key: cord-0950243-owr59495 authors: Paramasivan, Kalaivani; Mutturi, Sarma title: Recent advances in the microbial production of squalene date: 2022-04-15 journal: World J Microbiol Biotechnol DOI: 10.1007/s11274-022-03273-w sha: f303ce0e738ce7cf2484be70c580748f200da70a doc_id: 950243 cord_uid: owr59495 Squalene is a triterpene hydrocarbon, a biochemical precursor for all steroids in plants and animals. It is a principal component of human surface lipids, in particular of sebum. Squalene has several applications in the food, pharmaceutical, and medical sectors. It is essentially used as a dietary supplement, vaccine adjuvant, moisturizer, cardio-protective agent, anti-tumor agent and natural antioxidant. With the increased demand for squalene along with regulations on shark-derived squalene, there is a need to find alternatives for squalene production which are low-cost as well as sustainable. Microbial platforms are being considered as a potential option to meet such challenges. Considerable progress has been made using both wild-type and engineered microbial strains for improved productivity and yields of squalene. Native strains for squalene production are usually limited by low growth rates and lesser titers. Metabolic engineering, which is a rational strain engineering tool, has enabled the development of microbial strains such as Saccharomyces cerevisiae and Yarrowia lipolytica, to overproduce the squalene in high titers. This review focuses on key strain engineering strategies involving both in-silico and in-vitro techniques. Emphasis is made on gene manipulations for improved precursor pool, enzyme modifications, cofactor regeneration, up-regulation of limiting reactions, and downregulation of competing reactions during squalene production. Process strategies and challenges related to both upstream and downstream during mass cultivation are detailed. GRAPHICAL ABSTRACT: [Image: see text] Squalene is a naturally occurring linear triterpene hydrocarbon (C 30 H 50 ) comprising six double bonds (Fig. 1) . Squalene was named after the shark family "Squalidae" (Tsujimoto 1916) . The molecular weight of squalene is 410.7 g/mol, it has a melting point of − 75 °C, a boiling point of 285 °C, and a refractive index of 1.49 (Popa et al. 2015) . Squalene has several applications in the food, chemical, and healthcare sectors. It is used as a dietary supplement (Naziri et al. 2011a) , moisturizer, cardioprotective agent, anti-tumor agent, a natural antioxidant in addition to its antifungal (Smith 2000) , antibacterial, and antivirulence activities (Bavisetty et al. 2015) . It has been used as a vaccine adjuvant in seasonal influenza and COVID-19 vaccines (Tateno et al. 2020) . Squalene intake in the Mediterranean region and the United States is in the 200-400 mg/day and 30 mg/day ranges, respectively (Newmark 1999) . Squalane, a stable hydrogenated derivative of natural squalene is favored in the cosmetic industries as it penetrates human skin and imparts smoothness without greasy feels (Kim and Karadeniz 2012) . With the increased demand for squalene along with regulations on shark-derived squalene, there is a need to find alternatives for squalene production which are low-cost as well as sustainable (Paramasivan 2019) . In contrast to plant sterols Fig. 1 Squalene biosynthesis pathway in yeast (the MVA pathway) and bacteria (the MEP pathway). CDP-ME, 4-diphosphocytidyl-2-C-methylerythritol; CDP-MEP, CDP-2-C-methyl-D-erythritol 2-phosphate; DMAPP, dimethylallyl pyrophosphate; DXP, 1-Deoxy-D-xylulose 5-phosphate; dxr-1-deoxy-D-xylulose-5-phosphate reductoisomerase; dxs, 1-deoxyxylulose-5-phosphate synthase; ERG8, Phosphomevalonate kinase; ERG10, Acetyl-CoA Cacetyltransferase; ERG13, Hydroxy methyl glutaryl-CoA synthase; ERG12, Mevalonate kinase; ERG19, Mevalonate pyrophosphate decarboxylase; ERG9, Squalene synthase; ERG20, farnesyl pyrophosphate synthase; FPP, Farnesyl Pyrophosphate; GPP, Geranyl Pyrophosphate; HMB-PP, 4-Hydroxy-3-methyl-but-2-enyl pyrophosphate; IDI1, isopentenyl pyrophosphate: dimethylallyl pyrophosphate isomerase; IPP, Isopentenyl Pyrophosphate; ispD-2-C-methyl-D-erythritol-4-phosphate cytidylyl transferase; ispE, 4-diphophocy-tidyl-2-C-methyl-D-erythritol kinase; ispF, 2-C-methyl-D-erythritol 2,4-cyclodiphosphate synthase; ispG, HMB-PP synthase; ispH, HMB-PP reductase; HMGR, Hydroxy methyl glutaryl CoA reductase; MEP, 2-C-methylerythritol 4-phosphate; MEcPP, 2-C-methyl-D-erythritol-2,4-cyclodiphosphate such as sitosterol and stigmasterol, and animal sterols such as cholesterol, ergosterol acts as a predominant sterol in the cell wall of the fungus. Squalene is a ergosterol pathway intermediate in S. cerevisiae. It is produced in yeast through the mevalonate (MVA) biosynthesis process and in bacteria through the methylerythritol 4-phosphate (MEP) biosynthesis process (Fig. 1) . However, squalene, as an intermediary chemical, is produced at lower quantities in wild-type strains than other macromolecules and is usually below the detection level. Squalene is a biomolecule of economic importance, and efforts are undertaken in recent years to synthesize it utilizing S. cerevisiae as a cell factory (Table 1 ) (Mantzouridou and Tsimidou 2010; Naziri et al. 2011b; Rasool et al. 2016a; Han et al. 2018; Wei et al. 2018; Paramasivan et al. 2019 ). In the last decade, only a few patents on the generation of squalene in modified yeast have been granted (Walker et al. 2009; Choi et al. 2012) . Amyris, Inc., a biotech firm, has commercially synthesized squalene using a modified yeast strain (Fisher et al. 2013) . Biotechnological synthesis of squalene has also been pursued in recent years in various other species such as Escherichia coli, Synechocystis sp, Yarrowia lipolytica, and Kluyveromyces lactis other than S. cerevisiae (Table 2) (Bergquist et al. 2014; Drozdíková et al. 2015; Huang et al. 2018) . Squalene synthesis in microbes, plants, and mammals and their applications has been recently reviewed (Gohil et al. 2019 ). In the current review, metabolic engineering and process strategies for squalene enhancement in different