Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Copy number variations (CNVs) contribute to the adaptation process in two possible ways. First, they may have a direct role, in which a certain number of copies often provide a selective advantage. Second, CNVs can also indirectly contribute to adaptation because a higher copy number increases the so-called "mutational target size." In this study, we show that the copy number amplification of FLO11D in the osmotolerant yeast Zygosaccharomyces rouxii promotes its further adaptation to a flor-formative environment, such as osmostress static culture conditions. We demonstrate that a gene, which was identified as FLO11D, is responsible for flor formation and that its expression is induced by osmostress under glucose-free conditions, which confer unique characteristics to Z. rouxii, such as osmostress-dependent flor formation. This organism possesses zero to three copies of FLO11D, and it appears likely that the FLO11D copy number increased in a branch of the Z. rouxii tree. The cellular hydrophobicity correlates with the FLO11D copy number, and the strain with a higher copy number of FLO11D exhibits a fitness advantage compared to a reference strain under osmostress static culture conditions. Our data indicate that the FLO gene-related system in Z. rouxii has evolved remarkably to adapt to osmostress environments.

Free full text 


Logo of geneticsLink to Publisher's site
Genetics. 2013 Oct; 195(2): 393–405.
PMCID: PMC3781968
PMID: 23893487

Adaptation of the Osmotolerant Yeast Zygosaccharomyces rouxii to an Osmotic Environment Through Copy Number Amplification of FLO11D

Abstract

Copy number variations (CNVs) contribute to the adaptation process in two possible ways. First, they may have a direct role, in which a certain number of copies often provide a selective advantage. Second, CNVs can also indirectly contribute to adaptation because a higher copy number increases the so-called “mutational target size.” In this study, we show that the copy number amplification of FLO11D in the osmotolerant yeast Zygosaccharomyces rouxii promotes its further adaptation to a flor-formative environment, such as osmostress static culture conditions. We demonstrate that a gene, which was identified as FLO11D, is responsible for flor formation and that its expression is induced by osmostress under glucose-free conditions, which confer unique characteristics to Z. rouxii, such as osmostress-dependent flor formation. This organism possesses zero to three copies of FLO11D, and it appears likely that the FLO11D copy number increased in a branch of the Z. rouxii tree. The cellular hydrophobicity correlates with the FLO11D copy number, and the strain with a higher copy number of FLO11D exhibits a fitness advantage compared to a reference strain under osmostress static culture conditions. Our data indicate that the FLO gene-related system in Z. rouxii has evolved remarkably to adapt to osmostress environments.

Keywords: biofilm, copy number variations (CNVs), yeast, adaptation, flor

AN organism adapts to adverse environments to survive and produce progeny. Adaptive evolution is the process through which a population becomes better suited to its environment via mutation, selection, and random drift (Bürger 2000; Cressman 2003; Ewens 2004; Nowak and Sigmund 2004). Types of mutations include nucleotide changes (Steiner et al. 2007; Barrick and Lenski 2009), transposition events (Wilke and Adams 1992; Aminetzach et al. 2005), and copy number variations (CNVs), including gene amplifications (duplication) and deletions (Brown et al. 1998; Perry et al. 2007; Gresham et al. 2010). CNVs can have a phenotypic impact and can consequently alter the fitness of an allele through the following mechanisms: (i) changing the coding sequence of a gene (Yamanaka et al. 2009; Schlattl et al. 2011), (ii) creating paralogs that can diverge from each other and take on new or specialized functions (neofunctionalization or subfunctionalization, respectively) (Ohno 1970; Innan and Kondrashov 2010), and (iii) altering the expression level of a gene (gene dosage effect) (Stranger et al. 2007; Yamanaka et al. 2009; Iskow et al. 2012). In humans, most CNVs overlapping genes are under purifying (negative) selection (Conrad et al. 2010), but a handful of CNVs are thought to be under positive selection (Cooper et al. 2007; Hurles et al. 2010; Iskow et al. 2012) such as AMY1 (Perry et al. 2007). In yeast, there are several examples of how the (increased) copy number can provide a direct selective advantage (Gresham et al. 2008; Voordeckers et al. 2012).

In nature, budding yeast exhibits a number of adaptive responses, such as filamentation, invasive growth, flocculation, and biofilm formation, to overcome a deleterious environment (Fidalgo et al. 2006). The FLO (flocculation) gene family plays a central role in the mediation of these adaptive responses. For example, Saccharomyces cerevisiae has five FLO genes: FLO1, FLO5, FLO9, FLO10, and FLO11 (Teunissen and Ateensma 1995). FLO1, FLO5, FLO9, and FLO10 confer cell–cell adhesion (flocculation). Smukalla et al. (2008) showed that flocculation protects FLO1-expressing cells from multiple stresses, including antimicrobial and ethanol stress. In contrast, FLO11 is responsible for filamentation, invasive growth, and biofilm formation, such as a buoyant biofilm, which is also called flor (Guo et al. 2000; Zara et al. 2005; Fidalgo et al. 2006). The regulation of FLO11 is remarkably complex. At least three well-known signaling cascades regulate FLO11 expression in response to environment changes: the Ras-cAMP pathway, the MAP kinase-dependent filamentous growth pathway, and the glucose repression pathway (Madhani and Fink 1997; Rupp et al. 1999; Gagiano et al. 2002; Kuchin et al. 2002; Vyas et al. 2003; Schwartz and Madhani 2004; Verstrepen and Klis 2006). Accordingly, FLO11-dependent flor formation is considered as an adaptation to nutrient-limited and anoxic conditions because flor formation enables yeast to continue its growth through the utilization of oxygen and ethanol, which are produced by the yeast itself. In addition to these pathways, FLO11 is also regulated by the pH response pathway, noncoding RNAs, and the chromatin state (Halme et al. 2004; Barrales et al. 2008, 2012; Bumgarner et al. 2009; Octavio et al. 2009). A recent study showed that the noncoding RNAs contribute to a variegated transcription of FLO11 through the modulation of transcription factor recruitment (Bumgarner et al. 2012).

The FLO genes share a common structure, including a C-terminal glycosylphosphatidylinositol (GPI)-anchor addition site, an N-terminal carbohydrate- or peptide-binding domain or domain of unknown function, and a central domain containing multiple serine and threonine-rich repeats (Verstrepen and Klis 2006). The latter domain is also referred to as a domain with intragenic tandem repeats. The variation in the intragenic repeat number and/or the distribution of the different repetitive units provides a functional diversity of the cell-surface antigens that allows rapid adaptation to the environment (Verstrepen et al. 2005; Fidalgo et al. 2006, 2008; Zara et al. 2009). However, the relationship between adaptation and the copy number of the FLO gene itself, but not the number of the intragenic repeat units, has not been investigated.

It is known that not only S. cerevisiae, but also the osmotolerant yeast Zygosaccharomyces rouxii, form flor (Tomita and Yamamoto 1997). Z. rouxii has played a central role in the production of traditional fermented foods (e.g., soy sauce and soybean paste) in Japan; however, it is also known to cause yeast spoilage in the food industry. In soy sauce manufacturing, for example, Z. rouxii produces alcohol and aromatic flavors, such as 4-hydroxy-2(or 5)-ethyl-5(or 2)-methyl-3(2H)-furanone. In addition, the flor-forming strain produces off-flavors, such as isovaleric acid, in soy sauce. Interestingly, it is known that flor is formed in a NaCl-dependent manner in Z. rouxii (Tomita and Yamamoto 1997). We predicted that the NaCl-dependent ability to form flor by osmotolerant yeast is a reflection of a physiological role of flor formation under high-osmolarity conditions.

In this study, we demonstrate the molecular mechanism that underlies the ability of NaCl-dependent flor formation in Z. rouxii. Through a series of experiments, we found that FLO11D, which is responsible for flor formation in Z. rouxii, is a good example of a gene that exhibits copy number amplification that was positively selected. We also show that the copy number amplification of FLO11D enables Z. rouxii to be further adapted to a flor-formative environment, such as osmostress static culture conditions, through direct competition experiments.

