Skip to main content

The SREBP (Sterol Regulatory Element-Binding Protein) pathway: a regulatory bridge between carotenogenesis and sterol biosynthesis in the carotenogenic yeast Xanthophyllomyces dendrorhous

Abstract

Xanthophyllomyces dendrorhous is a basidiomycete yeast that naturally produces the red–orange carotenoid astaxanthin, which has remarkable antioxidant properties. The biosynthesis of carotenoids and sterols share some common elements that have been studied in X. dendrorhous. For example, their synthesis requires metabolites derived from the mevalonate pathway and in both specific pathways, cytochrome P450 enzymes are involved that share a single cytochrome P450 reductase, CrtR, which is essential for astaxanthin biosynthesis, but is replaceable for ergosterol biosynthesis. Research on the regulation of carotenoid biosynthesis is still limited in X. dendrorhous; however, it is known that the Sterol Regulatory Element-Binding Protein (SREBP) pathway, which is a conserved regulatory pathway involved in the control of lipid metabolism, also regulates carotenoid production in X. dendrorhous. This review addresses the similarities and differences that have been observed between mammal and fungal SREBP pathways and what it is known about this pathway regarding the regulation of the production of carotenoids and sterols in X. dendrorhous.

Background

The use of microbial platforms as natural carotenoid producers has gained interest as an alternative to the production of carotenoids by chemical synthesis due to health concern of synthetic carotenoids [1]. Even the cost associated with the production of carotenoids using microorganisms could be reduced using agro-industrial wastes as low-cost substrates, which also have an ecological aspect [2, 3]. The basidiomycetous yeast Xanthophyllomyces dendrorhous is a natural source of astaxanthin. This red–orange carotenoid has remarkable antioxidant activity [4] and is gaining increasing attention as evidence indicates that it is an effective molecule preventing oxidative stress-mediated and age-related diseases [5]. Astaxanthin is also used in aquaculture as a food additive, principally because it improves muscle pigmentation of aquatic species, and subsequently product quality and price [6]. There are several reviews related to astaxanthin biosynthesis in X. dendrorhous [7,8,9,10] that make this yeast one of the best candidates for commercial production of astaxanthin; however, this process has not been economically efficient to date [11]. On the other hand, research on the mechanisms regulating the carotenoid production, which could be used as a tool to improve the production of carotenoids in X. dendrorhous, is still limited. It is known that astaxanthin production is regulated by environmental and nutritional conditions. For example, reactive oxygen species induce carotenoid production, which is probably related to the antioxidant protective role of carotenoids [12,13,14]. At the nutritional level, the production of carotenoids in X. dendrorhous is regulated by glucose‐dependent repression, depending on the DNA‐binding regulator Mig1 and the co‐repressor complex Cyc8-Tup1, which repress the expression of several genes including genes involved in carotenogenesis [15, 16]. Recently, it has been demonstrated that carotenogenesis in X. dendrorhous is regulated by the SREBP (Sterol Regulatory Element-Binding) pathway [17], which is a conserved pathway in several organisms involved in lipid homeostasis that is activated depending on sterol demand [18]. In X. dendrorhous, this pathway regulates several genes of the mevalonate (MVA) pathway and from ergosterol biosynthesis; interestingly, it also regulates two carotenogenic genes (Fig. 1) [17]. Therefore, considering the aforementioned findings, the goal of this review is to gather and discuss the latest reports related to the SREBP pathway in fungi, focusing on the common aspects found in the regulation of the biosynthesis of sterols and carotenoids in X. dendrorhous.

Fig. 1
figure 1

Biosynthesis of astaxanthin and ergosterol in X. dendrorhous. The production of carotenoids and sterols requires isopentenyl pyrophosphate (IPP) from the mevalonate pathway [9]. Carotenoid biosynthesis begins with the production of geranylgeranyl pyrophosphate (GGPP) from farnesyl pyrophosphate (FPP) and IPP by GGPP synthase encoded by crtE [66, 68]. Then, the bi-functional enzyme phytoene-β-carotene synthase encoded by crtYB condenses two GGPP molecules producing phytoene [69]. Subsequently, phytoene undergoes four desaturation reactions carried out by the phytoene desaturase enzyme encoded by crtI, producing lycopene [70]. The cyclization of both ends of lycopene by the lycopene cyclase activity of the phytoene-β-carotene synthase produces β-carotene [69], which is then transformed into astaxanthin via intermediate xanthophylls. To date, X. dendrorhous is the only known organism that produces astaxanthin from β-carotene through a cytochrome P450 system [71], which is composed of the cytochrome P450 enzyme astaxanthin synthase (CrtS, encoded by crtS) [72, 73] and a cytochrome P450 reductase (CPR) (named CrtR in X. dendrorhous, encoded by crtR) [64]. CrtS catalyzes the hydroxylation and ketolation of carbons at positions 3 and 4, respectively, of each end ring of β-carotene to finally produce astaxanthin [72], while CrtR assists CrtS in these reactions [64]. In ergosterol biosynthesis, two cytochrome P450 enzymes are involved, which are encoded by CYP51 (lanosterol 14α-demethylase) [74] and CYP61 (C-22 sterol desaturase) [49]. Arrows: catalytic steps with the corresponding enzyme encoding gene. Genes in red: Direct Sre1 targets identified by ChIP-exo [17]. CrtR was included as a redox partner of the cytochrome P450 monooxygenases Cyp51, Cyp61 and CrtS, and squalene epoxidase (ERG1). 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA), mevalonate (MVA), dimethylallyl-pyrophosphate (DMAPP). Other genes: ERG10 (acetyl-CoA C-acetyltransferase), HMGS (HMG-CoA synthase), HMGR (HMG-CoA reductase), MVK (mevalonate kinase), PMK (phosphomevalonate kinase), MVD (mevalonate diphosphate decarboxylase), idi (IPP isomerase), FPS (FPP synthase). ERG represent to enzyme-encoding genes involved in ergosterol biosynthesis identified in Saccharomyces cerevisiae. Figure adapted from [17]