microbes have been comprehensively discussed with emphasis on squalene synthesis in yeast. The discussed engineering strategies could also be effectively applied for the synthesis of different terpenes in yeast as well as in other microbes. In S. cerevisiae, 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG-CoA) reductase is translated as two different isomers, Hmg1p and Hmg2p. Hmg1p accounts for nearly 83 percent of overall enzyme activity in the wild-type strain (Basson et al. 1986 ). Further analysis revealed that these two variants have a membrane-binding region (1-552 amino acids) that covers the ER (endoplasmic reticulum) as well as the catalytic domain (553-1046 amino acids) ). HMG-CoA reductase is a major regulatory enzyme in the ergosterol biosynthesis pathway and its overexpression has proven essential in most metabolic engineering efforts of S. cerevisiae towards terpene synthesis (Rodwell et al. 1976 ). Later, it was revealed that upregulation of the catalytic domain of HMG1, known as tHMG1, enhanced squalene levels in S. cerevisiae (Donald et al. 1997; Polakowski et al. 1998 ). Overexpression of membrane-bound Hmg1p results in the development of ER membrane stacks, also known as karmellae (Wright et al. 1988 ). To make a soluble and nonmembrane bound, a truncated form of the HMG-CoA reductase enzyme (tHMG1) has been overexpressed (Polakowski et al. 1998 ). Moreover to avoid feed-back inihibition of ergosterol, it was expressed under a constitutive promoter in their studies. This theory established the basis for the upregulation of tHMG1 in an array of studies targeting the synthesis of terpenes in S. cerevisiae (Donald et al. 1997; Polakowski et al. 1998; Thompson et al. 2014; Zhuang and Chappell 2015; Kwak et al. 2017; Han et al. 2018; Paramasivan et al. 2019; Liu et al. 2020a, b; Zhu et al. 2021 ) ( Table 1) . Overexpression of tHMG1 encoding solely the catalytic domain of protein to circumvent regulation did not raise ergosterol content as predicted, but it did boost squalene production by nearly 30 times in S. cerevisiae (Thompson et al. 2014 ). Full-length HMG1 overexpression has also been attempted and has been shown to increase squalene levels despite the feedback regulation (Tokuhiro et al. 2009 ). It might be attributable to the multi-copy integration of this gene in their studies; however, the exact theory is unclear. Hmg2p, a major isoform of this reductase in low oxygen conditions, has therefore turned into a target for upregulation during anaerobic growth to synthesize squalene (Mantzouridou and Tsimidou 2010) . Upregulation of structural genes, particularly those in the sterol pathway ahead of squalene, have been identified as key targets for squalene production in S. cerevisiae (Fig. 1 ) (Rasool et al. 2016a; Li et al. 2020; Liu et al. 2020a, b; Zhou et al. 2021; Zhu et al. 2021; Meng et al. 2020; Sun et al. 2020) . The genes include ERG10 coding for acetyl-CoA C-acetyltransferase, ERG13 coding for HMG-CoA synthase, HMGR, ERG12 coding for mevalonate kinase, ERG8 coding for phosphomevalonate kinase, IDI1 coding for isopentenyl pyrophosphate: dimethylallyl pyrophosphate isomerase, ERG20 coding for farnesyl pyrophosphate synthase, and ERG9 coding for squalene synthase. In a study by Rasool et al. (2016a) , overexpression of the structural genes in the MVA pathway such as HMG1, IDI1, ERG20, and ERG9 has been attempted during the synthesis of squalene and has enhanced the squalene up to 10-folds (Table 1 ; Figs. 1, 2a) . Further, the presence of squalene monooxygenase inhibitor, terbinafine, together with overexpression of the MVA pathway genes has increased squalene titer to 304.49 mg/l (Rasool et al. 2016a ). Overexpression of the MVA pathway genes combined with the carbon source controlled fed-batch fermentation has resulted in a strain with squalene synthesis ability of 9.5 g/l (Table 1 ) (Li et al. 2020 ). Deletion of the competing reactions is an effective method for constructing an efficient microbial chassis (Pickens et al. 2014) (Fig. 2b) . This approach is referred to as either knockout which is typically applied by deleting a gene engaged in the competing reactions or knockdown which is reducing the levels of gene expression by introducing a weak promoter, and occasionally by allowing a system to regulate expression through the use of a programmable or customizable promoter (referred as repression). Through a random mutation in the ERG1 gene, Kamimura et al. (1994) achieved a squalene production of 5 mg/g DCW (dry cell weight) by disrupting the ergosterol biosynthesis pathway in S. cerevisiae. Garaiová et al. (2014) investigated squalene increase in the mutant strains of ERG1 grown in the medium supplemented with terbinafine, ergosterol biosynthesis inhibitor and obtained a squalene production of 1000 µg per 10 9 cells. Knockout of the ERG6 gene coding for delta (24)-sterol C-methyltransferase along with a site-specific mutation in HMG2 (K6R) has enhanced squalene to 20-fold (Mantzouridou and Tsimidou 2010) . Likewise, knockout of ERG6 gene along with knockdown of ERG11 gene involved in ergosterol biosynthesis has enhanced squalene up to 43 mg/g DCW in S. cerevisiae (Paramasivan et al. 2018) . In another study, knockdown of downstream genes involved in ergosterol synthesis and the alcohol dehydrogenase genes involved in ethanol biosynthesis combined with several other strategies has enhanced squalene to 304.49 mg/l (Rasool et al. 2016a) . Also, knockout of the SHC gene coding for squalene hopene cyclase in the Rhodopseudomonas palustris and the cyanobacterial Synechocystis sp has enhanced squalene up to 70-fold and 27-fold, respectively Xu et al. 2016 ) ( Table 2 ). In Yarrowia lipolytica, knockout of PEX10 encoding a peroxisomal membrane E3 ubiquitin ligase, and URE2 encoding a transcriptional regulator involved in the repression of nitrogen catabolite has enhanced squalene yield (Wei et al. 2021 ). Rox1p is a repressor transcriptional factor protein involved in the regulation of ergosterol biosynthesis (Henry et al. 2002) . ROX1 gene deletion has enhanced the mevalonate and terpenoid