Materials and Methods

Strains, media, and genetic methods

The yeast strains used in this study are listed in Table 1. Z. rouxii Z3 and Z1 were isolated from the flor of yeasts grown on the surface of soy sauce mash at a soy sauce factory (Yamasa Corporation, Chiba, Choshi, Japan) and the laboratory, respectively, in 2009. Z. rouxii 3K was isolated from soy sauce mash as a non-flor forming strain at the soy sauce factory. Z. rouxii 1501 was isolated from soy sauce as a flor-forming strain in 1972. S. cerevisiae 133d flo11Δ was generously provided by J. I. Ibeas (Fidalgo et al. 2006), and BY4743 was purchased from Funakoshi Co. (Tokyo). The FLO11D gene disruption was performed using PCR-mediated gene replacement, and the gene deletion was confirmed using PCR and Southern blot analyses. pCUG, pCUZ, and pCUN were used as PCR templates for gene disruption, and each of these possesses G418-, Zeocin-, and Nourseothricin-resistant genes surrounded by two loxP sequences. The primers that were used in this study are listed in Supporting Information, Table S1. The transformations of Z. rouxii and S. cerevisiae were performed by electroporation and lithium acetate procedure, respectively (Gietz et al. 1995; Watanabe et al. 2010). The bacterial transformations, bacterial DNA manipulation, and plasmid constructions were performed using standard methods (Sambrook and Russell 2001).

Table 1

Strains used in this study
StrainsGenotypeSource
Z. rouxii 3KSoy sauce
1501Soy sauce
ATCC46251Unknown
ATCC2623Black-grape must
Z1Soy sauce
Z3Soy sauce/Watanabe et al. (2010)
NBRC0505Shoyu yeast, Kikkoman
NBRC0506Shoyu yeast, Yamasa
NBRC0845Mash of tamari-soya
NBRC1876Soy bean paste
NBRC1877Soy bean paste
NBRC1877ura3Δura3Δ::loxP/ura3Δ::loxPThis study
Z3 flo11aΔflo11aΔ::loxP-KanMX-loxPThis study
Z3 flo11bΔflo11bΔ::loxP-KanMX-loxPThis study
Z3 flo11cΔflo11cΔ::loxP-ZeoMX-loxPThis study
Z3 flo11abΔflo11abΔ::loxP-KanMX-loxPThis study
Z3 flo11abcΔflo11abΔ::loxP-KanMX-loxP flo11cΔ::loxP-ZeoMX-loxPThis study
Z3 ura3Δura3Δ::loxP/ura3Δ::loxPThis study
Z3 flo11dΔ1ura3Δ::loxP/ura3Δ::loxP flo11dΔ::loxP-KanMX-loxPThis study
Z3 flo11dΔ2ura3Δ::loxP/ura3Δ::loxP flo11dΔ::loxP-KanMX-loxPThis study
flo11dΔ::loxP-ZeoMX-loxP
Z3 flo11dΔ3ura3Δ::loxP/ura3Δ::loxP flo11dΔ::loxP-KanMX-loxPThis study
flo11dΔ::loxP-ZeoMX-loxP flo11dΔ::loxP-NatMX-loxP
ATCC46251 flo11dΔ1flo11dΔ::loxP-KanMX-loxPThis study
ATCC46251 flo11dΔ2flo11dΔ::loxP-KanMX-loxP flo11dΔ::loxP-ZeoMX-loxPThis study
ATCC46251 flo11dΔ3flo11dΔ::loxP-KanMX-loxP flo11dΔ::loxP-ZeoMX-loxPThis study
flo11dΔ::loxP-NatMX-loxP
NBRC0845 flo11dΔ1flo11dΔ::loxP-KanMX-loxPThis study
NBRC0845 flo11dΔ2flo11dΔ::loxP-KanMX-loxP flo11dΔ::loxP-ZeoMX-loxPThis study
1501 flo11dΔ1flo11dΔ::loxP-KanMX-loxPThis study
S. cerevisiae BY4743MATahis3Δ1/his3Δ1 leu2Δ0/leu2Δ0 LYS2/lys2Δ0 met15Δ0 /MET15Funakoshi Co.
ura3Δ0/ ura3Δ0
133d flo11ΔMATa ura3-52 flo11ΔJ. I. Ibeas/Fidalgo et al. (2006)

Standard YPD medium (1% yeast extract, 2% Bacto peptone, and 2% glucose) containing 200 mg/liter G418, 200 mg/liter Zeocin, and/or 5 mg/liter Nourseothricin and synthetic complete dextrose medium lacking uracil (SCD ura) were used for the transformant selection. YPD containing 1.8 M NaCl was used as the flor-forming media for Z. rouxii. Synthetic complete galactose medium lacking uracil (SCG ura-) was used as the flor assay media for S. cerevisiae. YPD plates supplemented with 0.35% agar and YPD plates supplemented with 0.40% agar and 1.8 M NaCl were used to investigate mat formation. All of the yeasts were incubated at 28°. For the incubation of ura3Δ mutants in Z. rouxii, 0.17% uracil was used to supplement the medium due to their significant requirement for uracil.

Assays for flor formation, mat formation, and determination of cellular hydrophobicity

The flor formation in Z. rouxii was assayed by inoculating precultured cells in glass tubes containing 10 ml of YPD or YPD supplemented with 1.8 M NaCl. The mat formation was assayed by inoculating cells from a fresh colony onto YPD soft agar plates containing 1.8 M NaCl. The flor formation in S. cerevisiae was assayed by inoculating cells from a fresh colony into glass tubes containing 10 ml of SCG ura- medium. The cells were grown at 28° for 3–7 days under static conditions and photographed. The exact incubation periods are written in the legend for Figures 16. The assays for the determination of the cellular hydrophobicity were performed as previously described (Barrales et al. 2008) with little modification. Briefly, the cells were grown in SCD containing 1.8 M NaCl, and then a portion of the cells was transferred to fresh medium. The cells were grown to an OD600 of ~1.0, and then 1.2 ml of the culture was overlaid with 600 μl of octane and vortexed for 3 min. The OD600 of the aqueous layer was recorded, and the relative difference compared with the initial OD600 was used to determine the percentage of cellular hydrophobicity. The detailed colony morphology was observed using a digital microscope VHX-1000 equipped with VH-Z250R/Z250W (Keyence, Osaka, Japan).

An external file that holds a picture, illustration, etc.
Object name is 393fig1.jpg

Spontaneous flor on the surface of soy sauce mash and in manufactured soy sauce. (A) Spontaneous flor on the soy sauce mash in a concrete tank. The white pixels on the mash are the flor. (B) Spontaneous flor on manufactured soy sauce in glass bottles. (C) Appearance of the flor formation process. Z. rouxii Z3 was inoculated into soy sauce mash or soy sauce and incubated at 28° for 3 days. Bar, 30 μm.

An external file that holds a picture, illustration, etc.
Object name is 393fig6.jpg

Copy number variations of FLO11D in Z. rouxii. (A) PCR amplification of the FLO11D locus. (B) Flor formation in wild-type, single, double, and triple FLO11D gene mutants. A fresh colony was inoculated in 10 ml of YPD containing 1.8 M NaCl and incubated at 28° for 5 days. (C) Mat formation in wild-type, single, double, and triple FLO11D gene mutants. A fresh colony was inoculated onto YPD plates supplemented with 0.4% agar and 1.8 M NaCl and incubated at 28° for 7 days. (D) Karyotype profiles of Z. rouxii strains. The untreated chromosome (top) and NotI-digested chromosome (bottom) pattern was analyzed by Southern blot using the Flo11 domain-coding region of the FLO11D gene as a probe. (E) Summary of the loss and gain of FLO11D allele events. The tree shows the cluster based on the NotI-digested chromosome length polymorphism in D. Each event was manually mapped on the tree.

ura3 mutant construction in Z. rouxii NBRC1877 and Z3

We attempted to identify the URA3 gene coded on the P subgenome in Z. rouxii because of the requirement for the auxotrophic mutant in the allopolyploid of Z. rouxii. The genomes from ATCC2623 and ATCC46251 were digested with BglII, SpeI, and SacI, and the fragments were analyzed by Southern blot using the coding region of URA3 from ATCC2623 as a probe. The specific signal from URA3 coded on the P subgenome was detected at ~2.2 kb in the BglII-digested ATCC46251 genome. The band was recovered and cloned into the same site of pZEAK, which was constructed from pZEU (Pribylova et al. 2007b) by removing the URA3 marker genes and disrupting the BamHI recognition site derived from pSRI. S. cerevisiae BY4743 (a uracil auxotrophic mutant) was transformed with the resulting plasmid and selected on an SCD ura- plate. The sequence analysis of the plasmid extracted from the transformants revealed the URA3 gene coded on the P subgenome (AB665419).

