryotic Genomes. Mol Biol Evol. 2008;25:787–794. pmid:18258610
  2. 2.
    Albertin W, Marullo P. Polyploidy in fungi: evolution after whole-genome duplication. Proc R Soc B Biol Sci. 2012;279:2497–2509. pmid:22492065
  3. 3.
    Gregory TR, Mable BK. Polyploidy in Animals. The Evolution of the Genome. Elsevier. 2005:427–517.
  4. 4.
    Wendel JF. The wondrous cycles of polyploidy in crops. Am J Bot. 2015;102:1753–1756. pmid:26451037
  5. 5.
    Otto SP, Whitton J. POLYPLOID INCIDENCE AND EVOLUTION. Annu Rev Genet. 2000;34:401–437. pmid:11092833
  6. 6.
    Ohno S. Evolution by Gene Duplication. Springer Berlin Heidelberg; 1970.
  7. 7.
    Fisher KJ, Buskirk SW, Vignogna RC, Marad DA, Lang GI. Adaptive genome duplication impacts patterns of molecular evolution in Saccharomyces cerevisiae. PLoS Genet. 2018;14:e1007396. pmid:29799840
  8. 8.
    Sharp NP, Sandell L, James CG, Otto SP. The genome-wide fee and spectrum of spontaneous mutations differ between haploid and diploid yeast. Proc Natl Acad Sci U S A. 2018;115:E5046–E5055. pmid:29760081
  9. 9.
    Marad DA, Buskirk SW, Lang GI. Altered entry to useful mutations slows adaptation and biases fastened mutations in diploids. Nat Ecol Evol. 2018;2:882–889. pmid:29581586
  10. 10.
    Gerstein AC, Cleathero LA, Mandegar MA, Otto SP. Haploids adapt sooner than diploids throughout a spread of environments. J Evol Biol. 2011;24:531–540. pmid:21159002
  11. 11.
    Zeyl C, Vanderford T, Carter M. An Evolutionary Benefit of Haploidy in Massive Yeast Populations. Science. 2003;299:555–558. pmid:12543972
  12. 12.
    Aggeli D, Marad DA, Liu X, Buskirk SW, Levy SF, Lang GI. Overdominant and partially dominant mutations drive clonal adaptation in diploid Saccharomyces cerevisiae. Genetics. 2022:221. pmid:35435209
  13. 13.
    Johnson MS, Gopalakrishnan S, Goyal J, Dillingham ME, Bakerlee CW, Humphrey PT, et al. Phenotypic and molecular evolution throughout 10,000 generations in laboratory budding yeast populations. Elife. 2021;10:1–28. pmid:33464204
  14. 14.
    Herskowitz I. Life cycle of the budding yeast Saccharomyces cerevisiae. Microbiol Rev. 1988;52:536–553. pmid:3070323
  15. 15.
    Peter J, De Chiara M, Friedrich A, Yue J-X, Pflieger D, Bergström A, et al. Genome evolution throughout 1,011 Saccharomyces cerevisiae isolates. Nature. 2018;556:339–344. pmid:29643504
  16. 16.
    Katz Ezov T, Boger-Nadjar E, Frenkel Z, Katsperovski I, Kemeny S, Nevo E, et al. Molecular-Genetic Biodiversity in a Pure Inhabitants of the Yeast Saccharomyces cerevisiae From “Evolution Canyon”: Microsatellite Polymorphism. Ploidy and Controversial Sexual Standing. Genetics. 2006;174:1455–1468. pmid:16980391
  17. 17.
    Gerstein AC, Chun H-JE, Grant A, Otto SP. Genomic Convergence towards Diploidy in Saccharomyces cerevisiae. PLoS Genet. 2006;2:e145. pmid:17002497
  18. 18.
    Gerstein AC, Sharp NP. The inhabitants genetics of ploidy change in unicellular fungi. FEMS Microbiol Rev. 2021;006:1–20. pmid:33503232
  19. 19.
    Todd RT, Forche A, Selmecki A. Ploidy Variation in Fungi: Polyploidy, Aneuploidy, and Genome Evolution. Microbiol Spectr. 2017:5. pmid:28752816
  20. 20.
    Harari Y, Ram Y, Rappoport N, Hadany L, Kupiec M. Spontaneous Modifications in Ploidy Are Widespread in Yeast. Curr Biol. 2018;28:825–835.e4. pmid:29502947
  21. 21.
    Mable BK, Otto SP. Masking and purging mutations following EMS remedy in haploid, diploid and tetraploid yeast (Saccharomyces cerevisiae). Genet Res (Camb). 2001;77:9–26. pmid:11279834
  22. 22.
    Venkataram S, Dunn B, Li Y, Agarwala A, Chang J, Ebel ER, et al. Improvement of a Complete Genotype-to-Health Map of Adaptation-Driving Mutations in Yeast. Cell. 2016;166:1585–1596.e22. pmid:27594428
  23. 23.
    Zörgö E, Chwialkowska Okay, Gjuvsland AB, Garré E, Sunnerhagen P, Liti G, et al. Historic Evolutionary Commerce-Offs between Yeast Ploidy States. PLoS Genet. 2013;9:e1003388. pmid:23555297
  24. 24.