The SREBP pathway in mammals and in fungi

The SREBP pathway is involved in the regulation of lipid homeostasis and metabolism, and our knowledge originates primarily from studies in mammalian cells. Therefore, it is necessary to highlight the key elements of the mammalian SREBP pathway (Fig. 2A), before establishing the differences found in fungi, specifically in X. dendrorhous. The central components of the SREBP pathway are the SREBP proteins, which are a family of basic-helix-loop-helix leucine zipper (bHLH-LZ) transcription factors that are initially synthesized as inactive precursors that are anchored to the Endoplasmic Reticulum (ER) membrane [19]. In mammals, three isoforms of these transcription factors have been described: SREBP-1a, SREBP-1c [20, 21] and SREBP-2 [22]. The first two are encoded by a single gene and differ due to the use of alternative transcription start sites. In all three cases, the precursor form is bound to the ER membrane in a harpin fashion with the NH2-terminal (the transcription factor domain, bHLH-LZ) and COOH-terminal (the regulatory domain) domains facing the cytoplasm. These terminal domains are separated by a short hydrophilic sequence (the lumenal loop) which projects into the lumen of the ER [23]. In the ER membrane, SREBP may form a complex with the SREBP cleavage-activating protein (SCAP), which itself harbors a sterol sensing domain [24]. Specifically, this interaction occurs between the WD repeat domain of the SCAP C-terminal domain and the SREBP C-terminal regulatory domain [25]. When sterol levels are sufficient, the Insig (Insulin induced gene) proteins bind to cholesterol-loaded SCAP protein to retain the SCAP-SREBP complex in the ER [26, 27]. On the other hand, when sterol levels decrease, the SCAP-SREBP complex clusters into COPII-coated vesicles that bud from the ER [28], which are then transported to the Golgi apparatus where SREBP is sequentially processed by proteolytic cleavage for activation in two steps. First, site-1 protease (S1P, a subtilisin-related serine protease) cuts SREBP in its ER lumenal loop [29] and next, site-2 protease (S2P, a metallopeptidase) hydrolyzes SREBP within the first transmembrane segment [30]. As a result, the N-terminal bHLH-LZ domain of SREBP is released and migrates to the nucleus where it activates the transcription of target genes by binding to Sterol Regulatory Elements (SREs) at their promoter region [21].

Fig. 2
figure 2

The SREBP pathway in A mammals and B X. dendrorhous. The SREBP pathway is involved in the regulation of lipid homeostasis and metabolism, and is regulated by several independent mechanisms including the use of subcellular compartmentalization to ensure its activation, when required. In sterol-replete mammalian cells, the Insig proteins (isoforms: Insig-1 and Insig-2) bind cholesterol-loaded protein SCAP to retain the SCAP-SREBP complex in the endoplasmic reticulum membrane. On the other hand, when cells are depleted of sterols, the SREBP-SCAP complex is transported to the Golgi apparatus where SREBP is sequentially processed by proteases S1P and S2P. As a result, the N-terminal domain (NH2) of SREBP, which contains the basic helix-loop-helix leucine zipper (bHLH-LZ) domain, is released, migrates to the nucleus, and binds as a dimer to the sterol regulatory elements (SREs) in the promoter region of target genes activating their transcription. In X. dendrorhous, the SREBP homolog is Sre1, which is processed by a S2P homolog, named Stp1. Figure adapted from [17]

Components of the SREBP pathway in ascomycete and basidiomycete fungi

SREBP-like protein encoding genes have been identified in several fungal genomes, and some have proven to be functional (Table 1). The SREBP pathway in fungi is also involved in lipid homeostasis and metabolism, and in some cases, it has been demonstrated that it is activated under low oxygen conditions regulating the hypoxic response. Most studies of fungal SREBP pathways have been carried out in the ascomycetes Schizosaccharomyces pombe [31] and species of the genus Aspergillus such as Aspergillus fumigatus [32], while studies in basidiomycetes have focused on Cryptococcus neoformans [33, 34]. In these fungi, the SREBP pathway is also involved in resistance to azole compounds, which are antifungal drugs that negatively affect ergosterol biosynthesis [35], and in pathogenesis in A. fumigatus and C. neoformans [32, 34]. Novel functions of the SREBP pathway in fungi include the maintenance of cell polarity in A. fumigatus [32], and the existence of a regulatory link in protein secretion under lignocellulolytic conditions in Neurospora crassa and Trichoderma reesei [36, 37]. Differences in the components of the SREBP pathway, including the proteolytic activation mechanism of the transcription factor SREBP, between the mammalian and fungal SREBP pathways have been detected, as well as dissimilarities between ascomycetes and basidiomycetes, and even differences within each division (Table 1).

Table 1 Components of SREBP pathways described in some fungi

Ascomycetes (S. pombe, species of the genus Aspergillus and N. crassa)

Schizosaccharomyces pombe

In the fission yeast S. pombe, homologs of SREBP, SCAP and Insig proteins have been identified (Sre1, Scp1 and Ins1, respectively) [31]. Interestingly, S. pombe has a second SREBP homolog (Sre2), whose role is still unknown [31]. The proteolytic activation of S. pombe Sre1 is mediated by a different mechanism to the one described in mammalian cells, as it lacks S1P and S2P homologs. Sre1 activation depends on the Golgi Dsc (defective for SREBP cleavage) E3 ligase complex (proteins Dsc1 to Dsc5; Dsc1 has E3 ubiquitin ligase activity) [38, 39] and rhomboid protein 2 (Rbd2) [40], that binds the AAA-ATPase Cdc48, which probably recognizes ubiquitinylated Sre1 and recruits it for Rbd2 cleavage. However, a second cut in Sre1 by another protease is not ruled out [41].

Species of the genus Aspergillus

In Aspergillus, the proteolytic processing of the SREBP homolog (SrbA) [32, 42] appears to be like that of S. pombe since A. fumigatus and A. nidulans also lack S1P and S2P homologs. In A. fumigatus, the homologs of the Golgi Dsc E3 ligase complex (DscA to DscD) [43], and rhomboid protease (RbdB) [44] are involved in SrbA processing. On the other hand, a second SREBP homolog identified in A. fumigatus (SrbB), is probably independent of proteolytic cleavage as no transmembrane domains are predicted in its structure [45]. Interestingly, A. nidulans SrbA is sequentially processed by Dsc-linked proteolysis followed by an aspartyl protease (SppA) [42]. Unlike the fission yeast, no homologs of SCAP have been detected in Aspergillus. Interestingly, a SppA homolog (Ypf1) was also detected in S. pombe, but it is not involved in the response to hypoxia conditions [42].