Z. rouxii NBRC1877 and the Z3 ura3 mutant were obtained through a two-step deletion of the ura3 gene (XM002499041, AB665419). The primers that were used in the gene disruption are indicated in Table S1. The removal of the G418 and Zeocin resistance markers from the genomes were performed as previously described (Pribylova et al. 2007b) with little modification. The cells were transformed with pZCRE, and the transformants were grown selectively on SCD ura- plates for 2 days and then nonselectively in liquid YPD supplemented with 0.17% uracil (YPDU) for 24 hr. The cultures were diluted and plated on YPDU to obtain single colonies. Two days later, the colonies were replated on YPDU, YPDU + G418, YPDU + G418 + Zeocin, and SCD ura- plates, and those clones with both G418- and Zeo-sensitive and Ura- phenotypes were selected. The removal of the G418 and Zeocin resistance markers from the genome was confirmed using PCR analysis.

Plasmid construction

To identify the flor-forming gene through functional cloning, Z3 genomic libraries were prepared. The Z3 genomic DNA was partially digested with Sau3AI, and the resulting fragments were separated by agarose gel electrophoresis. The 6- to 9-kb DNA fragments were recovered using a PCR and Gel Purification Kit (Promega, Fitchburg, WI) and cloned into the BamHI site of pZEUneo, which was constructed from pZEU (Pribylova et al. 2007b) by disrupting the BamHI recognition site derived from pSRI. Escherichia coli DH5α cells were transformed with the resulting plasmids, and the transformants were selected by Luria broth medium containing carbenicillin. The resulting Z3 genomic library was extracted from these bacteria and used for functional cloning. The FLO11D gene was amplified by PCR from the Z. rouxii Z3 genomic DNA using the primers pFLO11D_F and pFLO11D_R to append the EcoRI and SalI digestion sites onto either end of the PCR product. The PCR product was digested with the respective restriction enzymes and ligated into EcoRI- and SalI-digested pZEU (Pribylova et al. 2007a) to generate pFLO11D. To drive FLO11D under the GAL1 promoter, pYFLO11D were constructed. Briefly, the coding sequence of FLO11D was amplified by PCR from the Z. rouxii Z3 genomic DNA using the primers pYFLO11D_F and pYFLO11D_R. The resulting DNA fragments and pYES2 (Life Technologies, Carlsbad, CA) digested with BamHI and HindIII were introduced into S. cerevisiae BY4743 and then plated onto a SCD ura- plate. After the resulting plasmids were introduced into E. coli and were amplified, BY4743 and 133d flo11Δ were transformed by the plasmid. The PCR analyses were initiated using KOD plus neo (Toyobo, Osaka, Japan) according to the manufacturer’s instructions.

DNA sequencing, alignment, and analysis

All of the DNA sequences were obtained through outsourcing (Fasmac, Atugi, Japan). All of the BLASTP and PSI-BLAST searches (Altschul et al. 1997) applied an expected value cutoff of 10−5 at the hemiascomycete yeast genome database (Sherman et al. 2009; http://www.genolevures.org/) and through the National Center for Biotechnology Information. The protein sequences were aligned using ClustalX (Larkin et al. 2007). The initial formatting of the alignments was conducted in GeneDoc (http://www.psc.edu/biomed/genedoc). For the phylogenetic inferences, we used the neighbor-joining method (Saitou and Nei 1987) with 1000 bootstrap interactions. The tree was visualized in NJplot (Perrière and Gouy 1996). The predicted N-terminal signal peptides were detected with the SignalP 3.0 algorithm (Emanuelsson et al. 2007). The C-terminal GPI anchor attachment signals were detected using the fungal big-Π prediction algorithm (Eisenhaber et al. 2004).

Southern and Northern blot analyses

For the Southern blot analysis, the genomic DNA of yeast cells was extracted using standard protocols (Sambrook and Russell 2001) and digested with the appropriate restriction enzyme. The digested DNA was separated by 1% agarose gel electrophoresis and transferred onto a Hybond-N+ membrane (GE Healthcare, Buckinghamshire, UK) by upward capillary transfer.

For the Northern blot analysis, the total RNA was prepared with Isogen (NIPPON gene, Tokyo) according to the manufacturer’s instructions with little modification. Briefly, the total RNA was extracted after the cells were homogenized in a Micro Smash MS-100 bead beater (TOMY, Tokyo) at 3000 × g for 300 sec with adequate amounts of glass beads (0.1 mm in diameter) and Isogen solution. A 5-μg aliquot of the total RNA was run on a gel and blotted.

The Flo11 domain-coding region, which is associated with the Flo11p N-terminal region in the Pfam database (Finn et al. 2010), was used to probe the membranes. These experiments were performed using a PCR digoxigenin probe synthesis kit (Roche, Basel, Switzerland) and detection starter kit II (Roche) according to the manufacturer’s instructions.

Chromosomal DNA isolation and pulsed-field gel electrophoresis

The chromosomal DNA in plug was prepared according to a previous study with little modifications (Pribylova et al. 2007a). Instead of Lyticase, Zymolyase 100T (SEIKAGAKU Corporation, Tokyo) was used, and the cell treatment was performed overnight at 30°. The NotI-digested plug was prepared according to a previous study (Lim et al. 1994). The 1% agarose gels were prepared from pulsed-field certified agarose (Bio-Rad, Hercules, CA) and run on a CHEF apparatus (Bio-Rad) in 0.5× TBE buffer at 10°. The running conditions for chromosomal DNA separation were as follows: switch time, 350–400 sec; run time, 240 hr; angle, 120°; and voltage, 3 V/cm. The running conditions for NotI-digested chromosomal DNA separation were as follows: switch time, 60–120 sec; run time, 24 hr; angle, 120°; and voltage, 6 V/cm.

The DNA banding patterns were analyzed with Quantity One Basic (Bio-Rad) and manually. The cluster analysis of the band patterns was performed using SPSS version 11.5 with a word algorithm.

Fitness measurements

Yeast cells (wild-type, single, double, and triple FLO11D gene mutants) were independently grown in YPDU, and then a portion of the cells was transferred to fresh medium. The cells were grown to an OD600 of 1.0, and then 100 μl from each culture was mixed with its reference strain: wild type was mixed with single mutant, single mutant was mixed with double mutant, and double mutant was mixed with triple mutant. The mixture was inoculated into YPDU containing 1.8 M NaCl and incubated at 28° with or without shaking. After 7 days of incubation, the culture was vortexed for 3 min, and 100 μl of the culture was harvested. The ratio of the two competitors was quantified at the initial and final time points as follows: The culture was diluted and plated on YPDU medium to obtain a single colony. Three days later, the colonies were counted to calculate the colony formation units, and at least 208 colonies were randomly selected and transferred to YPDU or YPDU containing G418, Zeocin, or Nourseothricin to calculate the composition ratio of wild-type, single, double, and triple FLO11D gene mutants. The selective advantage, s, of each strain was calculated as s = [In(Sf/Rf)-ln(Si/Ri)]/T, where S and R are the numbers of sample and reference strains, respectively; the subscripts refer to the final and the initial populations, and T is the number of generations during the competition. The fitness of the reference strain was designated 1, and the fitness of the sample strain was 1 + s.