    Gerstein AC, Lim H, Berman J, Hickman MA. Ploidy tug-of-war: Evolutionary and genetic environments affect the speed of ploidy drive in a human fungal pathogen. Evolution (NY). 2017;71:1025–1038. pmid:28195309
  25. 25.
    Shen XX, Opulente DA, Kominek J, Zhou X, Steenwyk JL, Buh KV, et al. Tempo and Mode of Genome Evolution within the Budding Yeast Subphylum. Cell. 2018;175:1533–1545.e20. pmid:30415838
  26. 26.
    Molinet J, Urbina Okay, Villegas C, Abarca V, Oporto CI, Villarreal P, et al. A Saccharomyces eubayanus haploid useful resource for analysis research. Sci Rep. 2022;12:5976. pmid:35396494
  27. 27.
    Galgoczy DJ, Cassidy-Stone A, Llinás M, O’Rourke SM, Herskowitz I, DeRisi JL, et al. Genomic dissection of the cell-type-specification circuit in Saccharomyces cerevisiae. Proc Natl Acad Sci U S A. 2004;101:18069–18074. pmid:15604142
  28. 28.
    Haber JE. Mating-type genes and MAT switching in Saccharomyces cerevisiae. Genetics. 2012;191:33–64. pmid:22555442
  29. 29.
    Brisse S, Pannier C, Angoulvant A, de Meeus T, Diancourt L, Faure O, et al. Uneven Distribution of Mating Sorts amongst Genotypes of Candida glabrata Isolates from Scientific Samples. Eukaryot Cell. 2009;8:287–295. pmid:19151326
  30. 30.
    Muñoz JF, Gade L, Chow NA, Loparev VN, Juieng P, Berkow EL, et al. Genomic insights into multidrug-resistance, mating and virulence in Candida auris and associated rising species. Nat Commun. 2018;9:1–13. pmid:30559369
  31. 31.
    Vanheule A, De Boevre M, Moretti A, Scauflaire J, Munaut F, De Saeger S, et al. Genetic Divergence and Chemotype Range within the Fusarium Head Blight Pathogen Fusarium poae. Toxins (Basel). 2017;9:255. pmid:28832503
  32. 32.
    Guo L, Blatt AZ, Geiser DM, del Mar Jiménez-Gasco M, Kuldau GA. Mating sort and spore killing characterization of Fusarium verticillioides strains. Mycol Prog. 2015;14:16.
  33. 33.
    Kwon-Chung KJ, Bennett JE. DISTRIBUTION OF α AND α MATING TYPES OF CRYPTOCOCCUS NEOFORMANS AMONG NATURAL AND CLINICAL ISOLATES. Am J Epidemiol. 1978;108:337–340. pmid:364979
  34. 34.
    Hull CM, Heitman J. Genetics of Cryptococcus neoformans. Annu Rev Genet. 2002;36:557–615. pmid:12429703
  35. 35.
    Desjardins CA, Giamberardino C, Sykes SM, Yu CH, Tenor JL, Chen Y, et al. Inhabitants genomics and the evolution of virulence within the fungal pathogen Cryptococcus neoformans. Genome Res. 2017;27:1207–1219. pmid:28611159
  36. 36.
    Alvarez-Perez S, Blanco JL, Alba P, Garcia ME. Mating sort and invasiveness are considerably related in Aspergillus fumigatus. Med Mycol. 2010;48:273–277. pmid:19634068
  37. 37.
    Cheema MS, Christians JK. Virulence in an insect mannequin differs between mating varieties in Aspergillus fumigatus. Med Mycol. 2011;49:202–207. pmid:20818921
  38. 38.
    Zheng Q, Hou R, Juanyu Z, Ma J, Wu Z, Wang G, et al. The MAT Locus Genes Play Totally different Roles in Sexual Replica and Pathogenesis in Fusarium graminearum. PLoS ONE. 2013;8:e66980. pmid:23826182
  39. 39.
    Xu W, Liang G, Peng J, Lengthy Z, Li D, Fu M, et al. The affect of the mating sort on virulence of Mucor irregularis. Sci Rep. 2017;7:10629. pmid:28878325
  40. 40.
    Forgács L, Borman AM, Prépost E, Tóth Z, Kardos G, Kovács R, et al. Comparability of in vivo pathogenicity of 4 Candida auris clades in a neutropenic bloodstream an infection murine mannequin. Emerg Microbes Infect. 2020;9:1160–1169. pmid:32486923
  41. 41.
    Kwon-Chung KJ, Edman JC, Wickes BL. Genetic affiliation of mating varieties and virulence in Cryptococcus neoformans. Infect Immun. 1992;60:602–605. pmid:1730495
  42. 42.
    Nielsen Okay, Marra RE, Hagen F, Boekhout T, Mitchell TG, Cox GM, et al. Interplay Between Genetic Background and the Mating-Sort Locus in Cryptococcus neoformans Virulence Potential. Genetics. 2005;171:975–983. pmid:15965241
  43. 43.
    Nielsen Okay, Cox GM, Litvintseva AP, Mylonakis E, Malliaris SD, Benjamin DK, et al. Cryptococcus neoformans α Strains Preferentially Disseminate to the Central Nervous System throughout Coinfection. Infect Immun. 2005;73:4922–4933.