Neurospora crassa

Like S. pombe and A. fumigatus, N. crassa has homologs of SREBP (SAH-2 and SRE-2), SCAP (SCP-1), SppA (Spp-1), Rbd2 (RBD-2) and of components of the Golgi Dsc E3 ligase complex (Dsc-1 to Dsc-6) [36]. In this ascomycete, proteins SRE-2 and Spp-1 are not related to hypoxia adaptation and are therefore not required for the function of the SREBP pathway. Based on a model, the activation of SAH-2 probably involves homologs of the Golgi Dsc E3 ligase complex and rhomboid protease Rbd2 [36].

Basidiomycetes (C. neoformans and X. dendrorhous)

Cryptococcus neoformans

In the case of the basidiomycete C. neoformans, unlike S. pombe and Aspergillus, it was shown that the SREBP homolog (Sre1) [33] is processed by a homolog of mammalian S2P (Stp1) [34, 46]. Interestingly, like S. pombe and N. crassa, this basidiomycete has a SCAP homolog, which was shown to be involved in the SREBP pathway [33].

Xanthophyllomyces dendrorhous

In the basidiomycete X. dendrorhous, SREBP and S2P homologs were recently described (Sre1 and Stp1, respectively), which were proven to be involved in the SREBP pathway in this yeast [47, 48] (Fig. 2B).

The SREBP pathway in X. dendrorhous

Xanthophyllomyces dendrorhous produces the carotenoid astaxanthin and a carotenoid overproduction phenotype was observed in an ergosterol biosynthesis mutant, which was one of the first evidence that suggested that carotenoid production in this yeast could be regulated, at least in part, by the SREBP pathway. Cyp61 is a cytochrome P450 enzyme that catalyzes the second last step of ergosterol biosynthesis (Fig. 1), and carotenoid content increased approximately twofold compared to those of the wild-type strain when this gene was interrupted [49]. Recent works revealed that this phenotype depends on the SREBP pathway as it was demonstrated that the X. dendrorhous SREBP homolog was activated in the cyp61 mutant and mutations that avoided Sre1 activation in this strain, brought carotenoid levels back to wild levels [47, 48]. This section summarizes our current knowledge of the SREBP pathway in X. dendrorhous.

Xanthophyllomyces dendrorhous Sre1 is conserved and is probably activated independent of SCAP

Xanthophyllomyces dendrorhous Sre1 has the characteristic bHLH-LZ DNA binding domain with a tyrosine (Y364), which distinguishes SREBPs from other bHLH transcription factors [50]. It also lacks a DUF2014 domain [48], which is a domain of unknown function that is present at the C-terminal of some SREBP homologs and may be important for interaction between SREBP and SCAP, and therefore, for the ER membrane retention of SREBP [51]. However, a potential SCAP encoding gene is not distinguished in the X. dendrorhous genome, suggesting that this yeast lacks a SCAP homolog. As in X. dendrorhous, C. neoformans Sre1 also lacks the DUF2014 domain, but this basidiomycete does have a SCAP homolog (Scp1) [33]. A. fumigatus SrbA harbors the DUF2014 domain, but this ascomycete lacks a SCAP homolog [42]. Thus, it is possible that X. dendrorhous has a SCAP-independent Sre1 activation mechanism that could also be conserved in other fungi lacking SCAP, like the pathogen A. fumigatus. In mammals, the SCAP and the Insig proteins are important components of the SREBP pathway, as they bind cholesterol and cholesterol hydroxylated derivatives (such as 25-hydroxycholesterol), respectively [52, 53], and in this way they regulate SREBP retention at the ER membrane. Like C. neoformans and Aspergillus, X. dendrorhous lacks an Insig homolog, and, even though an Insig homolog was identified in S. pombe, it is not involved in the SREBP pathway of this yeast [31]. This evidence suggests that the SREBP pathways in fungi are Insig-independent and in some fungal species, this pathway may also be SCAP-independent, as in the case of X. dendrorhous.

Sre1 is cleaved by Stp1, but the “sterol level sensor” and the “sterol-signal” that induce Sre1 activation by proteolytic cleavage in X. dendrorhous are still unknown

Xanthophyllomyces dendrorhous lacks a S1P protease homolog involved in Sre1 cleavage, but it is known that a S2P protease homolog (Stp1) is involved in this process [47]. A similar Sre1 activation mechanism was described in C. neoformans, as although it also lacks a S1P homolog, it does possess a S2P homolog involved in Sre1 activation [46]. However, the SCAP homolog of C. neoformans is required for Sre1 activation in response to lower sterol levels under low oxygen conditions [33]. In S. pombe, the Sre1-Scp1 complex senses ergosterol, and Sre1 transport and activation depends on the ergosterol concentration in the ER [54]. In X. dendrorhous, the sterol signal that favors Sre1 activation is still unknown. However, in mutants that do not produce ergosterol and overproduce carotenoids (cyp61 mutants), Sre1 is mainly in its activated form (Sre1N, N-terminal bHLH-LZ domain of Sre1) under standard laboratory conditions, correlating with the higher transcript levels of genes regulated by Sre1 that may be responsible in part for the carotenoid overproduction phenotype [47]. However, other ergosterol biosynthesis mutants (mutants of genes ERG3 and ERG4) that also do not produce ergosterol, do not overproduce carotenoids, and transcript levels of genes regulated by Sre1 are the same as in the wild-type strain [55]. These observations suggest that it is not the absence of ergosterol itself that triggers Sre1 activation in X. dendrorhous, but rather Sre1 activation depends on other alterations in sterol composition.