Results

Identification of the gene conferring floatability to Z. rouxii

In soy sauce manufacturing, flor is naturally formed on the surface of soy sauce mash and soy sauce products by the flor-forming Z. rouxii (Figure 1, A and B). This strain reduces the quality of soy sauce products by producing off-flavors. We first observed the flor formation process to investigate how a flor would be developed on the surface of soy sauce mash and soy sauce. After the cells elongated and formed chains on the surface of the substrate, the cells began to elongate along the z-axis (Figure 1C, arrows), and this elongation was followed by the acquisition of a fluffy morphology (Figure 1C). It is known that the Z. rouxii F51 strain forms flor in NaCl-containing medium, but Z. rouxii never forms flor under NaCl-free conditions (Tomita and Yamamoto 1997). We replicated the experiment using a different genetic background in liquid static culture and on soft agar plates or normal agar plates to compare the morphologic changes. Flor yeasts display morphological development, such as flor, fluffy colony morphology, and mat, in NaCl-containing medium (Figure 2). These results are in agreement with the previous report, with the exception of the weak flor formation observed in the absence of NaCl. In contrast, non-flor-forming yeasts never developed flor, regardless of whether the medium contained NaCl (Figure 2A). These results suggest that the morphological development in Z. rouxii is strongly induced by NaCl.

An external file that holds a picture, illustration, etc.
Object name is 393fig2.jpg

NaCl-dependent morphological changes. (A) Flor formation in YPD or YPD containing 1.8 M NaCl. A fresh colony was inoculated into YPD or YPD containing 1.8 M NaCl and incubated at 28° for 5 days under static culture conditions. (B) Mat formation on YPD soft agar plates or YPD soft agar plates containing 1.8 M NaCl. A fresh colony was inoculated onto YPD plates supplemented with 0.35% agar or 0.4% agar containing 1.8 M NaCl and incubated at 28° for 7 days. (C) Assay for colony fluffiness. Z. rouxii Z3 was plated on YPD or YPD containing 1.8 M NaCl and incubated at 28° for 3 or 4 days, respectively. Bar, 0.3 mm.

It is known that the FLO11 gene is responsible for flor formation in S. cerevisiae. A recent study showed that the N-terminal domain of Flo11p, which is referred to as the Flo11 domain, self-interacts and explains the cell–cell interaction capacity of FLO11-expressing cells (Goossens and Willaert 2012). The Flo11 domain is conserved in the S. cerevisiae Flo11 protein and in the respective proteins from some related species within Saccharomycotina but was not found in any other fungal lineage (Linder and Gustafsson 2008). We searched for the FLO11 homolog in the Z. rouxii genome database (Sherman et al. 2009) to identify the gene conferring floatability to Z. rouxii. BLASTP searches were used with an expected value cutoff of 10−5 using the S. cerevisiae Flo11 domain as the query. As a result, three candidates were found, including ZYRO0F02728g, ZYRO0F02750g, and ZYRO0A13662g, which were designated FLO11A, FLO11B, and FLO11C, respectively (Figure S1A). These candidates have the Flo11 domain in the N terminus, Ser/Thr-rich repeats in the middle domain, and the GPI-attachment signal in the C terminus, indicating that not only the N-terminal region but also the overall structure of these candidates is similar to that of S. cerevisiae Flo11p. To investigate the role of FLO11A, FLO11B, and FLO11C, we constructed single, double, and triple gene mutants; however, all of the mutants formed flor and mat normally (Figure S1, C and D). The Southern blot analysis using the Flo11 domain coding region as a probe indicated that these strains had FLO11A, FLO11B, and FLO11C, as well as additional and multiple FLO11-like genes (Figure S1B). The signals from the additional FLO11-like genes would be derived from the P subgenome because it is presumed that the Z. rouxii in soy sauce mash is primarily of an allopolyploid and/or diploid mosaic linage (Tanaka et al. 2012; Watanabe et al. 2013) with one parent similar to Z. rouxii CBS732 and one NCYC3042 parent, which is informally called Z. pseudorouxii (James et al. 2005; Solieri et al. 2006, 2007, 2013; Gordon and Wolfe 2008).

We then attempted conventional functional cloning strategies by transforming the non-flor yeast NBRC1877 ura3Δ with Z3 genomic libraries that contain 20,000 independent clones (average insert size: 6 kbp) to select the floating clones. Approximately 200 colonies that formed on the SCD ura- plate were inoculated into 10 ml of SCD ura- containing 1.8 M NaCl, and the yeasts growing on the surface of the medium were recovered and purified. Consequently, we recovered a total of four colonies that form weak flor from the ~ 20,000 colonies that were screened (Figure 3A). The sequence analysis of the plasmid extracted from the abovementioned four colonies identified FLO-like genes (Figure 3B). All of these genes were homologous to each other, except for a difference in the number of repeats in the middle domain and a few base substitutions in the promoter region (Figure S2). The BLASTP analysis of the N-terminal region of this gene revealed a Flo11 domain. To compare the sequence of the Flo11 domain, we searched and aligned homologous sequences. Similar to the result reported by Linder and Gustafsson (2008), the homology search did not detect a Flo11 domain in any other fungal lineages except Saccharomycotina. The phylogenetic analysis revealed that the Flo11 domain obtained above is similar to that of Flo11ap and Flo11bp, but some differences existed (Figure S3). These results suggest that the gene is novel. Thus, we designated it FLO11D. The FLO11D-specific signals detected by Southern blot and PCR analyses were obtained only from the flor-forming strain (Figure 3C), indicating that FLO11D may be responsible for flor formation. If FLO11D truly played a role in flor formation in Z. rouxii, its expression would be detected during the flor-forming phase. We prepared total RNA sequentially from sedimenting cells and floating cells to investigate the FLO11D gene expression (Figure 3D). FLO11D transcripts were detected in floating cells after 2 and 3 days of culture by Northern blot analysis (Figure 3E). In contrast, FLO11A, FLO11B, and FLO11C gene expression was not observed by Northern blot analysis, but the expression of these genes was slightly detected by RT-PCR analysis regardless of the culture phase (data not shown). These results suggest that only FLO11D is upregulated at the flor-forming phase. Finally, to demonstrate the role of the FLO11D gene in the morphological change, we constructed a FLO11D deletion mutant in Z3. As shown in Figure 4, Z3 possesses three copies of FLO11D, and the complete deletion of FLO11D in Z3 abolished flor, mat, and fluffy colony morphology development. The plasmids containing FLO11D restored the ability to form flor, mat, and fluffy colony morphology, indicating that this gene is required for the morphological development (Figure 4, B–E). To verify that FLO11D is directly responsible for flor formation, FLO11D was heterologously expressed under the GAL1 promoter in S. cerevisiae BY4743 and 133d flo11Δ. As a result, S. cerevisiae harboring pYFLO11D formed weak flor under galactose- but not glucose-containing conditions (Figure 4F). These results suggest that FLO11D is directly responsible for flor formation.

An external file that holds a picture, illustration, etc.
Object name is 393fig3.jpg

Identification of FLO11D gene. (A) Flor formation in the transformants obtained from functional cloning. A fresh colony was inoculated to 10 ml of SCD ura- containing 1.8 M NaCl and incubated at 28° for 4 days. (B) A DNA restriction map of the FLO11D gene using ScaI endonuclease (top) and the primary structure of Flo11dp (bottom). (C) Detection of FLO11D by Southern blot and PCR analyses. ScaI-digested genomic DNA was analyzed by Southern blot analysis using the Flo11 domain-coding region of the FLO11D gene as a probe (top). FLO11D was amplified by PCR using FLO11D-specific primers (bottom). The URA3 gene was used as a positive control in both analyses. (D) Sequential flor formation assay in YPD containing 1.8 M NaCl. Z3 was inoculated into 20 ml of YPD containing 1.8 M NaCl and incubated at 28° for 3 days under static culture conditions. (E) Detection of FLO11D transcripts by Northern blot analysis. The total RNA was extracted from the yeast cells, and the FLO11D expression was measured by Northern blot analysis using the Flo11 domain-coding region of the FLO11D gene as a probe.

An external file that holds a picture, illustration, etc.
Object name is 393fig4.jpg

FLO11D is required for NaCl-dependent morphological changes. (A) PCR amplification of the FLO11D locus. (B) Flor formation in wild-type, single, double, and triple FLO11D gene mutants with an empty vector or a FLO11D-containing vector (pFLO11D). A fresh colony was inoculated in 10 ml of YPD containing 1.8 M NaCl and incubated at 28° for 5 days. (C) Mat formation in wild-type, single, double, and triple FLO11D gene mutants with an empty vector or pFLO11D. A fresh colony was inoculated onto YPD plates supplemented with 0.4% agar and 1.8 M NaCl and incubated at 28° for 7 days. (D) Microscopic analysis of C. Bar, 0.5 mm. (E) Assay for colony fluffiness. Z. rouxii Z3 or flo11dΔ3 strains were plated on YPDU containing 1.8 M NaCl and incubated at 28° for 4 days. Bar, 0.3 mm. (F) Flor formation assay in S. cerevisiae BY4743 and 133d flo11Δ with an empty vector or a FLO11D-containing vector (pYFLO11D). A fresh colony was inoculated in 10 ml of SCD ura- and SCG ura- and incubated at 28° for 4 days.