  44. 44.
    Lin X, Nielsen Okay, Patel S, Heitman J. Influence of Mating Sort, Serotype, and Ploidy on the Virulence of Cryptococcus neoformans. Infect Immun. 2008;76:2923–2938. pmid:18426889
  45. 45.
    Lin X, Huang JC, Mitchell TG, Heitman J. Virulence Attributes and Hyphal Progress of C. neoformans Are Quantitative Traits and the MATα Allele Enhances Filamentation. PLoS Genet. 2006;2:e187. pmid:17112316
  46. 46.
    Solar S, Coelho MA, David-Palma M, Priest SJ, Heitman J. The Evolution of Sexual Replica and the Mating-Sort Locus: Hyperlinks to Pathogenesis of Cryptococcus Human Pathogenic Fungi. Annu Rev Genet. 2019;53:417–444. pmid:31537103
  47. 47.
    Libkind D, Hittinger CT, Valeŕio E, Gonçalves C, Dover J, Johnston M, et al. Microbe domestication and the identification of the wild genetic inventory of lager-brewing yeast. Proc Natl Acad Sci U S A. 2011;108:14539–14544. pmid:21873232
  48. 48.
    Nespolo RF, Villarroel CA, Oporto CI, Tapia SM, Vega-Macaya F, Urbina Okay, et al. An Out-of-Patagonia migration explains the worldwide range and distribution of Saccharomyces eubayanus lineages. PLOS Genet. 2020;16:e1008777. pmid:32357148
  49. 49.
    Langdon QK, Peris D, Eizaguirre JI, Opulente DA, Buh KV, Sylvester Okay, et al. Postglacial migration formed the genomic range and world distribution of the wild ancestor of lager-brewing hybrids. PLOS Genet. 2020;16:e1008680. pmid:32251477
  50. 50.
    Peris D, Sylvester Okay, Libkind D, Gonçalves P, Sampaio JP, Alexander WG, et al. Inhabitants construction and reticulate evolution of Saccharomyces eubayanus and its lager-brewing hybrids. Mol Ecol. 2014;23:2031–2045. pmid:24612382
  51. 51.
    Peris D, Langdon QK, Moriarty RV, Sylvester Okay, Bontrager M, Charron G, et al. Advanced Ancestries of Lager-Brewing Hybrids Had been Formed by Standing Variation within the Wild Yeast Saccharomyces eubayanus. PLoS Genet. 2016;12:1–20. pmid:27385107
  52. 52.
    Eizaguirre JI, Peris D, Rodríguez ME, Lopes CA, De Los RP, Hittinger CT, et al. Phylogeography of the wild Lager-brewing ancestor (Saccharomyces eubayanus) in Patagonia. Environ Microbiol. 2018;20:3732–3743. pmid:30105823
  53. 53.
    Bing J, Han PJ, Liu WQ, Wang QM, Bai FY. Proof for a far east asian origin of lager beer yeast. Curr Biol Elsevier. Might 19 2014:R380–R381. pmid:24845661
  54. 54.
    Gibson B, Geertman JMA, Hittinger CT, Krogerus Okay, Libkind D, Louis EJ, et al. New yeasts-new brews: Trendy approaches to brewing yeast design and improvement. FEMS Yeast Res. 2017;17:1–13. pmid:28582493
  55. 55.
    Gorter De Vries AR, Voskamp MA, Van Aalst ACA, Kristensen LH, Jansen L, Van Den Broek M, et al. Laboratory evolution of a Saccharomyces cerevisiae × S. eubayanus hybrid below simulated lager-brewing circumstances. Entrance Genet. 2019:10. pmid:31001314
  56. 56.
    Diderich JA, Weening SM, Van Den Broek M, Pronk JT, Daran JMG. Collection of Pof- saccharomyces eubayanus variants for the development of S. cerevisiae × S. eubayanus hybrids with diminished 4-Vinyl guaiacol formation. Entrance Microbiol. 2018;9:1–17. pmid:30100898
  57. 57.
    Brouwers N, Gorter de Vries AR, van den Broek M, Weening SM, Elink Schuurman TD, Kuijpers NGA, et al. In vivo recombination of Saccharomyces eubayanus maltose-transporter genes yields a chimeric transporter that allows maltotriose fermentation. PLoS Genet. 2019;15:e1007853. pmid:30946741
  58. 58.
    Brickwedde A, Brouwers N, van den Broek M, Gallego Murillo JS, Fraiture JL, Pronk JT, et al. Structural, physiological and regulatory evaluation of maltose transporter genes in Saccharomyces eubayanus CBS 12357T. Entrance Microbiol. 2018;9:1–18. pmid:30147677
  59. 59.
    Baker EP, Peris D, Moriarty RV, Li XC, Fay JC, Hittinger CT. Mitochondrial DNA and temperature tolerance in lager yeasts. Sci Adv. 2019:5. pmid:30729163
  60. 60.
    He Y, Dong J, Yin H, Zhao Y, Chen R, Wan X, et al. Wort composition and its impression on the flavour-active increased alcohol and ester formation of beer–a evaluate. J Inst Brew. 2014;120:157–163.
  61. 61.