synthesis in Saccharomyces cerevisiae (Özaydın et al. 2013) . So far, this approach has not been demonstrated in yeast squalene synthesis. Site-specific mutagenesis is a critical enzyme engineering technique to improve target compounds in the cells (Fig. 2c) . Directed evolution improves enzyme characteristics such as stability, solubility, and specificity, resulting in the generation of customized enzymes (Kaur and Sharma 2006) . Site-directed mutagenesis aims to introduce deliberate mutations at a specific codon in the genes, resulting in a changed codon. This method has shown to be an invaluable tool in synthetic biology for improving protein characteristics, and it has been used to promote squalene synthesis in yeast and other microbes. As squalene epoxide synthesis reaction is catalyzed by the ERG1 gene, attempts were made to modify the ERG1 gene to reduce the flux towards the downstream ergosterol biosynthesis pathway. In a study, glutamine Q at position 443 was mutagenized to UAG in Erg1p that in turn increased squalene up to 30-40-fold in the engineered S. cerevisiae (Garaiová et al. 2014 ) (Table 1) . Several other point mutational studies have been made in the ERG1 gene. In a study by Zhou et al. (2021) , glycine G at position 30 was mutagenized to serine S in Erg1p to reduce the specific activity. The study resulted in a strain with squalene production ability up to 3.5 g/l (Table 1) . Sitespecific mutations have also been made in another critical gene, HMG2. Primarily, lysine K at positions 6, and 357 on Hmg2p were identified to be crucial for protein degradation, and so these lysines were substituted to evaluate the Hmgp2 stability (Gardner and Hampton 1999) . In a study by Mantzouridou and Tsimidou (2010) , lysine K at position 6 has been mutagenized to asparagine R in Hmg2p which resulted in a strain that produced 5.2 mg/g DCW of squalene (Table 1) . Hmg2P (K6R) has also been successful in improving several other terpenes Mutturi 2017a, 2017b) . During the synthesis of squalene, Garaiová et al. (2014) downregulated squalene epoxidase coded by ERG1 and expressed multiple enzyme mutant variants of the ERG1 gene. Based on reverse engineering approaches, a causal mutation for high levels of squalene accumulation in yeast cells has been identified in Paramasivan et al. (2021) when S. cerevisiae cells were adapted in the presence of terbinafine. In this study it was demonstrated that phenylalanine at position 420 of Erg1p was replaced with isoleucine (F420I), which resulted in improved squalene and terbinafine resistance. Under aerobic growth, Upc2p (uptake control protein) mutants are involved in sterol uptake (Vik Å and Rine 2001) . Mutations in Upc2p such as G888A has been demonstrated to enhance external sterol (ergosterol) uptake, downregulation of internal ergosterol formation, and thereby realigning the allocation of resources for improvement in specific terpene synthesis. So far, this strategy has not been applied towards squalene synthesis studies in yeast. This section describes the heterologous expression-based metabolic engineering approaches that have been developed to augment squalene synthesis in S. cerevisiae and other microbes (Fig. 2d) . Heterologous enzymes are sometimes more efficient in catalysis when compared to the native enzyme. In this regard, several attempts have been made to enhance the squalene synthesis in microbes by heterologous expression of non-native enzymes. In a study, as an alternative to ERG20, heterologous expression of IspA coding for farnesyl diphosphate synthase from E. coli was carried to obtain squalene levels of 1.24 g/l in S. cerevisiae (Choi et al. 2012 ) (Table 1 ). In another study by Zhuang and Chappell (2015) , squalene synthase from Botryococcus braunii was expressed in S. cerevisiae which led to squalene synthesis of nearly 270 mg/l. Expression of squalene synthase from humans has also become another predominant strategy towards squalene improvement in host cells such as E. coli. (Furubayashi et al. 2014; Katabami et al. 2015; Xu et al. 2019) . Human SQS has a high catalytic efficiency towards FPP (kcat/ Km = 0.51 μM −1 s −1 ). Similarly, squalene synthase from T. elongatus BP-1 has an even higher catalytic efficiency (kcat/Km = 1.8 μM −1 s −1 ) and has been expressed in E. coli for increased squalene synthesis (Katabami et al. 2015) . Also, the squalene synthase from S. cerevisiae (ScSQS) along with the entire MVA pathway genes has been expressed in E. coli towards squalene enhancement (Sun et al. 2020) . In another study by Ghimire et al. (2009) , hopD (farnesyl diphosphate synthase) and hopAB (squalene/phytoene synthases) genes from Streptomyces peucetius have been heterologously expressed to augment squalene synthesis in yeast cells leading to an increased titer of 11.8 mg/l in E. coli. A novel heterologous pathway of squalene biosynthesis from Zymomonas mobilis and Rhodopseudomonas palustris has been expressed and characterized in E. coli. The pathway consists of three genes, hpnD, hpnC, and hpnE expressing enzymes that convert FPP to PSPP (presqualene diphosphate), PSPP to HSQ (hydroxysqualene) and HSQ to squalene, respectively (Pan et al. 2015) . Since the biosynthesis of a metabolite includes multiple reactions within the cell, improvements in essential upstream molecules generated may affect enhancing downstream pathway components. Acetyl-CoA is an example of an early intermediate metabolite that contributes to the TCA cycle, sterol, fatty acids, and polyketides biosynthesis. In S. cerevisiae, cytoplasmic acetyl-CoA is formed via a PDHbypass, which predominantly promotes sterol and fatty acid pathway (Shiba et al. 2007; Chen et al. 2013; Krivoruchko et al. 2015) . Pulling the flux towards squalene biosynthesis exhausts acetyl-CoA supply in S. cerevisiae cells impairing its functions. To overcome this, the ACS1 gene coding for acetyl-CoA synthetase has been overexpressed in a study targeting squalene improvement where ACS1 combined with the ERG10 gene expression has significantly enhanced squalene biosynthesis in yeast (Zhu et al. 2021) . Cytoplasmic acetyl-CoA