Osmotic stress-induced FLO11D expression under glucose-free conditions

Z. rouxii forms flor in a NaCl-dependent manner, and, to the best of our knowledge, this ability is a Z. rouxii-specific characteristic. We hypothesized that this unique feature was due to NaCl-dependent FLO11D expression. To test this hypothesis, Z3 was incubated with shaking in synthetic complete dextrose medium (SCD) and was then transferred to various conditions (SCD, SCD containing NaCl, SC, or SC containing NaCl). We prepared total RNA from these cultures after 30, 60, and 90 min and then measured FLO11D expression by Northern blot analysis. FLO11D-specific signals were detected only in SC containing 1.8 M NaCl (Figure 5A), suggesting that FLO11D is repressed by glucose and induced by NaCl under glucose-free conditions. To examine other carbon sources in the repression of FLO11D, FLO11D expression was measured by Northern blot analysis in SC containing 1.8 M NaCl and other carbon sources, such as galactose, ethanol, and glycerol. As a result, these carbon sources did not repress the FLO11D expression under our experimental conditions (data not shown).

An external file that holds a picture, illustration, etc.
Object name is 393fig5.jpg

FLO11D expression and flor formation under osmostress conditions. (A) The regulation of FLO11D expression. The cells were grown to an OD600 of 1.0 in SCD, washed, and transferred to SCD, SCD containing 1.8 M NaCl, SC, or SC containing 1.8 M NaCl. After 30, 60, and 90 min, the total RNA was extracted and analyzed by Northern blot analysis using the Flo11 domain-coding region of the FLO11D gene as a probe. (B) Flor formation of Z3 in YPD medium or YPD medium containing 1.8 M of various chemicals (NaCl, KCl, NaNO3, and sorbitol). A fresh colony was inoculated into the above-mentioned medium and incubated at 28° for 7 days. (C) FLO11D gene expression in response to various osmostress conditions. The cells were grown to an OD600 of 1.0 in SCD, washed, and transferred to SC, SC containing 1.8 M NaCl, SC containing 1.8 M KCl, SC containing 1.8 M NaNO3, or SC containing 1.8 M sorbitol. After 60 min, the total RNA was extracted and analyzed by Northern blot analysis using the Flo11 domain-coding region of the FLO11D gene as a probe.

To demonstrate whether FLO11D expression is specifically induced by NaCl, we observed flor formation in YPD medium containing various chemicals (NaCl, MgCl2, KCl, MgSO4, Na2SO4, NaNO3, and sorbitol). Z. rouxii Z3 formed flor in medium containing 1.8 M NaCl, KCl, NaNO3, and sorbitol after a 7-day incubation (Figure 5B) and in the medium containing 0.9 M MgCl2, 1.8 M MgSO4, and Na2SO4 after a 16-day incubation (Figure S4). The result of the Northern blot analysis clearly demonstrated that FLO11D was induced not only by NaCl but also by KCl, NaNO3, and sorbitol under glucose-free conditions (Figure 5C). These results indicate that FLO11D is most likely induced by osmostress under glucose-free conditions. Consequently, flor formation can be observed in the late phase of culture, when the depletion of glucose occurs and Z. rouxii cells are subjected to osmotic stress.

FLO11D copy number variations

To confirm the requirement of FLO11D for flor formation in other genetic background strains, we constructed FLO11D gene mutants in the ATCC46251, NBRC0845, and 1501 strains. This analysis also showed that FLO11D played a role in flor formation, and there were FLO11D copy number variations (i.e., Z3, ATCC46251, NBRC0845, and 1501 possessed three, three, two, and one copies of FLO11D, respectively) (Figure 6, A–C).

Using a combination of pulsed-field gel electrophoresis (PFGE) and Southern blot analyses, we investigated how many FLO11D allele amplification events occurred in Z. rouxii. With respect to the NBRC1877, 1501, NBRC0845, ATCC46251, Z3, and Z1 strains, zero, one, one, three, two, and three copies of FLO11D, respectively, were detected by chromosomal PFGE analysis, whereas zero, one, two, three, three, and three copies, respectively, were detected by NotI-digested chromosomal PFGE analysis (Figure 6D). The latter analysis is in agreement with the FLO11D gene disruption experiment, indicating that the NBRC1877, 1501, NBRC0845, ATCC2623, Z3, and Z1 strains possess zero, one, two, three, three, and three copies of the FLO11D allele, respectively. These results also indicate that the first and second FLO11D alleles are located on either the same or different chromosomes, which have the same mobility in NBRC0845 and Z3. Interestingly, the mobility of the third FLO11D allele of Z3 was different from that of ATCC46251 and Z1, indicating that either the acquisition of the third FLO11D allele occurred independently two times or a sequence alteration in the third FLO11D allele occurred in Z3. We next compared the NotI-digested karyotype profiles of various Z. rouxii strains by cluster analysis, and the FLO11D amplification events were mapped on the tree (Figure 6E). The loss or gain of FLO11D allele events can be explained by the dendrogram, which suggested that the FLO11D allele originated from a single origin and that sequence alteration in the third FLO11D allele occurred in Z3 after its acquisition.

Adaptation to osmostress environments by copy number amplification of FLO11D

In S. cerevisiae, it has been reported that the length and type of repetitive units in the Flo11p coding repeats domain affect the Flo11p-associated functions, including cellular hydrophobicity (Fidalgo et al. 2006; Fidalgo et al. 2008; Zara et al. 2009); however, the relationship between its functions and the copy number of the FLO gene itself has not been elucidated. To investigate this relationship, we measured the cellular hydrophobicity of FLO11D mutants in Z3. As a result, the cellular hydrophobicity was correlated with the FLO11D copy number (Figure 7A).

An external file that holds a picture, illustration, etc.
Object name is 393fig7.jpg

Biological roles of FLO11D copy number variation. (A) Cellular hydrophobicity. The cell cultures were overlaid with octane and mixed. The OD600 of the aqueous layer was recorded, and the relative difference compared with the initial OD600 was used to determine the percentage of cellular hydrophobicity. The error bars represent the standard deviation between three independent experiments. The results were analyzed by one-way ANOVA with Bonferroni post test. (B) Effects of changes in the FLO11D copy number on the yeast fitness under osmostress conditions. An equal number of reference and sample strains (combinations of wild-type and single FLO11D gene mutants, single and double FLO11D gene mutants, and double and triple FLO11D gene mutants) was inoculated into 10 ml of YPD containing 1.8 M NaCl and incubated at 28° with or without shaking. The ratio of the two competitors was quantified at the initial and final time points (7-day incubation) based on its drug resistance. The fitness of the reference strain was designated as 1 and the fitness of the sample strain as 1 + s. The selective advantage, s, of each strain was calculated by the formula described in Materials and Methods. The error bars represent the standard deviation between three independent experiments. The results were analyzed by t-test.

We hypothesized that the copy number amplification of FLO11D promoted adaptation to osmostress environments because of their osmo-ergic properties and the correlation between the FLO11D copy number and cellular hydrophobicity. To test this hypothesis, we incubated an equivalent mixture of wild-type and single FLO11D mutant, single and double FLO11D mutants, and double and triple FLO11D mutants and sequentially traced the population that remained in the culture. In a comparison between static culture and shaking culture conditions in YPD medium containing 1.8 M NaCl, the strain with more copy numbers of FLO11D showed a considerable fitness advantage compared to the reference strain under static culture conditions: the wild-type strain was dominant over the single mutant, the single FLO11D mutant was dominant over the double FLO11D mutant, and the double FLO11D mutant was dominant over the triple FLO11D mutant (Figure 7B). These results suggest that the FLO11D copy number amplification promotes further adaptation to a flor-formative environment, such as osmostress static culture conditions.