    Fox G. Infrared spectral evaluation of sugar profiles of worts from various grist to liquor ratios utilizing infusion and ramping mash kinds. J Inst Brew. 2016;122:437–445.
  62. 62.
    Peris D, Ubbelohde EJ, Kuang MC, Kominek J, Langdon QK, Adams M, et al. Macroevolutionary range of traits and genomes within the mannequin yeast genus Saccharomyces. Nat Commun. 2023;14:690. pmid:36755033
  63. 63.
    Brouwers N, Brickwedde A, Gorter de Vries AR, van den Broek M, Weening SM, van den Eijnden L, et al. Himalayan saccharomyces eubayanus genome sequences reveal genetic markers explaining heterotic maltotriose consumption by saccharomyces pastorianus hybrids. Appl Environ Microbiol. 2019:85. pmid:31519660
  64. 64.
    Baker EP, Hittinger CT. Evolution of a novel chimeric maltotriose transporter in Saccharomyces eubayanus from mother or father proteins unable to carry out this operate. PLoS Genet. 2019;15:e1007786. pmid:30946740
  65. 65.
    Bergin SA, Allen S, Hession C, Cinnéide EÓ, Ryan A, Byrne KP, et al. Identification of European isolates of the lager yeast mother or father Saccharomyces eubayanus. FEMS Yeast Res. 2022;22:1–9. pmid:36473696
  66. 66.
    Buskirk SW, Peace RE, Lang GI. Hitchhiking and epistasis give rise to cohort dynamics in adapting populations. Proc Natl Acad Sci U S A. 2017;114:8330–8335. pmid:28720700
  67. 67.
    Lang GI, Rice DP, Hickman MJ, Sodergren E, Weinstock GM, Botstein D, et al. Pervasive genetic hitchhiking and clonal interference in forty evolving yeast populations. Nature. 2013;500:571–574. pmid:23873039
  68. 68.
    Aggeli D, Li Y, Sherlock G. Modifications within the distribution of health results and adaptive mutational spectra following a single first step in the direction of adaptation. Nat Commun. 2021;12:1–14. pmid:34465770
  69. 69.
    Fisher KJ, Vignogna RC, Lang GI. Overdominant Mutations Prohibit Adaptive Lack of Heterozygosity at Linked Loci. Genome Biol Evol. 2021:13. pmid:34363476
  70. 70.
    James TY, Michelotti LA, Glasco AD, Clemons RA, Powers RA, James ES, et al. Adaptation by lack of heterozygosity in Saccharomyces cerevisiae clones below divergent choice. Genetics. 2019;213:665–683. pmid:31371407
  71. 71.
    Sui Y, Qi L, Wu JK, Wen XP, Tang XX, Ma ZJ, et al. Genome-wide mapping of spontaneous genetic alterations in diploid yeast cells. Proc Natl Acad Sci U S A. 2020;117:28191–28200. pmid:33106417
  72. 72.
    Smukowski Heil CS, Massive CRL, Patterson Okay, Hickey ASM, Yeh CLC, Dunham MJ. Temperature desire can bias parental genome retention throughout hybrid evolution. PLoS Genet. 2019;15:e1008383. pmid:31525194
  73. 73.
    Smukowski Heil CS, DeSevo CG, Pai DA, Tucker CM, Hoang ML, Dunham MJ. Lack of Heterozygosity Drives Adaptation in Hybrid Yeast. Mol Biol Evol. 2017;34:1596–1612. pmid:28369610
  74. 74.
    Gerstein AC, Kuzmin A, Otto SP. Loss-of-heterozygosity facilitates passage via Haldane’s sieve for Saccharomyces cerevisiae present process adaptation. Nat Commun. 2014;5:3819. pmid:24804896
  75. 75.
    Smukowski HC. Lack of Heterozygosity and Its Significance in Evolution. J Mol Evol. 2023;91:369–377. pmid:36752826
  76. 76.
    Orr HA, Otto SP. Does diploidy improve the speed of adaptation? Genetics. 1994;136:1475–1480. pmid:8013920
  77. 77.
    Selmecki AM, Maruvka YE, Richmond PA, Guillet M, Shoresh N, Sorenson AL, et al. Polyploidy can drive fast adaptation in yeast. Nature. 2015;519:349–352. pmid:25731168
  78. 78.
    Weiss RL, Kukora JR, Adams J. The connection between enzyme exercise, cell geometry, and health in Saccharomyces cerevisiae. Proc Natl Acad Sci U S A. 1975;72:794–798. pmid:1093169
  79. 79.
    Lewis WM. Nutrient Shortage as an Evolutionary Explanation for Haploidy. Am Nat. 1985;125:692–701.
  80. 80.
    Bessho Okay, Iwasa Y, Day T. The evolutionary benefit of haploid versus diploid microbes in nutrient-poor environments. J Theor Biol. 2015;383:116–129. pmid:26247141
  81. 81.
    Mable BK. Ploidy evolution within the yeast Saccharomyces cerevisiae: a check of the nutrient limitation speculation. J Evol Biol. 2001;14:157–170. pmid:29280580
  82. 82.
    Gerstein AC, Otto SP. Cryptic Health Benefit: Diploids Invade Haploid Populations Regardless of Missing Any Obvious Benefit as Measured by Commonplace Health Assays. PLoS ONE. 2011;6:e26599. pmid:22174734
  83. 83.