augmentation is one of the primary strategies applied to improve squalene synthesis. Erg10p is the foremost enzyme in the MVA pathway that pulls the flux towards squalene synthesis by converting acetyl-CoA to acetoacetyl-CoA. ERG10 gene overexpression has been attempted in several studies that aim to improve squalene synthesis (Rasool et al. 2016b; Kwak et al. 2017; Li et al. 2020; Liu et al. 2020a, b; Zhou et al. 2021; Zhu et al. 2021) . Overexpression of tHMG1 along with the ERG10 gene in a xylose-fermenting strain has improved the squalene levels to 150 mg/l using xylose as a carbon source . Likewise, endogenous gene atoB coding for acetyl-CoA acetyltransferase has been overexpressed in an attempt to enhance squalene synthesis in E. coli (Katabami et al. 2015 ). Yet another promising strategy to improve acetyl-CoA is by conversion of ethanol to acetyl-CoA. In an interesting study by Li et al. (2020) , the ADH2 gene that converts ethanol to acetyl-CoA has been overexpressed together with the heterologous expression of ADA from Dickeya zeae which converts acetaldehyde to acetyl-CoA in S. cerevisiae. As the ADA gene coding enzyme does not require ATP consumption, the metabolic burden on the cells during squalene synthesis is reduced. In addition, ACL1 and ACL2 genes coding for ATP citrate lyase from Yarrowia lipolytica that converts citrate to acetyl-CoA have been overexpressed in S. cerevisiae towards squalene synthesis. This strategy has proven to enhance the squalene content in the cells significantly (Liu et al. 2020a, b) . In yeast cells, NADPH is one of the ubiquitous cofactors (Agledal et al. 2010) . Several enzymes in the sterol pathway of S. cerevisiae require NADPH as a cofactor. Overexpression of the MVA pathway genes such as tHMG1, HMG2, and ERG9 would deplete cytosolic NADPH. Hence, the expression of NADPH regeneration-related genes has become a popular approach to stimulate terpenes and other metabolite syntheses (Brown et al. 2015) (Fig. 2d) . In yeast cells, NADH kinases and NADP + -dependent dehydrogenases are responsible for NADPH synthesis, and regeneration respectively (Shi et al 2005 (Shi et al , 2011 . Precisely, NADH kinase is involved in the phosphorylation of NADH to NADPH while the dehydrogenases are involved in the reduction of NADP + to NADPH. POS5 gene codes for the enzyme that harbors both NAD kinase as well as NADH kinase activity (Strand et al. 2003) while another enzyme glucose-6-phosphate dehydrogenase coded by ZWF1 has NADP + -dependent dehydrogenase activity. In S. cerevisiae, ZWF1 is a prime target for promoting cytosolic NADPH regeneration. ZWF1 gene overexpression enhanced squalene synthesis to 2.3fold, whereas POS5 overexpression with mitochondrial targeting sequence (MTS) has enhanced squalene production to sixfold upon overexpression in yeast cells (Paramasivan & Mutturi, 2017a, b) . In the same study, ZWF1 and POS5 (with MTS) gene expression combined with tHMG1 expression has synergistically enhanced squalene to 22.5fold and 27.5-fold respectively. In another study, POS5 gene expression was combined with tHMG1 expression and knockout of ADK1 and LYS1 genes identified by insilico based genome-scale models towards squalene increase (Fig. 2e) . The genetic modifications combined with fedbatch cultivation have enhanced squalene levels to 1.9 g/l. Similarly, zwf expression in E. coli combined with several other strategies has enhanced squalene synthesis to 52 mg/l with net increment up to 21-folds (Xu et al. 2019) (Table 1 ). In addition, to reduce the metabolic burden caused by the NADPH cofactor, an alternative NADH-based enzyme has been used as a target towards squalene synthesis (Li et al. 2020; Liu et al. 2020a, b) . Since NADH is more readily available compared to NADPH, NADH-based HMGR from Silicibacter pomeroyi has been heterologously expressed in S. cerevisiae for squalene improvement. NADH-HMGR overexpression combined with several other strategies has resulted in a strain with squalene levels of 9.5 g/l (Li et al. 2020 ) and 11 g/l (Liu et al. 2020a, b) (Table 1) . Promoters are critical for the control of gene expression thereby regulating the metabolic pathways in S. cerevisiae. Promoter-based engineering strategies are gaining an advantage in recent times as they could significantly affect gene expression levels. ERG1 gene codes for squalene monooxygenase in the MVA pathway downstream of squalene in S. cerevisiae. Being an essential gene, disruption of ERG1 leads to a lethal phenotype and therefore repression of ERG1 gene by promoter replacement has become a prominent strategy in the development of cell factories towards squalene biosynthesis (Hull et al. 2014; Manzoor et al. 2020; Liu et al. 2020a, b; Zhu et al. 2021) . Also, the usage of repressible promoters provides better economic feasibility when compared to the enzyme inhibitors at an industrial level. Initially, doxycycline repressible tet07-CYC1 promoter has been utilized to control the ERG1 expression by substituting the native ERG1 promoter at the chromosome. In a study by Hull et al. (2014) , ERG1 gene under repressible tet07-CYC1 promoter system along with grass juice as a carbon source has enhanced squalene up to 18 mg/l. Later, two iron and copper repressible promoters P AFT1 and P CTR1 , respectively are characterized and used to repress the ERG1 gene in S. cerevisiae towards squalene bioproduction. ERG1 repression with metal ion repressible P CTR1 promoter has enhanced squalene up to twofold when compared to P AFT1 promoter (Manzoor et al. 2020 ). In the same study, TFBS TEF1 and TFBS HHF2 were engineered into different constitutive promoters thereby enhancing the strength of the yeast constitutive promoters. The engineered promoters such as P HHF2 , P IRA1 , P RHO1 , and P PET9 with TFBS TEF1 were utilized to drive the transcription of HMG1, IDI1, ERG20, and ERG9 genes respectively. This study resulted in a strain with squalene improvement to nearly 74-fold (Manzoor et al. 2020) . Further, P HXT1 is a glucose inducible promoter and causes repression when the glucose levels in the medium become low or zero. The