Additional regulation mechanisms on SREBPs

Besides proteolytic activation, SREBPs are regulated by covalent modifications and by interaction with other proteins. This has been studied more in mammals than in fungi. In mammals, regulation of SREBP by phosphorylation-dependent degradation has been reported [56, 57]. For example, Glycogen Synthase Kinase—3 (GSK-3) has been implicated in the phosphorylation of the precursor form of SREBP-1c in a specific serine [58]. Thus, phosphorylated SREBP-1c has lower affinity for SCAP, and free SREBP-1c is targeted for degradation [58]. In S. pombe, the active Sre1 transcription factor (Sre1N) is regulated by a casein kinase 1 family member (Hhp2) that promotes Sre1N proteasomal degradation through phosphorylation of Sre1N at specific residues [59]. In the X. dendrorhous genome, genes encoding putative kinase homologs to S. pombe Hhp2 (CDZ96742.1) and to mammalian GSK-3 (CDZ96841.1) were identified, and several serine and threonine residues of Sre1N are predicted to be potential phosphorylation sites. However, studies are needed to determine if indeed phosphorylation by these potential kinases or by others has a role in the regulation of Sre1 in X. dendrorhous. Additionally, in S. pombe the protein Ofd1 (2-oxo-glutarate Fe(II) dioxygenase) accelerates Sre1N proteasomal degradation and inhibits Sre1N DNA binding under normoxic conditions [60]. X. dendrorhous has a potential Ofd1 homolog (CED84823.1), but it may not be involved in the X. dendrorhous SREBP pathway as the phenotype of strains that only produce the active Sre1 transcription factor was not modified with the OFD1 mutation [61].

Role of the SREBP pathway in X. dendrorhous

As a SREBP homolog, X. dendrorhous Sre1 is involved in lipid homeostasis and in resistance against antifungal drugs [47, 48], which is observed in A. fumigatus by the direct regulation by Sre1 of a cdr1B homolog gene [17] that encodes an ABC transporter that contributes to the resistance to azoles [62]. Interestingly, A. fumigatus cdr1B is regulated by AtrR, which is a transcription factor that also regulates the synthesis of ergosterol and shares target genes with SrbA [63]. At this point, it would be interesting to explore the existence and role of possible counterpart of AtrR in X. dendrorhous. Importantly, the SREBP pathway plays a role in carotenogenesis through the regulation of carotenogenic genes crtE and crtR (Fig. 1) [17]. In addition, small ChIP-exo peaks were observed in the promoter region of the astaxanthin synthase encoding gene (crtS gene), although no SRE elements were detected. However, when comparing transcript levels among strains having an activated SREBP pathway and sre1 mutants by RNA-seq analysis, the transcriptional profile of crtS was similar to that of crtR (higher transcript levels in strains having an active SREBP pathway and lower levels in sre1 mutants), suggesting that crtS might be regulated by Sre1 at some level [17]. CrtR is the only cytochrome P450s reductase in this yeast, and it participates in both: carotenoid (assisting the cytochrome P450 enzyme astaxanthin synthase, CrtS) and ergosterol (assisting the cytochrome P450 enzymes Cyp51 and Cyp61, and probably squalene epoxidase) biosynthesis (Fig. 1). However, CrtR is only essential for astaxanthin biosynthesis [64] as the crtR mutants of X. dendrorhous do not produce astaxanthin and accumulate β-carotene, but, although in a lower proportion when compared to the wild-type strain, they do produce ergosterol. This last observation supported that there is an alternative electron donor to the cytochrome P450 reductase enzyme (probably the NADH-dependent cytochrome b5 reductase and cytochrome b5 via) that in X. dendrorhous may assist the production of ergosterol, but not that of astaxanthin [65]. The regulation of crtR by Sre1 is proof of links between carotenoid and sterol production in X. dendrorhous.

On the other hand, carotenogenesis depends on metabolites from the MVA pathway. It is important to note that in non-carotenogenic organisms, the MVA pathway is usually considered as the first steps of the sterol biosynthesis pathway. Therefore, these genes are generally considered as genes of sterol biosynthesis. However, products of the MVA pathway are also precursors of other metabolites than sterols; for example, carotenoids in non-photosynthetic organisms as in X. dendrorhous. In X. dendrorhous, Sre1 directly regulates the transcription of three genes of the MVA pathway (Fig. 1). Overexpression of genes of the MVA pathway and of gene crtE in X. dendrorhous, favors carotenoid production [66, 67]; therefore, the regulation of genes of the MVA pathway and is another evidence of regulatory bridging between the production of carotenoids and sterols in this yeast.

Conclusions

The SREBP pathway in fungi is still not fully understood and current knowledge is based on studies on few fungal models. In ascomycetes, the proteolytic activation of the SREBP homolog depends on several components of the SREBP pathway, which include proteins that anchor SREBP to the ER membrane and mechanisms that include at least one protease involved in the processing of SREBP. In X. dendrorhous, like the basidiomycete C. neoformans, Sre1 cleavage depends on the mammalian S2P homolog, the Stp1 protease. However, after the first hydrolysis performed by Stp1, the possibility of a second cleavage of Sre1 by another protease is not ruled out. On the other hand, unlike C. neoformans, X. dendrorhous lacks a SCAP homolog, which is the sterol sensor protein that retains SREBP in the ER membrane. Thus, Sre1 processing in X. dendrorhous is probably SCAP-independent and depends on sterol composition changes that are still unknown. This mechanism is different from what is observed in mammals and in some fungi, where the transcription factor is activated in response to sterol depletion in a SCAP-dependent manner. These differences raise important questions in X. dendrorhous about how this yeast monitors sterol levels and regulates the stability and proteolytic activation of Sre1. On the other hand, it is likely that Sre1 in X. dendrorhous is also regulated by post-translational modifications, as reported in the yeast S. pombe; however, this has not yet been explored in the carotenogenic yeast. Another difference between members of the ascomycota and basidiomycota divisions is the presence of more than one SREBP homolog in ascomycetes. However, in some fungi, not all SREBP homologs have been shown to be involved in the regulation of lipid metabolism or in other functions of the SREBP pathway. Basidiomycetes X. dendrorhous and C. neoformans harbor only one Sre1 homolog, which is involved in lipid homeostasis, which in turn regulates other processes, for example, carotenogenesis in X. dendrorhous. The regulation of carotenoid biosynthesis in X. dendrorhous by the transcription factor Sre1 is thus a novel function of the SREBP pathway and is a regulatory bridge between the biosynthesis of sterols and carotenoids in this species. The recent data regarding the SREBP pathway in X. dendrorhous contributes to our understanding about the regulation of carotenogenesis in this yeast and to our knowledge of SREBP pathways in fungi.