    Lang GI, Murray AW, Botstein D. The price of gene expression underlies a health trade-off in yeast. Proc Natl Acad Sci U S A. 2009;106:5755–5760. pmid:19299502
  84. 84.
    Galitski T, Saldanha AJ, Kinds CA, Lander ES, Fink GR. Ploidy Regulation of Gene Expression. Science. 1999;285:251–254. pmid:10398601
  85. 85.
    Wu CY, Alexander Rolfe P, Gifford DK, Fink GR. Management of Transcription by Cell Dimension. PLoS Biol. 2010;8:e1000523. pmid:21072241
  86. 86.
    Dephoure N, Hwang S, O’Sullivan C, Dodgson SE, Gygi SP, Amon A, et al. Quantitative proteomic evaluation reveals posttranslational responses to aneuploidy in yeast. Elife. 2014;3:1–27. pmid:25073701
  87. 87.
    Loehlin DW, Carroll SB. Expression of tandem gene duplicates is usually higher than twofold. Proc Natl Acad Sci U S A. 2016;113:5988–5992. pmid:27162370
  88. 88.
    Cromie GA, Tan Z, Hays M, Jeffery EW, Dudley AM. Dissecting Gene Expression Modifications Accompanying a Ploidy-Based mostly Phenotypic Change. G3 Genes|Genomes|Genetics. 2017;7:233–246. pmid:27836908
  89. 89.
    Hose J, Yong CM, Sardi M, Wang Z, Newton MA, Gasch AP. Dosage compensation can buffer copy-number variation in wild yeast. Elife. 2015:4. pmid:25955966
  90. 90.
    Torres EM, Springer M, Amon A. No present proof for widespread dosage compensation in S. cerevisiae. Elife. 2016:5. pmid:26949255
  91. 91.
    de la Cerda G-CR, Hokamp Okay, Roche F, Thompson G, Timouma S, Delneri D, et al. Aneuploidy influences the gene expression profiles in Saccharomyces pastorianus group I and II strains throughout fermentation. PLoS Genet. 2022;18:e1010149. pmid:35389986
  92. 92.
    Torres EM, Sokolsky T, Tucker CM, Chan LY, Boselli M, Dunham MJ, et al. Results of Aneuploidy on Mobile Physiology and Cell Division in Haploid Yeast. Science. 2007;317:916–924. pmid:17702937
  93. 93.
    Kaya A, Mariotti M, Tyshkovskiy A, Zhou X, Hulke ML, Ma S, et al. Molecular signatures of aneuploidy-driven adaptive evolution. Nat Commun. 2020;11:1–14. pmid:32001709
  94. 94.
    Sheltzer JM, Torres EM, Dunham MJ, Amon A. Transcriptional penalties of aneuploidy. Proc Natl Acad Sci U S A. 2012;109:12644–12649. pmid:22802626
  95. 95.
    Levine J, Tanouye L, Michels CA. The united statesMAL) is a bidirectional promotor component required for the expression of each the MAL61 and MAL62 genes of the Saccharomyces MAL6 locus. Curr Genet. 1992;22:181–189. pmid:1525871
  96. 96.
    Vidgren V, Kankainen M, Londesborough J, Ruohonen L. Identification of regulatory components within the AGT1 promoter of ale and lager strains of brewer’s yeast. Yeast. 2011;28:579–594. pmid:21755532
  97. 97.
    Hu Z, Nehlin JO, Ronne H, Michels CA. MIG1-dependent and MIG1-independent glucose regulation of MAL gene expression in Saccharomyces cerevisiae. Curr Genet. 1995;28:258–266. pmid:8529272
  98. 98.
    Kumar A. The Advanced Genetic Foundation and Multilayered Regulatory Management of Yeast Pseudohyphal Progress. Annu Rev Genet. 2021;55:1–21. pmid:34280314
  99. 99.
    Liu H, Kinds CA, Fink GR. Parts of the Yeast Pheromone Response Pathway Required for Filamentous Progress of Diploids. Science. 1993;262:1741–1744. pmid:8259520
  100. 100.
    Gavrias V, Andrianopoulos A, Gimeno CJ, Timberlake WE. Saccharomyces cerevisiae TEC1 is required for pseudohyphal progress. Mol Microbiol. 1996;19:1255–1263. pmid:8730867
  101. 101.
    Madhani HD, Fink GR. Combinatorial Management Required for the Specificity of Yeast MAPK Signaling. Science. 1997;275:1314–1317. pmid:9036858
  102. 102.
    Cullen PJ, Sprague GF. The Regulation of Filamentous Progress in Yeast. Genetics. 2012;190:23–49. pmid:22219507
  103. 103.
    Cullen PJ, Sprague GF. Glucose depletion causes haploid invasive progress in yeast. Proc Natl Acad Sci U S A. 2000;97:13619–13624. pmid:11095711
  104. 104.
    Köhler T, Wesche S, Taheri N, Braus GH, Mösch H-U. Twin Function of the Saccharomyces cerevisiae TEA/ATTS Household Transcription Issue Tec1p in Regulation of Gene Expression and Mobile Improvement. Eukaryot Cell. 2002;1:673–686. pmid:12455687
  105. 105.