native promoter of ERG1 when replaced with the HXT1 promoter combined with multiple other strategies has significantly enhanced squalene titer to 11 g/l (Liu et al. 2020a, b) . Similarly, the P HXT1 has been used to repress the ERG1 which in combination with the compartmentalization of genes in mitochondria leads to the highest production of squalene reported to date which is 21.1 g/l (Table 1; Zhu et al. 2021) . Another noteworthy and most commonly used repressible promoter is the methionine repressible promoter (P MET3 ) which has been extensively used to repress the ERG9 gene towards other terpene production in yeast (Westfall et al. 2012 ). However tunable P MET3 has not been used towards squalene synthesis so far. GAL80 negatively regulates gal regulon by binding to P GAL4 in the absence of galactose as well as in the presence of carbon sources such as glucose. Since galactose is an expensive carbon source compared to glucose, GAL80 gene knockout is critical to express the genes under the strong galactose inducible promoters P GAL1 and P GAL10 (Westfall et al. 2012 ). Thus, Li et al. (2020) have overexpressed the mevalonate pathway gene tHMG1 under the galactose inducible promoter in the GAL80 deletion strain where the gene activation happens in response to glucose as the negative regulator is not present. This leads to a 33-fold improvement in squalene synthesis. In an interesting investigation by Zhou et al. (2021) , the SRE (sterol response element) motif in ERG1 and ERG11 genes was replaced with the heterologous marO motif to downregulate the expression of both the genes thereby improving squalene and lanosterol accumulation, respectively. Replacement of the SRE motif reduces the feedback regulation by SREBP (sterol response element-binding proteins) at high sterol levels in the cells while marO insertion helps in controlling the metabolic flux. marO is a cis-element of mar operator bound by the transcriptional repressor protein, marR in E. coli. marO engineered near the TATA box of ERG1 and ERG11 promoter could efficiently repress the respective gene expression proving to be a promising alternative for gene repression studies in yeast S. cerevisiae. Owing to the abundant availability of precursors and cofactors such as acetyl-CoA and redox equivalents, respectively, harnessing organelles such as mitochondria, ER, and peroxisomes are considered as a viable strategy towards squalene production (Fig. 2f) . In a study by Zhu et al. (2021) , to surpass the cell toxicity and the metabolic strain imposed by the phosphorylated intermediates of the MVA pathway, a partial pathway from acetyl-CoA to MVA has been incorporated into the mitochondria by merging the mitochondrial targeting signal sequence with overexpression genes. Upon combining the cytoplasmic and mitochondrial engineering with a two-stage fed-batch fermentation, the squalene titers have synergistically improved to 21.1 g/l with 437.1 mg/g DCW (Table 1) . Similarly, introducing the MVA pathway in the peroxisomes by fusing the genes with peroxisomal targeting sequence has enhanced the squalene titers up to 11 g/l upon combining with P HXT1 -ERG1 and a two-stage fermentation (Table 1 ) (Liu et al. 2020a, b) . Another noteworthy ER-based metabolic engineering has enhanced squalene levels up to 71-fold where the overexpression of the INO2 gene has expanded the ER functions. The key function of ER is protein folding and overexpression of genes causes metabolic burden in the ER. Therefore, the size regulatory factor gene INO2 has been overexpressed in the yeast to restore the normal ER functions (Kim et al. 2019) . Hence, organelle engineering could be considered as a novel and effective alternative to improve squalene production. Squalene is stored in the cell membrane, peroxisomes, and lipid droplets in yeast cells. A lipid droplet found in all eukaryotic cells is a lipid storage organelle that is made of sterol esters, triacylglycerols, and squalene. It consists of neutral lipids and serves as a depot for non-polar storage lipids such as squalene and fatty acids. Neutral lipid production and storage is a complex and dynamic process. Lipid droplets are ER-derived storage organelles whose quantity, size, and content vary depending on nutritional requirements and growth circumstances (Farese and Walther 2009; Fei et al. 2009 ). Triacylglycerides and steryl esters are inert and are found in the lipid bodies unless they are hydrolyzed while squalene is either sequestered in lipid particles or involved in membrane biogenesis (Spanova et al. 2010; Grillitsch et al. 2011) . In a study by Wei et al. (2018) , the DGA1 gene coding for the diacylglycerol acyltransferase gene involved in fatty acid metabolism has been overexpressed in S. cerevisiae which enhanced squalene up to 250-fold upon the combination with the tHMG1 gene. DGA1 gene expression has proven to enhance lipid body formation in yeast cells thereby increasing the storage ability of the cells. Cell membrane engineering has been carried out to enhance the storage of squalene in E. coli. In this regard, tsr gene coding for a serine chemoreceptor protein has been overexpressed which in turn enhanced the fatty acid composition in membrane lipid composition leading to the formation of a multi-layered structure. The engineered strain has synthesized squalene up to 612 mg/l which is the maximum known squalene synthesis to date in E. coli (Meng et al. 2020 ) ( Table 2 ). Bioprocess conditions play a key role in the high-level production of compounds in microbial fermentation (Fig. 2g) . Under anaerobic fermentative conditions, Bhattacharjee et al. (2001) have generated squalene in a commercial yeast strain with a yield of up to 41 µg/g DCW. Besides, under optimized semi-anaerobic cultivation, inoculation levels, and growth conditions, Mantzouridou et al. (2009) reached a maximum squalene yield of 3.1 mg/l. Subsequently, overexpression of tHMG1 and DGA1 and growth under nitrogenlimited conditions have led to a squalene titer of 445.6 mg/l in galactose-based fed-batch cultivation (Wei et al. 2018 ). In another study by Han et