Discussion

Previous phenotypic studies have suggested that the flor formation of Z. rouxii occurs in a NaCl-dependent manner (Tomita and Yamamoto 1997). However, at the outset of this study, the molecular mechanism that underlies NaCl-dependent flor formation was unclear. In this study, we identified FLO11D as a gene responsible for buoyant biofilm formation in Z. rouxii (Figure 4), and the FLO11D gene expression is induced by osmostress under glucose-free conditions (Figure 5). Their osmo-ergic properties must permit the unique characteristics in Z. rouxii, such as osmostress-dependent buoyant biofilm formation.

Regulation of FLO11D

The osmostress-dependent expression of FLO11D is very unique. In S. cerevisiae, FLO11 is activated by nitrogen and carbon depletion via the MAP kinase-dependent filamentous growth pathway and the glucose repression pathway (Madhani and Fink 1997; Rupp et al. 1999; Gagiano et al. 2002; Kuchin et al. 2002; Vyas et al. 2003; Schwartz and Madhani 2004). Therefore, the formation of flor can be considered as an adaptive mechanism to nutrient starvation (Zara et al. 2011) because it ensures access to oxygen and therefore permits continued growth on nonfermentable ethanol. The glucose repression pathway between S. cerevisiae and Z. rouxii could be common because not only FLO11 expression but also FLO11D expression was repressed by glucose, suggesting that the flor formation is important to the adaption to nutrient starvation in Z. rouxii. Interestingly, a previous study showed that hyperosmotic stress inhibits the development of the fluffy colony morphology in S. cerevisiae (Furukawa et al. 2009), which are the opposite results from ours (Figure 2C). This fact raises the question of why the FLO11D-dependent morphological change is induced by osmostress in Z. rouxii. We hypothesize three possible reasons: (1) the FLO11D gene is under the HOG1 MAP kinase pathway, (2) there is signaling cross talk between the filamentous growth MAP kinase pathway and the HOG1 MAP kinase pathway because the two pathways share common components (Ste11p, Ste50p, Ste20p, and Cdc42p) in their signal transduction (Chen and Thorner 2007), or (3) there is an unknown pathway. To assess the possibility of hypothesis 1, we constructed the hog1Δ strain under the Z3 background and compared the expression level of FLO11D between Z3 and hog1Δ. As a result, FLO11D gene expression was observed under osmostress conditions in both Z3 and hog1Δ (data not shown). These results disprove hypothesis 1. We therefore infer that hypothesis 2 or 3 may contribute to the osmostress FLO11D expression. However, we cannot exclude the possibility that FLO11D activation in hog1Δ under osmostress conditions is an artificial consequence because hyperosmotic shock induces inappropriate activation of the MAP kinase-dependent filamentous growth pathway in S. cerevisiae hog1Δ (Davenport et al. 1999). Clearly, a more detailed analysis is needed to elucidate the pathway required for NaCl-dependent FLO11D expression in Z. rouxii. We speculate that the osmo-ergic FLO11D expression in Z. rouxii would be important for ecological niche segregation among yeast. For Z. rouxii, the adaptation to nutrient starvation thorough the formation of flor in a non-osmostress environment would not be effective for the expansion of Z. rouxii’s natural habitat because its competitor, S. cerevisiae, has advantages in growth rate in this type of environment. Conversely, the adaptation to nutrient starvation by forming flor observed in Z. rouxii under osmostress conditions would ensure its increased survival and thereby permit its occupancy of this type of niche.

Implications of FLO11D copy number amplification

We showed that there are FLO11D copy number variations in a Z. rouxii population and that the cellular hydrophobicity correlates with the FLO11D copy number (Figure 6 and Figure 7A). We also showed that the strain with more FLO11D copy numbers has a fitness advantage in a flor-formative environment, such as osmostress static culture conditions (Figure 7B), even though one copy of FLO11D is sufficient to form flor (Figure 4B and Figure 6B). These results suggest that the copy number of FLO11D is under positive selection under osmostress static culture conditions. A salt-containing food fermentation process, such as soy sauce and soy bean paste, is similar to the osmostress static culture conditions; therefore, these traditional food production processes can act as a positive selection pressure for the FLO11D copy number of Z. rouxii. This raises the question of why soy sauce mash is not occupied by flor-forming yeast with a high copy number of FLO11D. In fact, we demonstrated that the FLO11D copy number was variable in a Z. rouxii population isolated from salt-containing conditions (Figure 6). One possibility is that counterselection pressure exists in these artificial environments. During soy sauce fermentation, the flor is usually discarded by the manufacturer to maintain the organoleptic quality of the products, and a non-flor-forming strain of Z. rouxii is often added to soy sauce mash to stabilize the alcohol fermentation. The addition of yeast to the mash as a starter may lead to a cross between flor-forming and non-flor-forming yeast, which have variable FLO11D copy numbers, followed by sporulation.

In general, gene amplification (duplication) can have an important role in evolution through changes in the sequence, the acquisition of new or specialized functions (neofunctionalization or subfunctionalization), and increases in the expression level (gene dosage effect). For example, a high copy number of the human salivary amylase gene (AMY1), the yeast copper-binding metallothionein gene (CUP1), and the yeast high-affinity sulfate transporter (SUL1) acts as a gene dosage, and the yeast α-glucosidase gene MALS family exhibits aspects of three classical models, such as gene dosage, neo-, and subfunctionalization (Perry et al. 2007; Gresham et al. 2008; Warringer et al. 2011; Voordeckers et al. 2012). This study clearly indicates that the copy number amplification of FLO11D also contributes to adaptation in a gene-dosage-dependent manner. This study therefore may have profound implications that support part of the classical models of gene duplication built with past knowledge.

In this study, we have demonstrated that the osmo-ergic FLO11D gene is responsible for the NaCl-dependent morphological changes in Z. rouxii. Through a gene amplification event and natural selection during its evolution, Z. rouxii acquired an additional copy of the FLO11D gene, enabling its further adaptation to flor-formative environments, such as osmostress static culture conditions.

Supplementary Material

Supporting Information:

Acknowledgments

We thank Jose I. Ibeas for the yeast strains, two referees for their constructive comments, and the members of the soy sauce lab for fruitful discussions.