    Dorrity MW, Cuperus JT, Carlisle JA, Fields S, Queitsch C. Preferences in a trait determination decided by transcription issue variants. Proc Natl Acad Sci U S A. 2018;115:E7997–E8006. pmid:30068600
  106. 106.
    Heise B, van der Felden J, Kern S, Malcher M, Brückner S, Mösch H-U. The TEA Transcription Issue Tec1 Confers Promoter-Particular Gene Regulation by Ste12-Dependent and -Impartial Mechanisms. Eukaryot Cell. 2010;9:514–531. pmid:20118212
  107. 107.
    Borneman AR, Gianoulis TA, Zhang ZD, Yu H, Rozowsky J, Seringhaus MR, et al. Divergence of Transcription Issue Binding Websites Throughout Associated Yeast Species. Science. 2007;317:815–819. pmid:17690298
  108. 108.
    Brown CA, Murray AW, Verstrepen KJ. Speedy Enlargement and Practical Divergence of Subtelomeric Gene Households in Yeasts. Curr Biol. 2010;20:895–903. pmid:20471265
  109. 109.
    Stambuk BU, Silva MA, Panek AD, Araujo PS. Lively α-glucoside transport in Saccharomyces cerevisiae. FEMS Microbiol Lett. 1999;170:105–110. pmid:9919658
  110. 110.
    Nalabothu RL, Fisher KJ, LaBella AL, Meyer TA, Opulente DA, Wolters JF, et al. Codon Optimization Improves the Prediction of Xylose Metabolism from Gene Content material in Budding Yeasts. Mol Biol Evol. 2023;40:msad111. pmid:37154525
  111. 111.
    Kuang MC, Kominek J, Alexander WG, Cheng JF, Wrobel RL, Hittinger CT. Repeated cis-regulatory tuning of a metabolic bottleneck gene throughout evolution. Mol Biol Evol. 2018;35:1968–1981. pmid:29788479
  112. 112.
    Meurer M, Chevyreva V, Cerulus B, Knop M. The regulatable MAL32 promoter in Saccharomyces cerevisiae: traits and instruments to facilitate its use. Yeast. 2017;34:39–49. pmid:27714848
  113. 113.
    Stambuk BU, Araujo PS. Kinetics of lively alpha-glucoside transport in Saccharomyces cerevisiae. FEMS Yeast Res. 2001;1:73–78. pmid:12702465
  114. 114.
    Cousseau FEM, Alves SL, Trichez D, Stambuk BU. Characterization of maltotriose transporters from the Saccharomyces eubayanus subgenome of the hybrid Saccharomyces pastorianus lager brewing yeast pressure Weihenstephan 34/70. Lett Appl Microbiol. 2013;56:21–29. pmid:23061413
  115. 115.
    Vidgren V, Londesborough J. Characterization of the Saccharomyces bayanus-type AGT1 transporter of lager yeast. J Inst Brew. 2012;118:148–151.
  116. 116.
    Han E-Okay, Cotty F, Sottas C, Jiang H, Michels CA. Characterization of AGT1 encoding a basic α-glucoside transporter from Saccharomyces. Mol Microbiol. 1995;17:1093–1107. pmid:8594329
  117. 117.
    Horák J. Laws of sugar transporters: insights from yeast. Curr Genet. 2013;59:1–31. pmid:23455612
  118. 118.
    Vandermeulen MD, Cullen PJ. Ecological Inducers of the Yeast Filamentous Progress Pathway Reveal Surroundings-Dependent Roles for Pathway Elements. bioRxiv. 2023; 2023.05.25.542306. pmid:37732804
  119. 119.
    Duan S-F, Shi J-Y, Yin Q, Zhang R-P, Han P-J, Wang Q-M, et al. Reverse Evolution of a Basic Gene Community in Yeast Affords a Aggressive Benefit. Curr Biol. 2019;29:1126–1136.e5. pmid:30905601
  120. 120.
    New AM, Cerulus B, Govers SK, Perez-Samper G, Zhu B, Boogmans S, et al. Totally different Ranges of Catabolite Repression Optimize Progress in Steady and Variable Environments. PLoS Biol. 2014;12:1001764. pmid:24453942
  121. 121.
    Gallone B, Steensels J, Prahl T, Soriaga L, Saels V, Herrera-Malaver B, et al. Domestication and Divergence of Saccharomyces cerevisiae Beer Yeasts. Cell. 2016;166:1397–1410.e16. pmid:27610566
  122. 122.
    Gallone B, Mertens S, Gordon JL, Maere S, Verstrepen KJ, Steensels J. Origins, evolution, domestication and variety of Saccharomyces beer yeasts. Curr Opin Biotechnol. 2018;49:148–155. pmid:28869826
  123. 123.
    Hutzler M, Michel M, Kunz O, Kuusisto T, Magalhães F, Krogerus Okay, et al. Distinctive Brewing-Related Properties of a Pressure of Saccharomyces jurei Remoted From Ash (Fraxinus excelsior). Entrance Microbiol. 2021:12. pmid:33868204
  124. 124.