Availability of data and materials

Not applicable.

Abbreviations

SREBP:

Sterol Regulatory Element-Binding Protein

MVA:

Mevalonate

bHLH-LZ:

Basic-helix-loop-helix leucine zipper

ER:

Endoplasmic reticulum

SCAP:

SREBP cleavage-activating protein

S1P:

Site-1 protease

S2P:

Site-2 protease

SREs:

Sterol regulatory elements

Insig:

Insulin-induced gene

Dsc:

Defective for SREBP cleavage

Sre1N:

N-terminal bHLH-LZ domain of Sre1

References

  1. Saini RK, Keum YS. Microbial platforms to produce commercially vital carotenoids at industrial scale: an updated review of critical issues. J Ind Microbiol Biotechnol. 2019;46(5):657–74.

    Article  CAS  PubMed  Google Scholar 

  2. Gervasi T, Pellizzeri V, Benameur Q, Gervasi C, Santini A, Cicero N, et al. Valorization of raw materials from agricultural industry for astaxanthin and beta-carotene production by Xanthophyllomyces dendrorhous. Nat Prod Res. 2018;32(13):1554–61.

    Article  CAS  PubMed  Google Scholar 

  3. Gervasi T, Santini A, Daliu P, Salem A, Gervasi C, Pellizzeri V, et al. Astaxanthin production by Xanthophyllomyces dendrorhous growing on a low cost substrate. Agrofor Syst. 2020;94(4):1229–34.

    Article  Google Scholar 

  4. Naguib YM. Antioxidant activities of astaxanthin and related carotenoids. J Agric Food Chem. 2000;48(4):1150–4.

    Article  CAS  PubMed  Google Scholar 

  5. Sztretye M, Dienes B, Gonczi M, Czirjak T, Csernoch L, Dux L, et al. Astaxanthin: a potential mitochondrial-targeted antioxidant treatment in diseases and with aging. Oxid Med Cell Longev. 2019;2019:3849692.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Lim KC, Yusoff FM, Shariff M, Kamarudin MS. Astaxanthin as feed supplement in aquatic animals. Rev Aquacult. 2018;10(3):738–73.

    Article  Google Scholar 

  7. Lee PC, Schmidt-Dannert C. Metabolic engineering towards biotechnological production of carotenoids in microorganisms. Appl Microbiol Biotechnol. 2002;60(1–2):1–11.

    CAS  PubMed  Google Scholar 

  8. Visser H, van Ooyen AJ, Verdoes JC. Metabolic engineering of the astaxanthin-biosynthetic pathway of Xanthophyllomyces dendrorhous. FEMS Yeast Res. 2003;4(3):221–31.

    Article  CAS  PubMed  Google Scholar 

  9. Schmidt I, Schewe H, Gassel S, Jin C, Buckingham J, Humbelin M, et al. Biotechnological production of astaxanthin with Phaffia rhodozyma/Xanthophyllomyces dendrorhous. Appl Microbiol Biotechnol. 2011;89(3):555–71.

    Article  CAS  PubMed  Google Scholar 

  10. Barredo JL, García-Estrada C, Kosalkova K, Barreiro C. Biosynthesis of astaxanthin as a main carotenoid in the heterobasidiomycetous yeast Xanthophyllomyces dendrorhous. J Fungi (Basel). 2017;3(3):44.

    Article  CAS  Google Scholar 

  11. Dufossé L. Current carotenoid production using microorganisms. In: Singh OV, editor. Bio-pigmentation and biotechnological implementations. 1st ed. USA: Wiley; 2017. p. 87–106.

    Chapter  Google Scholar 

  12. Schroeder WA, Calo P, DeClercq ML, Johnson EA. Selection for carotenogenesis in the yeast Phaffia rhodozyma by dark-generated singlet oxygen. Microbiology. 1996;142(10):2923–9.

    Article  CAS  Google Scholar 

  13. Schroeder WA, Johnson EA. Carotenoids protect Phaffia rhodozyma against singlet oxygen damage. J Ind Microbiol. 1995;14(6):502–7.

    Article  CAS  Google Scholar 

  14. Schroeder WA, Johnson EA. Singlet oxygen and peroxyl radicals regulate carotenoid biosynthesis in Phaffia rhodozyma. J Biol Chem. 1995;270(31):18374–9.

    Article  CAS  PubMed  Google Scholar 

  15. Córdova P, Alcaíno J, Bravo N, Barahona S, Sepúlveda D, Fernández-Lobato M, et al. Regulation of carotenogenesis in the red yeast Xanthophyllomyces dendrorhous: the role of the transcriptional co-repressor complex Cyc8-Tup1 involved in catabolic repression. Microb Cell Fact. 2016;15(1):193.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Alcaíno J, Bravo N, Córdova P, Marcoleta AE, Contreras G, Barahona S, et al. The involvement of Mig1 from Xanthophyllomyces dendrorhous in catabolic repression: an active mechanism contributing to the regulation of carotenoid production. PLoS One. 2016;11(9): e0162838.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Gómez M, Campusano S, Gutiérrez MS, Sepúlveda D, Barahona S, Baeza M, et al. Sterol regulatory element-binding protein Sre1 regulates carotenogenesis in the red yeast Xanthophyllomyces dendrorhous. J Lipid Res. 2020;61(12):1658–74.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Eberle D, Hegarty B, Bossard P, Ferre P, Foufelle F. SREBP transcription factors: master regulators of lipid homeostasis. Biochimie. 2004;86(11):839–48.

    Article  CAS  PubMed  Google Scholar 

  19. Brown MS, Goldstein JL. The SREBP pathway: regulation of cholesterol metabolism by proteolysis of a membrane-bound transcription factor. Cell. 1997;89(3):331–40.