Footnotes

Communicating editor: M. Hampsey

Literature Cited

  • Altschul S. F., Madden T. L., Schäffer A. A., Zhang J., Zhang Z., et al. , 1997.  Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res. 25: 3389–3402. [Europe PMC free article] [Abstract] [Google Scholar]
  • Aminetzach Y. T., Macpherson J. M., Petrov D. A., 2005.  Pesticide resistance via transposition-mediated adaptive gene truncation in Drosophila. Science 309: 764–767. [Abstract] [Google Scholar]
  • Barrales R. R., Jimenez J., Ibeas J. I., 2008.  Identification of novel activation mechanisms for FLO11 regulation in Saccharomyces cerevisiae. Genetics 178: 145–156. [Europe PMC free article] [Abstract] [Google Scholar]
  • Barrales R. R., Korber P., Jimenez J., Ibeas J. I., 2012.  Chromatin modulation at the FLO11 promoter of Saccharomyces cerevisiae by HDAC and Swi/Snf complexes. Genetics 191: 791–803. [Europe PMC free article] [Abstract] [Google Scholar]
  • Barrick J. E., Lenski R. E., 2009.  Genome-wide mutational diversity in an evolving population of Escherichia coli. Cold Spring Harb. Symp. Quant. Biol. 74: 119–129. [Europe PMC free article] [Abstract] [Google Scholar]
  • Brown C. J., Todd K. M., Rosenzweig R. F., 1998.  Multiple duplications of yeast hexose transport genes in response to selection in a glucose-limited environment. Mol. Biol. Evol. 15: 931–942. [Abstract] [Google Scholar]
  • Bumgarner S. L., Dowell R. D., Grisafi P., Gifford D. K., Fink G. R., 2009.  Toggle involving cis-interfering noncording RNAs controls variegated gene expression in yeast. Proc. Natl. Acad. Sci. USA 106: 18321–18326. [Europe PMC free article] [Abstract] [Google Scholar]
  • Bumgarner S. L., Neuert G., Voight B. F., Symbor-Nagrabska A., Grisafi P., et al. , 2012.  Single-cell analysis reveals that noncoding RNAs contribute to clonal heterogeneity by modulating transcription factor recruitment. Mol. Cell 45: 470–482. [Europe PMC free article] [Abstract] [Google Scholar]
  • Bürger R., 2000.  The Mathematical Theory of Selection, Recombination, and Mutation. John Wiley & Sons, New York; Hoboken, NJ. [Google Scholar]
  • Chen R. E., Thorner J., 2007.  Function and regulation in MAPK signaling pathwasy: lessons learned from the yeast Saccharomyces cerevisiae. Biochim. Biophys. Acta 1773: 1311–1340. [Europe PMC free article] [Abstract] [Google Scholar]
  • Conrad D. F., Pinto D., Redon R., Feuk L., Gokcumen O., et al. , 2010.  Origins and functional impact of copy number variation in the human genome. Nature 464: 704–712. [Europe PMC free article] [Abstract] [Google Scholar]
  • Cooper G. M., Nickerson D. A., Eichler E. E., 2007.  Mutational and selective effects on copy number variants in the human genome. Nat. Genet. 39: S22–S29. [Abstract] [Google Scholar]
  • Cressman R., 2003.  Evolutionary Dynamics and Extensive Form Games. MIT Press, Cambridge, MA. [Google Scholar]
  • Davenport K. D., Williams K. E., Ullmann B. D., Gustin M. C., 1999.  Activation of the Saccharomyces cerevisiae filamentation/invasion pathway by osmotic stress in high-osmolarity glycogen pathway mutants. Genetics 153: 1091–1103. [Europe PMC free article] [Abstract] [Google Scholar]
  • Eisenhaber B., Schneider G., Wildpaner M., Eisenhaber F., 2004.  A sensitive predictor for potential GPI lipid modification sites in fungal protein sequences and its application to genome-wide studies for Aspergillus nidulans, Candida albicans, Neurospora crassa, Saccharomyces cerevisiae and Schizosaccharomyces pombe. J. Mol. Biol. 337: 243–253. [Abstract] [Google Scholar]
  • Emanuelsson O., Brunak S., von Heijne G., Nielsen H., 2007.  Locating proteins in the cell using TargetP, SignalP and related tools. Nat. Protoc. 2: 953–971. [Abstract] [Google Scholar]
  • Ewens W. J., 2004.  Mathematical Population Genetics. Springer, New York. [Google Scholar]
  • Fidalgo M., Barrales R. R., Ibeas J. I., Jimenez J., 2006.  Adaptive evolution by mutation in the FLO11 gene. Proc. Natl. Acad. Sci. USA 103: 11228–11233. [Europe PMC free article] [Abstract] [Google Scholar]
  • Fidalgo M., Barrales R. R., Jimenez J., 2008.  Cording repeat instability in the FLO11 gene of Saccharomyces yeasts. Yeast 25: 879–889. [Abstract] [Google Scholar]
  • Finn R. D., Mistry J., Tate J., Coggill P., Heger A., et al. , 2010.  The Pfam protein families database. Nucleic Acids Res. 38: D211–D222. [Europe PMC free article] [Abstract] [Google Scholar]
  • Furukawa K., Sidoux-Walter F., Hohmann S., 2009.  Expression of the yeast aquaporin Aqy2 affects cell surface properties under the control of osmoregulatory and morphogenic signalling pathways. Mol. Microbiol. 74: 1272–1286. [Abstract] [Google Scholar]
  • Gagiano M., Bauer F. F., Pretorius I. S., 2002.  The sensing of nutritional status and the relationship to filamentous growth in Saccharomyces cerevisiae. FEMS Yeast Res. 2: 433–470. [Abstract] [Google Scholar]
  • Gietz R. D., Schiestl R. H, Willems A. R., Woods R. A., 1995.  Studies on the transformation of intact yeast cells by the LiAc/SS-DNA/PEG procedure. Yeast 11: 355–360. [Abstract] [Google Scholar]
  • Goossens K. V., Willaert R. G., 2012.  The N-terminal domain of the Flo11 protein from Saccharomyces cerevisiae is an adhesion without mannose-binding activity. FEMS Yeast Res. 12: 78–87. [Abstract] [Google Scholar]
  • Gordon J. L., Wolfe K. H., 2008.  Recent allopolyploid origin of Zygosaccharomyces rouxii strain ATCC42981. Yeast 25: 449–456. [Abstract] [Google Scholar]
  • Gresham D., Desai M. M., Tucker C. M., Jenq H. T., Pai D. A., et al. , 2008.  The repertoire and dynamics of evolutionary adaptations to controlled nutrient-limited environments in yeast. PLoS Genet. 4: e1000303. [Europe PMC free article] [Abstract] [Google Scholar]
  • Gresham D., Usaite R., Germann S. M., Lisby M., Botstein D., et al. , 2010.  Adaptation to diverse nitrogen-limited environments by deletion or extrachromosomal element formation of the GAP1 locus. Proc. Natl. Acad. Sci. USA 107: 18551–18556. [Europe PMC free article] [Abstract] [Google Scholar]
  • Guo B., Styles C. A., Feng Q., Fink G. R., 2000.  A Saccharomyces gene family involved in invasive growth, cell-cell adhesion, and mating. Proc. Natl. Acad. Sci. USA 97: 12158–12163. [Europe PMC free article] [Abstract] [Google Scholar]
  • Halme A., Bumgarner S., Styles C., Fink G. R., 2004.  Genetic and epigenetic regulation of the FLO gene family generates cell-surface variation in yeast. Cell 116: 405–415. [Abstract] [Google Scholar]
  • Hurles M. E., Dermitzakis E. T., Tyler-Smith C., 2010.  The functional impact of structural variation in humans. Trends Genet. 24: 238–245. [Europe PMC free article] [Abstract] [Google Scholar]
  • Innan H., Kondrashov F., 2010.  The evolution of gene duplications: classifying and distinguishing between models. Nat. Rev. Genet. 11: 97–108. [Abstract] [Google Scholar]
  • Iskow R. C., Gokcumen O., Lee C., 2012.  Exploring the role of copy number variants in human adaptation. Trends Genet. 28: 245–257. [Europe PMC free article] [Abstract] [Google Scholar]
  • James S. A., Bond C. J., Stratford M., Roberts I. N., 2005.  Molecular evidence for the existence of natural hybrids in the genus Zygosaccharomyces. FEMS Yeast Res. 5: 747–755. [Abstract] [Google Scholar]
  • Kuchin S., Vyas V. K., Carlson M., 2002.  Snf1 protein kinase and the repressors Nrg1 and Nrg2 regulate FLO11, haploid invasive growth, and diploid pseudohyphal differentiation. Mol. Cell. Biol. 22: 3994–4000. [Europe PMC free article] [Abstract] [Google Scholar]
  • Larkin M. A., Blackshields G., Brown N. P., Chenna R., McGettigan P. A., et al. , 2007.  Clustal W and Clustal X version 2.0. Bioinformatics 23: 2947–2948. [Abstract] [Google Scholar]
  • Lim D., Trivedi H., Nath K., 1994.  Determination of Gardnerella vaginalis genome size by pulsed-field gel electrophoresis. DNA Res. 1: 115–122. [Abstract] [Google Scholar]