    Skoneczna A, Kaniak A, Skoneczny M. Genetic instability in budding and fission yeast—sources and mechanisms. FEMS Microbiol Rev. 2015;39:917–967. pmid:26109598
  125. 125.
    Klein HL. Spontaneous Chromosome Loss in Saccharomyces cerevisiae Is Suppressed by DNA Injury Checkpoint Capabilities. Genetics. 2001;159:1501–1509. pmid:11779792
  126. 126.
    Kumaran R, Yang SY, Leu JY. Characterization of Chromosome Stability in Diploid, Polyploid and Hybrid Yeast Cells. PLoS ONE. 2013;8:e68094. pmid:23874507
  127. 127.
    Heasley LR, Watson RA, Argueso JL. Punctuated Aneuploidization of the Budding Yeast Genome. Genetics. 2020;216:43–50. pmid:32753390
  128. 128.
    Krogerus Okay, Magalhães F, Kuivanen J, Gibson B. A deletion within the STA1 promoter determines maltotriose and starch utilization in STA1+ Saccharomyces cerevisiae strains. Appl Microbiol Biotechnol. 2019;103:7597–7615. pmid:31346683
  129. 129.
    Yamashita I, Nakamura M, Fukui S. Gene fusion is a potential mechanism underlying the evolution of STA1. J Bacteriol. 1987;169:2142–2149. pmid:3106330
  130. 130.
    Yamashita I, Maemura T, Hatano T, Fukui S. Polymorphic extracellular glucoamylase genes and their evolutionary origin within the yeast Saccharomyces diastaticus. J Bacteriol. 1985;161:574–582. pmid:3918018
  131. 131.
    Lo WS, Dranginis AM. FLO11, a yeast gene associated to the STA genes, encodes a novel cell floor flocculin. J Bacteriol. 1996;178:7144–7151. pmid:8955395
  132. 132.
    Lambrechts MG, Bauer FF, Marmur J, Pretorius IS. Muc1, a mucin-like protein that’s regulated by Mss10, is important for pseudohyphal differentiation in yeast. Proc Natl Acad Sci U S A. 1996;93:8419–8424. pmid:8710886
  133. 133.
    Rupp S, Summers E, Lo HJ, Madhani H, Fink G. MAP kinase and cAMP filamentation signaling pathways converge on the unusually massive promoter of the yeast FLO11 gene. EMBO J. 1999;18:1257–1269. pmid:10064592
  134. 134.
    Brückner S, Mösch HU. Choosing the proper way of life: adhesion and improvement in Saccharomyces cerevisiae. FEMS Microbiol Rev. 2012;36:25–58. pmid:21521246
  135. 135.
    Dranginis AM. Regulation of STA1 Gene Expression by MAT through the Life Cycle of Saccharomyces cerevisiae. Mol Cell Biol. 1989;9:3992–3998. pmid:2506439
  136. 136.
    Kim TS, Kim HY, Yoon JH, Kang HS. Recruitment of the Swi/Snf Advanced by Ste12-Tec1 Promotes Flo8-Mss11-Mediated Activation of STA1 Expression. Mol Cell Biol. 2004;24:9542–9556. pmid:15485921
  137. 137.
    Kim TS, Lee SB, Kang HS. Glucose Repression of STA1 Expression Is Mediated by the Nrg1 and Sfl1 Repressors and the Srb8-11 Advanced. Mol Cell Biol. 2004;24:7695–7706. pmid:15314176
  138. 138.
    Krogerus Okay, Gibson B. A re-evaluation of diastatic Saccharomyces cerevisiae strains and their function in brewing. Appl Microbiol Biotechnol. 2020;104:3745–3756. pmid:32170387
  139. 139.
    Gietz RD, Schiestl RH. Excessive-efficiency yeast transformation utilizing the LiAc/SS provider DNA/PEG methodology. Nat Protoc. 2007;2:31–34. pmid:17401334
  140. 140.
    Stoneman HR, Wrobel RL, Place M, Graham M, Krause DJ, de Chiara M, et al. CRISpy-Pop: A Internet Instrument for Designing CRISPR/Cas9-Pushed Genetic Modifications in Various Populations. G3 Genes|Genomes|Genetics. 2020;10:4287–4294. pmid:32963084
  141. 141.
    Bukhman YV, DiPiazza NW, Piotrowski J, Shao J, Halstead AGW, Bui MD, et al. Modeling Microbial Progress Curves with GCAT. BioEnergy Res. 2015;8:1022–1030.
  142. 142.
    R Core Staff. R: A Language and Surroundings for Statistical Computing. 2019;2: https://www.r-project.org. Out there from: http://www.r-project.org.
  143. 143.
    Hicks JB, Herskowitz I. INTERCONVERSION OF YEAST MATING TYPES I. DIRECT OBSERVATIONS OF THE ACTION OF THE HOMOTHALLISM (HO) GENE. Genetics. 1976;83:245–258. pmid:17248712
  144. 144.
    Bolger AM, Lohse M, Usadel B. Trimmomatic: a versatile trimmer for Illumina sequence knowledge. Bioinformatics. 2014;30:2114–2120. pmid:24695404
  145. 145.