    Article  CAS  PubMed  Google Scholar 

  20. Tontonoz P, Kim JB, Graves RA, Spiegelman BM. ADD1: a novel helix-loop-helix transcription factor associated with adipocyte determination and differentiation. Mol Cell Biol. 1993;13(8):4753–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Yokoyama C, Wang X, Briggs MR, Admon A, Wu J, Hua X, et al. SREBP-1, a basic-helix-loop-helix-leucine zipper protein that controls transcription of the low density lipoprotein receptor gene. Cell. 1993;75(1):187–97.

    Article  CAS  PubMed  Google Scholar 

  22. Hua X, Yokoyama C, Wu J, Briggs MR, Brown MS, Goldstein JL, et al. SREBP-2, a second basic-helix-loop-helix-leucine zipper protein that stimulates transcription by binding to a sterol regulatory element. Proc Natl Acad Sci USA. 1993;90(24):11603–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hua X, Sakai J, Ho YK, Goldstein JL, Brown MS. Hairpin orientation of sterol regulatory element-binding protein-2 in cell membranes as determined by protease protection. J Biol Chem. 1995;270(49):29422–7.

    Article  CAS  PubMed  Google Scholar 

  24. Nohturfft A, Brown MS, Goldstein JL. Topology of SREBP cleavage-activating protein, a polytopic membrane protein with a sterol-sensing domain. J Biol Chem. 1998;273(27):17243–50.

    Article  CAS  PubMed  Google Scholar 

  25. Sakai J, Nohturfft A, Cheng D, Ho YK, Brown MS, Goldstein JL. Identification of complexes between the COOH-terminal domains of sterol regulatory element-binding proteins (SREBPs) and SREBP cleavage-activating protein. J Biol Chem. 1997;272(32):20213–21.

    Article  CAS  PubMed  Google Scholar 

  26. Yang T, Espenshade PJ, Wright ME, Yabe D, Gong Y, Aebersold R, et al. Crucial step in cholesterol homeostasis: sterols promote binding of SCAP to INSIG-1, a membrane protein that facilitates retention of SREBPs in ER. Cell. 2002;110(4):489–500.

    Article  CAS  PubMed  Google Scholar 

  27. Yabe D, Brown MS, Goldstein JL. Insig-2, a second endoplasmic reticulum protein that binds SCAP and blocks export of sterol regulatory element-binding proteins. Proc Natl Acad Sci USA. 2002;99(20):12753–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Sun LP, Li L, Goldstein JL, Brown MS. Insig required for sterol-mediated inhibition of Scap/SREBP binding to COPII proteins in vitro. J Biol Chem. 2005;280(28):26483–90.

    Article  CAS  PubMed  Google Scholar 

  29. Duncan EA, Brown MS, Goldstein JL, Sakai J. Cleavage site for sterol-regulated protease localized to a Leu–Ser bond in the lumenal loop of sterol regulatory element-binding protein-2. J Biol Chem. 1997;272(19):12778–85.

    Article  CAS  PubMed  Google Scholar 

  30. Duncan EA, Dave UP, Sakai J, Goldstein JL, Brown MS. Second-site cleavage in sterol regulatory element-binding protein occurs at transmembrane junction as determined by cysteine panning. J Biol Chem. 1998;273(28):17801–9.

    Article  CAS  PubMed  Google Scholar 

  31. Hughes AL, Todd BL, Espenshade PJ. SREBP pathway responds to sterols and functions as an oxygen sensor in fission yeast. Cell. 2005;120(6):831–42.

    Article  CAS  PubMed  Google Scholar 

  32. Willger SD, Puttikamonkul S, Kim KH, Burritt JB, Grahl N, Metzler LJ, et al. A sterol-regulatory element binding protein is required for cell polarity, hypoxia adaptation, azole drug resistance, and virulence in Aspergillus fumigatus. PLoS Pathog. 2008;4(11): e1000200.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Chang YC, Bien CM, Lee H, Espenshade PJ, Kwon-Chung KJ. Sre1p, a regulator of oxygen sensing and sterol homeostasis, is required for virulence in Cryptococcus neoformans. Mol Microbiol. 2007;64(3):614–29.

    Article  CAS  PubMed  Google Scholar 

  34. Chun CD, Liu OW, Madhani HD. A link between virulence and homeostatic responses to hypoxia during infection by the human fungal pathogen Cryptococcus neoformans. PLoS Pathog. 2007;3(2): e22.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Lamb DC, Baldwin BC, Kwon-Chung KJ, Kelly SL. Stereoselective interaction of the azole antifungal agent SCH39304 with the cytochrome P-450 monooxygenase system isolated from Cryptococcus neoformans. Antimicrob Agents Chemother. 1997;41(7):1465–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Qin L, Wu VW, Glass NL. Deciphering the regulatory network between the SREBP pathway and protein secretion in Neurospora crassa. mBio. 2017;8(2):e00233-17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Reilly MC, Qin L, Craig JP, Starr TL, Glass NL. Deletion of homologs of the SREBP pathway results in hyper-production of cellulases in Neurospora crassa and Trichoderma reesei. Biotechnol Biofuels. 2015;8:121.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Stewart EV, Lloyd SJ, Burg JS, Nwosu CC, Lintner RE, Daza R, et al. Yeast sterol regulatory element-binding protein (SREBP) cleavage requires Cdc48 and Dsc5, a ubiquitin regulatory X domain-containing subunit of the Golgi Dsc E3 ligase. J Biol Chem. 2012;287(1):672–81.

    Article  CAS  PubMed  Google Scholar 

  39. Stewart EV, Nwosu CC, Tong Z, Roguev A, Cummins TD, Kim DU, et al. Yeast SREBP cleavage activation requires the Golgi Dsc E3 ligase complex. Mol Cell. 2011;42(2):160–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kim J, Ha HJ, Kim S, Choi AR, Lee SJ, Hoe KL, et al. Identification of Rbd2 as a candidate protease for sterol regulatory element binding protein (SREBP) cleavage in fission yeast. Biochem Biophys Res Commun. 2015;468(4):606–10.