al. (2018) , tHMG1 and bacterial ispA gene in tandem with terbinafine-based inhibition of squalene epoxidase have enhanced the squalene synthesis up to 2 g/l in glucose-based fed-batch cultivation. In some cases, the carbon sources to supplement the growth have been varied. For instance, grass juice and cane molasses have been used as a feedstock to increase squalene synthesis to 18 mg/l and 3.5 g/l respectively (Hull et al. 2014; Zhou et al. 2021) . In an interesting study by Kwak et al (2017) , xylose utilizing engineered strain was subjected to growth under xylose-based fed-batch cultivation. This study resulted in a strain with 532 mg/l of squalene titer. On the other hand, open loop-based exponential feeding of glucose in fed-batch cultivation has resulted in 1.9 g/l of squalene production using an engineered S. cerevisiae (Paramasivan et al. 2019 ). An optimized two-stage fed-batch cultivation using glucose and ethanol as a carbon source has been used to achieve a squalene titer of 11 g/l in an engineered strain (Liu et al. 2020a, b) . C/N ratio, pH, and temperature optimization have led to a significantly high squalene production of 2.4 g/l in the yeast-like fungus Pseudozyma sp. SD301, a native producer of squalene (Song et al. 2015) . Adaptive evolution is the use of selection pressure to leverage the innate capacity of an organism to undergo mutations toward the desired phenotype (Conrad et al. 2014) (Fig. 2h ). It entails a process of continuous evolution with a selective gain for the variant exhibiting the desired trait (Sonderegger and Sauer 2003; Çakar et al. 2005) . Adaptive laboratory evolution in combination with sequencing is a critical strategy in metabolic engineering for biomolecule synthesis (Conrad et al. 2014) . Lowering the activity of the downstream enzyme, squalene epoxidase is a key technique for enhancing squalene levels and may be accomplished using a specialized non-competitive enzymatic inhibitor, terbinafine (Ta et al. 2012; Garaiová et al. 2014; Rasool et al. 2016a ). Terbinafine has been extensively used in various studies and in different microbes to increase the squalene accumulation Naziri et al. 2011a, b; Drozdıkova et al. 2015; Han et al. 2018) . Naziri et al. (2011a, b) pioneered the cultivation of S. cerevisiae in terbinafine which led to at least ten times improvement in squalene. Following that, Garaiová et al. (2014) have produced 1000 µg of squalene per 10 9 cells in the presence of terbinafine with an engineered strain with Erg1p (Q443UAG). An engineered yeast strain has been subjected to grow in the presence of terbinafine as a selection pressure to enhance the squalene compound. This study resulted in a strain that produced 198 mg/l which was a 16.5-fold enhancement in comparison to the parent strain (Paramasivan et al. 2021) . Further, terbinafine addition to the medium for the native squalene producer Aurantiochytrium mangrovei has increased squalene up to 40% . Besides, terbinafine has also been used to enhance squalene content in Kluyveromyces lactis (Drozdıkova et al. 2015) . Han et al. (2018) have cultivated an engineered yeast strain in a fed-batch fermentation medium in the presence of terbinafine which has improved squalene synthesis up to twofold. However, large-scale cultivation of S. cerevisiae in the presence of terbinafine towards squalene synthesis could be an expensive strategy. Hence some studies reported the site-specific mutagenesis of squalene epoxidase and replacement of the promoter for ERG1 gene to downregulate the activity of this enzyme (Garaiová et al. 2014; Hull et al. 2014) . Such strategies resulted in improved squalene accumulation as discussed earlier. The genes involved in the MEP pathway are upregulated in combination with the heterologous expression of squalene synthase from Bacillus megaterium has enhanced the squalene synthesis up to 29-fold in Bacillus subtilis (Song et al. 2020 ) ( Table 2 ). Overexpression of dxs and idi genes, repression of idsA gene, and heterologous expression of ERG9 from S. cerevisiae has led to improved squalene levels (105 mg/l) in Corynebacterium glutamicum (Park et al. 2018 ) (Table 2) . Further, when protein engineering by expression of human squalene synthase was coupled with pathway engineering, fed-batch cultivation, and dodecane extraction, the C. glutamicum strain generated 1.5 g/l (Park et al. 2021) (Table 2 ). Squalene synthase from S. cerevisiae along with the overexpression of the MVA pathway genes and tsr gene has increased squalene in the engineered E. coli (Meng et al. 2020 ) ( Table 2) . Cyanobacterial species such as Synechococcus elongatus has been utilized for squalene synthesis by heterologous expression of dxs, idi, ispA from E. coli, and overexpression of the fusion protein CpcB1-ScSQS along with light optimization in photobioreactor which has increased the squalene content up to 79.2 mg/g DCW (Choi et al. 2017 ) ( Table 2) . Metabolic engineering of a novel species Rhodopseudomonas palustris has enhanced squalene up to 23.3 mg/g DCW with 178-fold net increment by the knockout of shc and crtB genes as well as the overexpression of dxs, crtE, and hpnD (Xu et al. 2021 ) ( Table 2 ). Overexpression of ylHMG1 and ylDGA1 along with fermentation optimization enhanced squalene to 731 mg/l in Yarrowia lipolytica. This is the highest reported so far in that species (Tang et al. 2021 ) ( Table 2 ). In a study by Kang et al (2021) , overexpression of squalene synthase, inhibition of squalene-hopene cyclase by ferulenol, knockout of pds-crtN-ald gene cluster, and fed-batch cultivation with potassium nitrate supplementation has improved squalene levels to 31.3 mg/l in Methylomonas sp. DH-1 ) ( Table 2 ). Concerns about squalene production from shark liver oil, and lower yields from plant sources, microbial platforms for squalene production have been sought. However, there exists challenges with microbial production systems as well. Some of the significant limitations of existing microbial squalene manufacturing platforms is lower squalene TRY (titer, productivity and yields) meant for industrial