    Koren S, Walenz BP, Berlin Okay, Miller JR, Bergman NH, Phillippy AM. Canu: scalable and correct long-read meeting through adaptive k-mer weighting and repeat separation. Genome Res. 2017;27:722–736. pmid:28298431
  146. 146.
    Walker BJ, Abeel T, Shea T, Priest M, Abouelliel A, Sakthikumar S, et al. Pilon: An Built-in Instrument for Complete Microbial Variant Detection and Genome Meeting Enchancment. PLoS ONE. 2014;9:e112963. pmid:25409509
  147. 147.
    Proux-Wéra E, Armisén D, Byrne KP, Wolfe KH. A pipeline for automated annotation of yeast genome sequences by a conserved-synteny strategy. BMC Bioinformatics. 2012;13:237. pmid:22984983
  148. 148.
    Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ. Fundamental native alignment search device. J Mol Biol. 1990;215:403–410. pmid:2231712
  149. 149.
    Gurevich A, Saveliev V, Vyahhi N, Tesler G. QUAST: high quality evaluation device for genome assemblies. Bioinformatics. 2013;29:1072–1075. pmid:23422339
  150. 150.
    Simão FA, Waterhouse RM, Ioannidis P, Kriventseva EV, Zdobnov EM. BUSCO: assessing genome meeting and annotation completeness with single-copy orthologs. Bioinformatics. 2015;31:3210–3212. pmid:26059717
  151. 151.
    Baker EC, Wang B, Bellora N, Peris D, Hulfachor AB, Koshalek JA, et al. The genome sequence of Saccharomyces eubayanus and the domestication of lager-brewing yeasts. Mol Biol Evol. 2015;32:2818–2831. pmid:26269586
  152. 152.
    Delcher AL. Quick algorithms for large-scale genome alignment and comparability. Nucleic Acids Res. 2002;30:2478–2483. pmid:12034836
  153. 153.
    Li H, Durbin R. Quick and correct brief learn alignment with Burrows–Wheeler remodel. Bioinformatics. 2009;25:1754–1760. pmid:19451168
  154. 154.
    Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics. 2009;25:2078–2079. pmid:19505943
  155. 155.
    Quinlan AR, Corridor IM. BEDTools: a versatile suite of utilities for evaluating genomic options. Bioinformatics. 2010;26:841–842. pmid:20110278
  156. 156.
    Garrison E, Marth G. Haplotype-based variant detection from short-read sequencing. arxiv.org. 2012.
  157. 157.
    Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES, Getz G, et al. Integrative genomics viewer. Nat Biotechnol. 2011;29:24–26. pmid:21221095
  158. 158.
    Monteiro PT, Oliveira J, Pais P, Antunes M, Palma M, Cavalheiro M, et al. YEASTRACT+: a portal for cross-species comparative genomics of transcription regulation in yeasts. Nucleic Acids Res. 2020;48:D642–D649. pmid:31586406
  159. 159.
    Kim D, Paggi JM, Park C, Bennett C, Salzberg SL. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat Biotechnol. 2019;37:907–915. pmid:31375807
  160. 160.
    Anders S, Pyl PT, Huber W. HTSeq—a Python framework to work with high-throughput sequencing knowledge. Bioinformatics. 2015;31:166–169. pmid:25260700
  161. 161.
    Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq knowledge with DESeq2. Genome Biol. 2014;15:550. pmid:25516281
  162. 162.
    Stephens M. False discovery charges: a brand new deal. Biostatistics. 2017;18:275–294. pmid:27756721
  163. 163.
    Zhao Y, Li M-C, Konaté MM, Chen L, Das B, Karlovich C, et al. TPM, FPKM, or Normalized Counts? A Comparative Examine of Quantification Measures for the Evaluation of RNA-seq Information from the NCI Affected person-Derived Fashions Repository. J Transl Med. 2021;19:269. pmid:34158060
  164. 164.
    Dillies M-A, Rau A, Aubert J, Hennequet-Antier C, Jeanmougin M, Servant N, et al. A complete analysis of normalization strategies for Illumina high-throughput RNA sequencing knowledge evaluation. Temporary Bioinform. 2013;14:671–683. pmid:22988256
  165. 165.
    Lin Y, Golovnina Okay, Chen Z-X, Lee HN, Negron YLS, Sultana H, et al. Comparability of normalization and differential expression analyses utilizing RNA-Seq knowledge from 726 particular person Drosophila melanogaster. BMC Genomics. 2016;17:28. pmid:26732976
  166. 166.
    Ellahi A, Thurtle DM, Rine J. The Chromatin and Transcriptional Panorama of Native Saccharomyces cerevisiae Telomeres and Subtelomeric Domains. Genetics. 2015;200:505–521. pmid:25823445
  167. 167.
    Eden E, Navon R, Steinfeld I, Lipson D, Yakhini Z. GOrilla: a device for discovery and visualization of enriched GO phrases in ranked gene lists. BMC Bioinformatics. 2009;10:48. pmid:19192299
  168. 168.
    Livak KJ, Schmittgen TD. Evaluation of Relative Gene Expression Information Utilizing Actual-Time Quantitative PCR and the two−ΔΔCT Technique. Strategies. 2001;25:402–408. pmid:11846609

[ad_2]

LEAVE A REPLY

Please enter your comment!
Please enter your name here