    Article  CAS  PubMed  Google Scholar 

  41. Hwang J, Ribbens D, Raychaudhuri S, Cairns L, Gu H, Frost A, et al. A Golgi rhomboid protease Rbd2 recruits Cdc48 to cleave yeast SREBP. EMBO J. 2016;35(21):2332–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bat-Ochir C, Kwak JY, Koh SK, Jeon MH, Chung D, Lee YW, et al. The signal peptide peptidase SppA is involved in sterol regulatory element-binding protein cleavage and hypoxia adaptation in Aspergillus nidulans. Mol Microbiol. 2016;100(4):635–55.

    Article  CAS  PubMed  Google Scholar 

  43. Willger SD, Cornish EJ, Chung D, Fleming BA, Lehmann MM, Puttikamonkul S, et al. Dsc orthologs are required for hypoxia adaptation, triazole drug responses, and fungal virulence in Aspergillus fumigatus. Eukaryot Cell. 2012;11(12):1557–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Dhingra S, Kowalski CH, Thammahong A, Beattie SR, Bultman KM, Cramer RA. RbdB, a rhomboid protease critical for SREBP activation and virulence in Aspergillus fumigatus. mSphere. 2016;1(2):e00035-16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chung D, Barker BM, Carey CC, Merriman B, Werner ER, Lechner BE, et al. ChIP-seq and in vivo transcriptome analyses of the Aspergillus fumigatus SREBP SrbA reveals a new regulator of the fungal hypoxia response and virulence. PLoS Pathog. 2014;10(11): e1004487.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Bien CM, Chang YC, Nes WD, Kwon-Chung KJ, Espenshade PJ. Cryptococcus neoformans Site-2 protease is required for virulence and survival in the presence of azole drugs. Mol Microbiol. 2009;74(3):672–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Gómez M, Gutiérrez MS, González AM, Gárate-Castro C, Sepúlveda D, Barahona S, et al. Metallopeptidase Stp1 activates the transcription factor Sre1 in the carotenogenic yeast Xanthophyllomyces dendrorhous. J Lipid Res. 2020;61(2):229–43.

    Article  PubMed  Google Scholar 

  48. Gutiérrez MS, Campusano S, González AM, Gómez M, Barahona S, Sepúlveda D, et al. Sterol regulatory element-binding protein (Sre1) promotes the synthesis of carotenoids and sterols in Xanthophyllomyces dendrorhous. Front Microbiol. 2019;10:586.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Loto I, Gutiérrez MS, Barahona S, Sepúlveda D, Martínez-Moya P, Baeza M, et al. Enhancement of carotenoid production by disrupting the C22-sterol desaturase gene (CYP61) in Xanthophyllomyces dendrorhous. BMC Microbiol. 2012;12:235.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Párraga A, Bellsolell L, Ferre-D’Amare AR, Burley SK. Co-crystal structure of sterol regulatory element binding protein 1a at 2.3 A resolution. Structure. 1998;6(5):661–72.

    Article  PubMed  Google Scholar 

  51. Maguire SL, Wang C, Holland LM, Brunel F, Neuveglise C, Nicaud JM, et al. Zinc finger transcription factors displaced SREBP proteins as the major sterol regulators during Saccharomycotina evolution. PLoS Genet. 2014;10(1): e1004076.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Radhakrishnan A, Ikeda Y, Kwon HJ, Brown MS, Goldstein JL. Sterol-regulated transport of SREBPs from endoplasmic reticulum to Golgi: oxysterols block transport by binding to Insig. Proc Natl Acad Sci USA. 2007;104(16):6511–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Sun LP, Seemann J, Goldstein JL, Brown MS. Sterol-regulated transport of SREBPs from endoplasmic reticulum to Golgi: Insig renders sorting signal in Scap inaccessible to COPII proteins. Proc Natl Acad Sci USA. 2007;104(16):6519–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Porter JR, Burg JS, Espenshade PJ, Iglesias PA. Ergosterol regulates sterol regulatory element binding protein (SREBP) cleavage in fission yeast. J Biol Chem. 2010;285(52):41051–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Venegas M, Barahona S, González AM, Sepúlveda D, Zúñiga GE, Baeza M, et al. Phenotypic analysis of mutants of ergosterol biosynthesis genes (ERG3 and ERG4) in the red yeast Xanthophyllomyces dendrorhous. Front Microbiol. 2020;11:1312.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Sundqvist A, Bengoechea-Alonso MT, Ye X, Lukiyanchuk V, Jin J, Harper JW, et al. Control of lipid metabolism by phosphorylation-dependent degradation of the SREBP family of transcription factors by SCF(Fbw7). Cell Metab. 2005;1(6):379–91.

    Article  CAS  PubMed  Google Scholar 

  57. Punga T, Bengoechea-Alonso MT, Ericsson J. Phosphorylation and ubiquitination of the transcription factor sterol regulatory element-binding protein-1 in response to DNA binding. J Biol Chem. 2006;281(35):25278–86.

    Article  CAS  PubMed  Google Scholar 

  58. Dong Q, Giorgianni F, Beranova-Giorgianni S, Deng X, O’Meally RN, Bridges D, et al. Glycogen synthase kinase-3-mediated phosphorylation of serine 73 targets sterol response element binding protein-1c (SREBP-1c) for proteasomal degradation. Biosci Rep. 2015;36(1): e00284.

    Article  PubMed  CAS  Google Scholar 

  59. Brookheart RT, Lee CY, Espenshade PJ. Casein kinase 1 regulates sterol regulatory element-binding protein (SREBP) to control sterol homeostasis. J Biol Chem. 2014;289(5):2725–35.

    Article  CAS  PubMed  Google Scholar 

  60. Hughes BT, Espenshade PJ. Oxygen-regulated degradation of fission yeast SREBP by Ofd1, a prolyl hydroxylase family member. EMBO J. 2008;27(10):1491–501.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Gárate-Castro C. Estudio y caracterización del gen OFD1 de la levadura carotenogénica Xanthophyllomyces dendrorhous. Santiago: Universidad de Chile; 2017. http://repositorio.uchile.cl/handle/2250/144145. Accessed 21 Oct 2021.