production. Moreover, obtaining toxin-free squalene from the microbes that naturally produce squalene such as Schizochytrium sp. and Aurantiochytrium sp. is challenging. Besides, physiology, genetics, and process constraints needs to be well studied for such organisms. Some of the prominent microbial platforms for squalene production includes S. cerevisiae, E. coli, C. glutamicum and Y. lipolytica. Metabolic engineering strategies in S. cerevisiae such as co-factor engineering, improved precursor supply, alterations outside sterol pathway, and process strategies have been thoroughly investigated and highest squalene titer of 21.1 g/l has been reported so far. However, majority of studies in S. cerevisiae focused on episomal expression, which presents a process issue when grown in a bioreactor. Amyris Inc. commercial synthesis of squalene is implemented in two stages, (i) farnesene is synthesized in the first stage using modified S. cerevisiae, and (ii) the obtained farnesene is transformed to squalene in the second stage utilizing chemical catalysis. Such hybrid production platforms can be exploited for commercial viability. Several microbial strains with high squalene productivity are developed in recent times which could serve as potential chassis for commercial production of squalene and also to compete with other low cost non-sustainable alternatives such as shark liver oil and vegetable oils. The present review on strain engineering and bioprocess strategies might be highly useful in improving squalene synthesis, leading to the development of a feasible and robust microbial platform for industrial production squalene. The phosphate makes a difference: cellular functions of NADP Saccharomyces cerevisiae contains two functional genes encoding 3-hydroxy-3-methylglutaryl-coenzyme A reductase Structural and functional conservation between yeast and human 3-hydroxy-3-methylglutaryl coenzyme A reductases, the rate-limiting enzyme of sterol biosynthesis Inhibition of virulence of Staphylococcus aureus -a food borne pathogen -by squalene, a functional lipid Production of squalene in Synechocystis sp. PCC 6803 Studies on fermentative production of squalene De novo production of the plant-derived alkaloid strictosidine in yeast Evolutionary engineering of multiple-stress resistant Saccharomyces cerevisiae The isolation and characterization of Pseudozyma sp Optimization of nitrogen source for enhanced production of squalene from thraustochytrid Aurantiochytrium sp Establishing a platform cell factory through engineering of yeast acetyl-CoA metabolism Improvement of squalene production from CO in Synechococcus elongatus PCC 7942 by metabolic engineering and scalable production in a photobioreactor Modified yeast strain and a method for producing squalene using the same Scalable cultivation of engineered cyanobacteria for squalene production from industrial flue gas in a closed photobioreactor Microbial laboratory evolution in the era of genome-scale science Effects of overproduction of the catalytic domain of 3-hydroxy-3-methylglutaryl coenzyme A reductase on squalene synthesis in Saccharomyces cerevisiae Production of squalene by lactose-fermenting yeast Kluyveromyces lactis with reduced squalene epoxidase activity Physiologic and metabolic characterization of Saccharomyces cerevisiae reveals limitations in the synthesis of the triterpene squalene Production of squalene in Synechocystis sp. PCC 6803 Towards a sustainable route for the production of squalene using cyanobacteria Enhanced production of squalene in the thraustochytrid Aurantiochytrium mangrovei by medium optimization and treatment with terbinafine Lipid droplets finally get a little Conditions of endoplasmic reticulum stress stimulate lipid droplet formation in Saccharomyces cerevisiae Squalane and isosqualane compositions and methods for preparing the same Construction of carotenoid biosynthetic pathways using squalene synthase Squalene epoxidase as a target for manipulation of squalene levels in the yeast Saccharomyces cerevisiae A highly conserved signal controls degradation of 3-hydroxy-3-methylglutaryl-coenzyme a (HMG-CoA) reductase in eukaryotes Improved squalene production via modulation of the methylerythritol 4-phosphate pathway and heterologous expression of genes from Streptomyces peucetius ATCC 27952 in Escherichia coli Engineering strategies in microorganisms for the enhanced production of squalene: advances, challenges and opportunities Lipid particles/ droplets of the yeast Saccharomyces cerevisiae revisited: lipidome meets proteome High level recombinant production of squalene using selected Saccharomyces cerevisiae strains ROX1 and ERG regulation in Saccharomyces cerevisiae: implications for antifungal susceptibility Extraction of squalene as value-added product from the residual biomass of Schizochytrium mangrovei PQ6 during biodiesel producing process Enhanced squalene biosynthesis in Yarrowia lipolytica based on metabolically engineered acetyl-CoA metabolism Co-production of ethanol and squalene using a Saccharomyces cerevisiae ERG1 (squalene epoxidase) mutant and agro-industrial feedstock Accumulation of squalene in a microalga Chlamydomonas reinhardtii by genetic modification of squalene synthase and squalene epoxidase genes Construction of squalene-accumulating Saccharomyces cerevisiae mutants by gene disruption through homologous recombination High-level squalene production from methane using a metabolically engineered Methylomonas sp. DH-1 strain Production of squalene by squalene synthases and their truncated mutants in Escherichia coli Directed evolution: an approach to engineer enzymes Thraustochytrid Aurantiochytrium sp. 18W-13a accummulates high amounts of squalene Biological importance and applications of squalene and squalane Tailoring the Saccharomyces cerevisiae endoplasmic reticulum for functional assembly of terpene synthesis pathway Microbial acetyl-CoA metabolism and metabolic engineering Enhanced isoprenoid production from xylose by engineered Saccharomyces cerevisiae Metabolic engineering of Saccharomyces cerevisiae to overproduce squalene The yeast