  • Linder T., Gustafsson C. M., 2008.  Molecular phylogenetics of ascomycotal adhesins-A novel family of putative cell-surface adhesive proteins in fission yeasts. Fungal Genet. Biol. 45: 485–497. [Abstract] [Google Scholar]
  • Madhani H. D., Fink G. R., 1997.  Combinatorial control required for the specificity of yeast MAPK signaling. Science 275: 1314–1317. [Abstract] [Google Scholar]
  • Nowak M. A., Sigmund K., 2004.  Evolutionary dynamics of biological games. Science 303: 793–799. [Abstract] [Google Scholar]
  • Octavio L. M., Gedeon K., Maheshri N., 2009.  Epigenetic and conventional regulation is distributed among activators of FLO11 allowing tuning of population-level heterogeneity in its expression. PLoS Genet. 5: e1000673. [Europe PMC free article] [Abstract] [Google Scholar]
  • Ohno S., 1970.  Evolution by Gene Duplication. Springer-Verlag, Berlin. [Google Scholar]
  • Perrière G., Gouy M., 1996.  WWW-Query: an on-line retrieval system for biological sequence banks. Biochimie 78: 364–369. [Abstract] [Google Scholar]
  • Perry G. H., Dominy N. J., Claw K. G., Lee A. S., Fiegler H., et al. , 2007.  Diet and the evolution of human amylase gene copy number variation. Nat. Genet. 39: 1256–1260. [Europe PMC free article] [Abstract] [Google Scholar]
  • Pribylova L., de Montigny J., Sychrova H., 2007a Osmoresistant yeast Zygosaccharomyces rouxii: the two most studied wild-type strains (ATCC2623 and ATCC42981) differ in osmotolerance and glycerol metabolism. Yeast 24: 171–180. [Abstract] [Google Scholar]
  • Pribylova L., de Montigny J., Sychrova H., 2007b Tools for the genetic manipulation of Zygosaccharomyces rouxii. FEMS Yeast Res. 7: 1285–1294. [Abstract] [Google Scholar]
  • Rupp S., Summers E., Lo H. J., Madhani H., Fink G., 1999.  MAP kinase and cAMP filamentation signaling pathways converge on the unusually large promoter of the yeast FLO11 gene. EMBO J. 18: 1257–1269. [Europe PMC free article] [Abstract] [Google Scholar]
  • Saitou N., Nei M., 1987.  The neighbor-joining method: a new method for reconstructing phylogenetic trees. Mol. Biol. Evol. 4: 406–425. [Abstract] [Google Scholar]
  • Sambrook J., Russell D. W., 2001.  Molecular Cloning: A Laboratory Manual. Cold Spring Habor Laboratory Press, Cold Spring Harbor, NY. [Google Scholar]
  • Schlattl A., Anders S., Waszak S. M., Huber W., Korbel J. O., 2011.  Relating CNVs to transcriptome data at fine resolution: assessment of the effect of variant size, type, and overlap with functional regions. Genome Res. 21: 2004–2013. [Europe PMC free article] [Abstract] [Google Scholar]
  • Schwartz M. A., Madhani H. D., 2004.  Principles of MAP kinase signaling specificity in Saccharomyces cerevisiae. Annu. Rev. Genet. 38: 725–748. [Abstract] [Google Scholar]
  • Sherman D. J., Martin T., Nikolski M., Cayla C., Souciet J. L., et al. , 2009.  Génolevures: protein families and synteny among complete hemiascomycetous yeast proteomes and genomes. Nucleic Acids Res. 37: D550–D554. [Europe PMC free article] [Abstract] [Google Scholar]
  • Smukalla S., Caldara M., Pochet N., Beauvais A., Guadagnini S., et al. , 2008.  FLO1 is a variable green beard gene that drives biofilm-like cooperation in budding yeast. Cell 135: 726–737. [Europe PMC free article] [Abstract] [Google Scholar]
  • Solieri L., Landi S., De Vero L., Giudici P., 2006.  Molecular assessment of indigenous yeast population from traditional balsamic vinegar. J. Appl. Microbiol. 101: 63–71. [Abstract] [Google Scholar]
  • Solieri L., Cassanelli S., Giudici P., 2007.  A new putative Zygosaccharomyces yeast species isolated from traditional balsamic vinegar. Yeast 24: 403–417. [Abstract] [Google Scholar]
  • Solieri L., Dakal C. T., Croce A. M., Giudici P., 2013.  Unravelling genomic diversity of the Zygosaccharomyces rouxii complex with a link to its life cycle. FEMS Yeast Res. 13: 245–258. [Abstract] [Google Scholar]
  • Steiner C. C., Weber J. N., Hoekstra H. E., 2007.  Adaptive variation in beach mice produced by two interacting pigmentation genes. PLoS Biol. 5: e219. [Europe PMC free article] [Abstract] [Google Scholar]
  • Stranger B. E., Forrest M. S., Dunning M., Ingle C. E., Beazley C., et al. , 2007.  Relative impact of nucleotide and copy number variation on gene expression phenotypes. Science 315: 848–853. [Europe PMC free article] [Abstract] [Google Scholar]
  • Tanaka Y., Watanabe J., Mogi Y., 2012.  Monitoring of the microbial communities involved in the soy sauce manufacturing process by PCR-denaturing gradient gel electrophoresis. Food Microbiol. 31: 100–106. [Abstract] [Google Scholar]
  • Teunissen A. W. R. H., Ateensma H. Y., 1995.  The dominant flocculation genes of Saccharomyces cerevisiae constitute a new subtelomeric gene family. Yeast 11: 1001–1013. [Abstract] [Google Scholar]
  • Tomita M., Yamamoto S., 1997.  Physiological characteristics of a halo-tolerant yeast Zygosacchaormyces rouxii (2)-film-forming strains grown on salty foods. J. Brewing Soc. Japan 92: 853–859. [Google Scholar]
  • Verstrepen K. J., Klis F. M., 2006.  Flocculation, adhesion and biofilm formation in yeasts. Mol. Microbiol. 60: 5–15. [Abstract] [Google Scholar]
  • Verstrepen K. J., Jansen A., Lewitter F., Fink G. R., 2005.  Intragenic tandem repeats generate functional variability. Nat. Genet. 37: 986–990. [Europe PMC free article] [Abstract] [Google Scholar]
  • Voordeckers K., Brown C. A., Vanneste K., van der Zande E., Voet A., et al. , 2012.  Reconstruction of ancestral metabolic enzymes reveals molecular mechanisms underlying evolutionary innovation through gene duplication. PLoS Biol. 10: e1001446. [Europe PMC free article] [Abstract] [Google Scholar]
  • Vyas V. K., Kuchin S., Berkey C. D., Carlson M., 2003.  Snf1 kinase with different beta-subunit isoforms play distinct foles in regulating haploid invasive growth. Mol. Cell. Biol. 23: 1341–1348. [Europe PMC free article] [Abstract] [Google Scholar]
  • Warringer J., Zörgö E., Cubillos F. A., Zia A., Gjuvsland A., et al. , 2011.  Trait variation in yeast is defined by population history. PLoS Genet. 7: e1002111. [Europe PMC free article] [Abstract] [Google Scholar]
  • Watanabe J., Uehara K., Mogi Y., Suzuki K., Watanabe T., et al. , 2010.  Improved transformation of the halo-tolerant yeast Zygosaccharomyces rouxii by electroporation. Biosci. Biotechnol. Biochem. 74: 1092–1094. [Abstract] [Google Scholar]
  • Watanabe J., Uehara K., Mogi Y., 2013.  Diversity of mating-type chromosome structures in the yeast Zygosaccharomyces rouxii caused by ectopic exchanges between MAT-like loci. PLoS ONE 8: e62121. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wilke C. M., Adams J., 1992.  Fitness effects of Ty transposition in Saccharomyces cerevisiae. Genetics 131: 31–42. [Europe PMC free article] [Abstract] [Google Scholar]
  • Yamanaka M., Kato Y., Angata T., Narimatsu H., 2009.  Deletion polymorphism of SIGLEC14 and its functional implications. Glycobiology 19: 841–846. [Abstract] [Google Scholar]
  • Zara S., Bakalinsky A. T., Zara G., Pirino G., Demontis M. A., et al. , 2005.  FLO11-based model for air-liquid interfacial biofilm formation by Saccharomyces cerevisiae. Appl. Environ. Microbiol. 71: 2934–2939. [Europe PMC free article] [Abstract] [Google Scholar]
  • Zara G., Zara S., Pinna C., Marceddu S., Budroni M., 2009.  FLO11 gene length and transcriptional level affect biofilm-forming ability of wild flor strains of Saccharomyces cerevisiae. Microbiology 155: 3838–3846. [Abstract] [Google Scholar]
  • Zara G., Budroni M., Mannazzu I., Zara S., 2011.  Air-liquid biofilm formation is dependent on ammonium depletion in Saccharomyces cerevisiae flor strain. Yeast 28: 809–814. [Abstract] [Google Scholar]

Articles from Genetics are provided here courtesy of Oxford University Press

Citations & impact 


Impact metrics

Jump to Citations
Jump to Data

Citations of article over time

Article citations


Go to all (10) article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.