  62. Hagiwara D, Miura D, Shimizu K, Paul S, Ohba A, Gonoi T, et al. A Novel Zn2-Cys6 transcription factor AtrR plays a key role in an azole resistance mechanism of Aspergillus fumigatus by co-regulating cyp51A and cdr1B expressions. PLoS Pathog. 2017;13(1): e1006096.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Paul S, Stamnes M, Thomas GH, Liu H, Hagiwara D, Gomi K, et al. AtrR is an essential determinant of azole resistance in Aspergillus fumigatus. mBio. 2019;10(2):e02563-18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Alcaíno J, Barahona S, Carmona M, Lozano C, Marcoleta A, Niklitschek M, et al. Cloning of the cytochrome p450 reductase (crtR) gene and its involvement in the astaxanthin biosynthesis of Xanthophyllomyces dendrorhous. BMC Microbiol. 2008;8:169.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Gutiérrez MS, Rojas MC, Sepúlveda D, Baeza M, Cifuentes V, Alcaíno J. Molecular characterization and functional analysis of cytochrome b5 reductase (CBR) encoding genes from the carotenogenic yeast Xanthophyllomyces dendrorhous. PLoS One. 2015;10(10): e0140424.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Alcaíno J, Romero I, Niklitschek M, Sepúlveda D, Rojas MC, Baeza M, et al. Functional characterization of the Xanthophyllomyces dendrorhous farnesyl pyrophosphate synthase and geranylgeranyl pyrophosphate synthase encoding genes that are involved in the synthesis of isoprenoid precursors. PLoS One. 2014;9(5): e96626.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Hara KY, Morita T, Mochizuki M, Yamamoto K, Ogino C, Araki M, et al. Development of a multi-gene expression system in Xanthophyllomyces dendrorhous. Microb Cell Fact. 2014;13:175.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Niklitschek M, Alcaíno J, Barahona S, Sepúlveda D, Lozano C, Carmona M, et al. Genomic organization of the structural genes controlling the astaxanthin biosynthesis pathway of Xanthophyllomyces dendrorhous. Biol Res. 2008;41(1):93–108.

    Article  PubMed  Google Scholar 

  69. Verdoes JC, Krubasik KP, Sandmann G, van Ooyen AJ. Isolation and functional characterisation of a novel type of carotenoid biosynthetic gene from Xanthophyllomyces dendrorhous. Mol Gen Genet. 1999;262(3):453–61.

    Article  CAS  PubMed  Google Scholar 

  70. Verdoes JC, Misawa N, van Ooyen AJ. Cloning and characterization of the astaxanthin biosynthetic gene encoding phytoene desaturase of Xanthophyllomyces dendrorhous. Biotechnol Bioeng. 1999;63(6):750–5.

    Article  CAS  PubMed  Google Scholar 

  71. Córdova P, González AM, Nelson DR, Gutiérrez MS, Baeza M, Cifuentes V, et al. Characterization of the cytochrome P450 monooxygenase genes (P450ome) from the carotenogenic yeast Xanthophyllomyces dendrorhous. BMC Genomics. 2017;18(1):540.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Ojima K, Breitenbach J, Visser H, Setoguchi Y, Tabata K, Hoshino T, et al. Cloning of the astaxanthin synthase gene from Xanthophyllomyces dendrorhous (Phaffia rhodozyma) and its assignment as a beta-carotene 3-hydroxylase/4-ketolase. Mol Genet Genomics. 2006;275(2):148–58.

    Article  CAS  PubMed  Google Scholar 

  73. Álvarez V, Rodríguez-Saíz M, de la Fuente JL, Gudina EJ, Godio RP, Martín JF, et al. The crtS gene of Xanthophyllomyces dendrorhous encodes a novel cytochrome-P450 hydroxylase involved in the conversion of beta-carotene into astaxanthin and other xanthophylls. Fungal Genet Biol. 2006;43(4):261–72.

    Article  PubMed  CAS  Google Scholar 

  74. Leiva K, Werner N, Sepúlveda D, Barahona S, Baeza M, Cifuentes V, et al. Identification and functional characterization of the CYP51 gene from the yeast Xanthophyllomyces dendrorhous that is involved in ergosterol biosynthesis. BMC Microbiol. 2015;15:89.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Bien CM, Espenshade PJ. Sterol regulatory element binding proteins in fungi: hypoxic transcription factors linked to pathogenesis. Eukaryot Cell. 2010;9(3):352–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Vaknin Y, Hillmann F, Iannitti R, Ben Baruch N, Sandovsky-Losica H, Shadkchan Y, et al. Identification and characterization of a novel Aspergillus fumigatus rhomboid family putative protease, RbdA, involved in hypoxia sensing and virulence. Infect Immun. 2016;84(6):1866–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Colot HV, Park G, Turner GE, Ringelberg C, Crew CM, Litvinkova L, et al. A high-throughput gene knockout procedure for Neurospora reveals functions for multiple transcription factors. Proc Natl Acad Sci USA. 2006;103(27):10352–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Chang YC, Ingavale SS, Bien C, Espenshade P, Kwon-Chung KJ. Conservation of the sterol regulatory element-binding protein pathway and its pathobiological importance in Cryptococcus neoformans. Eukaryot Cell. 2009;8(11):1770–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

The work was supported by FONDECYT 1160202, ENL08/20 from Vicerrectoría de Investigación y Desarrollo (VID) de la Universidad de Chile, and by the graduate scholarship ANID-PFCHA/Doctorado Nacional/2017-21170613 to M. Gómez.

Author information

Authors and Affiliations

Authors

Contributions

MG, MB, VC and JA provided strategic inputs. MG and JA wrote the manuscript. All authors have read and approved the final manuscript.

Corresponding author

Correspondence to Jennifer Alcaíno.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gómez, M., Baeza, M., Cifuentes, V. et al. The SREBP (Sterol Regulatory Element-Binding Protein) pathway: a regulatory bridge between carotenogenesis and sterol biosynthesis in the carotenogenic yeast Xanthophyllomyces dendrorhous. Biol Res 54, 34 (2021). https://doi.org/10.1186/s40659-021-00359-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40659-021-00359-x

Keywords