peroxisome: a dynamic storage depot and subcellular factory for squalene overproduction Genetic and bioprocess engineering to improve squalene production in Yarrowia lipolytica Observations on squalene accumulation in Saccharomyces cerevisiae due to the manipulation of HMG2 and ERG6 Squalene versus ergosterol formation using Saccharomyces cerevisiae: combined effect of oxygen supply, inoculum size, and fermentation time on yield and selectivity of the bioprocess Self-redirection of metabolic flux toward squalene and ethanol pathways by engineered yeast Extension of cell membrane boosting squalene production in the engineered Escherichia coli Optimization of culture conditions of the thraustochytrid Aurantiochytrium sp. strain 18W-13a for squalene production TLC screening of thraustochytrid strains for squalene production Squalene resources and uses point to the potential of biotechnology Enhanced squalene production by wild-type Saccharomyces cerevisiae strains using safe chemical means Squalene, olive oil, and cancer risk review and hypothesis Carotenoid-based phenotypic screen of the yeast deletion collection reveals new genes with roles in isoprenoid production Biosynthesis of squalene from farnesyl diphosphate in Bacteria: three steps catalyzed by three enzymes Metabolic engineering of Saccharomyces cerevisiae for overproduction of squalene Progress in terpene synthesis strategies through engineering of Saccharomyces cerevisiae Regeneration of NADPH coupled with HMG-CoA reductase activity increases squalene synthesis in Saccharomyces cerevisiae Studies on squalene biosynthesis and the standardization of its extraction methodology from Saccharomyces cerevisiae Systems-based Saccharomyces cerevisiae strain design for improved squalene synthesis Adaptive evolution of engineered yeast for squalene production improvement and its genome-wide analysis Heterologous production of squalene from glucose in engineered Corynebacterium glutamicum using multiplex CRISPR interference and high-throughput fermentation Microbial bioprocess for extracellular squalene production and formulation of nanoemulsions Simultaneous production of DHA and squalene from Aurantiochytrium sp. grown on forest biomass hydrolysates Metabolic engineering for the production of natural products Overexpression of a cytosolic hydroxymethylglutaryl-CoA reductase leads to squalene accumulation in yeast Methods for obtaining and determination of squalene from natural sources Green microalgae strain improvement for the production of sterols and squalene Overproduction of squalene synergistically downregulates ethanol production in Saccharomyces cerevisiae Engineering of the terpenoid pathway in Saccharomyces cerevisiae co-overproduces squalene and the non-terpenoid compound oleic acid Regulation of HMG-CoA reductase Screening and optimization of squalene production from microalgae Aurantiochytrium sp Rhodosporidium sp. DR37: a novel strain for production of squalene in optimized cultivation conditions Identification of ATP-NADH kinase isozymes and their contribution to supply of NADP(H) in Saccharomyces cerevisiae Role of mitochondrial NADH kinase and NADPH supply in the respiratory chain activity of Saccharomyces cerevisiae Engineering of the pyruvate dehydrogenase bypass in Saccharomyces cerevisiae for highlevel production of isoprenoids Squalene: potential chemo preventive agent Evolutionary engineering of Saccharomyces cerevisiae for anaerobic growth on xylose High production of squalene using a newly isolated yeast-like strain Production of squalene in Bacillus subtilis by squalene synthase screening and metabolic engineering Effect of lipid particle biogenesis on the subcellular distribution of squalene in the yeast Saccharomyces cerevisiae POS5 gene of Saccharomyces cerevisiae encodes a mitochondrial NADH kinase required for stability of mitochondrial DNA Using engineered Escherichia coli to synthesize squalene with optimized manipulation of squalene synthase and mevalonate pathway Accumulation of squalene is associated with the clustering of lipid droplets Metabolic engineering of Yarrowia lipolytica for improving squalene production Synthetic biologyderived triterpenes as efficacious immunomodulating adjuvants Overproduction of geranylgeraniol by metabolically engineered Saccharomyces cerevisiae A highly unsaturated hydrocarbon in shark liver oil Upc2p and Ecm22p, dual regulators of sterol biosynthesis in Saccharomyces cerevisiae Production of squalene using yeast U.S. Patent Application No Improved squalene production through increasing lipid contents in Saccharomyces cerevisiae Increased accumulation of squalene in engineered Yarrowia lipolytica through deletion of PEX10 and URE2 From the Cover: PNAS Plus: production of amorphadiene in yeast, and its conversion to dihydroartemisinic acid, precursor to the antimalarial agent artemisinin Increased amounts of HMG-CoA reductase induce "karmellae": a proliferation of stacked membrane pairs surrounding the yeast nucleus Metabolic engineering of Rhodopseudomonas palustris for squalene production Improving squalene production by enhancing the NADPH/NADP+ ratio, modifying the isoprenoid-feeding module and blocking the menaquinone pathway in Escherichia coli Improving squalene production by blocking the competitive branched pathways and expressing ratelimiting enzymes in Rhodopseudomonas palustris Impact of methyl jasmonate on squalene biosynthesis in microalga Schizochytrium mangrovei Bio-based squalene production by Aurantiochytrium sp. through optimization of culture conditions, and elucidation of the putative biosynthetic pathway genes Engineering of cis-element in Saccharomyces cerevisiae for efficient accumulation of value-added compound squalene via downregulation of the downstream metabolic flux Metabolic compartmentalization in yeast mitochondria: burden and solution for squalene overproduction Building terpene production platforms in yeast Publisher's Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations Author contributions KP and SM wrote the manuscript. All authors read and approved the manuscript.