Recombinant Ustilago maydis squalene synthase, also known as ERG9, is an enzyme that plays a crucial role in the biosynthesis of sterols in fungi. It is the first committed enzyme in the sterol biosynthesis pathway, converting farnesyl pyrophosphate (FPP) into squalene. This enzyme is essential for the production of ergosterol, a critical component of fungal cell membranes. The recombinant version of this enzyme in Ustilago maydis, a model fungus, offers insights into metabolic engineering and biotechnological applications.
Squalene synthase is pivotal in the biosynthesis of sterols, as it catalyzes the conversion of two molecules of FPP into squalene. This step is crucial for the synthesis of ergosterol, which is vital for maintaining the integrity and fluidity of fungal cell membranes. Inhibitors of squalene synthase have been explored as potential antifungal agents due to their ability to disrupt ergosterol biosynthesis .
While there is no specific data available on recombinant Ustilago maydis squalene synthase (ERG9), research on related enzymes and pathways provides valuable insights:
Metabolic Pathway Engineering: Studies on U. maydis have demonstrated its potential as a host for metabolic pathway engineering, particularly for the production of terpenoids and carotenoids .
Squalene Synthase in Other Organisms: In organisms like Candida glabrata, squalene synthase is essential for ergosterol biosynthesis, but its depletion does not impair growth in certain conditions due to the incorporation of exogenous sterols .
The recombinant expression of squalene synthase in U. maydis could have several applications:
Biotechnological Production: Enhancing sterol biosynthesis pathways could lead to improved production of valuable compounds like ergosterol or other sterol derivatives.
Metabolic Engineering: Understanding and manipulating the sterol biosynthesis pathway could provide insights into optimizing metabolic fluxes for the production of diverse compounds.
Organism | Function of Squalene Synthase | Importance |
---|---|---|
Ustilago maydis | Converts FPP to squalene for sterol biosynthesis | Essential for ergosterol production |
Candida glabrata | Critical for ergosterol biosynthesis, but not essential in vivo due to exogenous sterol incorporation | Potential antifungal target |
Saccharomyces cerevisiae | Essential for cell viability due to ergosterol biosynthesis | Target for antifungal drugs |
Recombinant Ustilago maydis Squalene Synthase (ERG9) catalyzes the condensation of two farnesyl pyrophosphate moieties to form squalene. This enzyme is the first committed step in the sterol biosynthesis pathway and is essential for ergosterol biosynthesis.
KEGG: uma:UMAG_04374
STRING: 5270.UM04374P0
Ustilago maydis offers several distinct advantages as a recombinant expression host for terpenoid biosynthesis compared to conventional systems. As a basidiomycete model organism, U. maydis provides metabolic compatibility with other higher basidiomycetes, potentially enabling more efficient expression of fungal sesquiterpenoid synthases. The fungus possesses an innate tolerance for substances that may be toxic to other microorganisms, which is particularly valuable when producing bioactive compounds. Additionally, U. maydis has a well-annotated genome, established molecular tools, and marker-free strain generation capabilities, allowing for sophisticated genetic engineering approaches. From a safety perspective, U. maydis is non-pathogenic in its yeast form and has been consumed by humans for centuries in Mexico (as corn smut or "huitlacoche"), suggesting minimal human health concerns for biotechnological applications . These characteristics collectively make U. maydis an excellent chassis for recombinant protein expression, particularly for enzymes involved in complex secondary metabolite pathways like the mevalonate pathway.
The isoprenoid biosynthesis pathway in U. maydis follows the evolutionarily conserved mevalonate (MVA) pathway found in most eukaryotes, which produces the key intermediates isopentyl diphosphate (IPP) and dimethylallyl diphosphate (DMAPP). These serve as building blocks for all terpenoids. In U. maydis, this pathway begins with Acetyl-CoA C-acetyltransferase (Aat1), which catalyzes the condensation of two acetyl-CoA molecules. Subsequent enzymatic steps through hydroxymethylglutaryl-CoA synthase (Hcs1) and other MVA pathway enzymes produce farnesyl pyrophosphate (FPP), which is the direct precursor for squalene synthase (potentially encoded by an ERG9 homolog). Interestingly, subcellular localization studies have revealed that, unlike in some other organisms, the initial reactions of the MVA pathway in U. maydis may occur in peroxisomes rather than the cytosol or endoplasmic reticulum, as Aat1 localizes to peroxisomes despite lacking a canonical peroxisomal targeting sequence . This unique subcellular compartmentalization may have implications for the optimization of recombinant squalene synthase expression and activity, as it suggests that metabolic engineering approaches may need to consider spatial organization of pathway enzymes to maximize precursor availability.
U. maydis naturally produces several terpenoid compounds, including carotenoids such as β-carotene and retinal, which compete with sterol biosynthesis for the common precursor FPP. Understanding this native metabolic context is crucial when designing expression strategies for recombinant squalene synthase. Previous studies have demonstrated that modifying the carotenoid pathway in U. maydis, such as deleting the bifunctional enzyme Car2 (which functions as both phytoene synthase and lycopene cyclase), can redirect metabolic flux toward heterologous pathways . This approach has been successful for producing other terpenoids like (+)-valencene and α-cuprenene. When expressing recombinant squalene synthase, researchers should consider similar strategies to reduce competition for FPP from native pathways. Additionally, the natural tolerance of U. maydis for terpenoid compounds suggests that engineering the upstream mevalonate pathway by overexpressing rate-limiting enzymes (such as the truncated HMG-CoA reductase Hmg1 N Δ 1–932) could increase precursor availability without toxic effects, potentially enhancing squalene synthase productivity . This metabolic engineering approach should be balanced against the needs of the organism for essential sterols, as complete redirection away from ergosterol biosynthesis would likely be detrimental to growth.
For recombinant squalene synthase expression in U. maydis, the selection of an appropriate promoter system is critical for achieving optimal enzyme production. Based on successful heterologous expression of other terpenoid pathway enzymes, the constitutively active promoter Prpl40, derived from the gene encoding ribosomal protein L40, has proven effective for consistent expression throughout the growth cycle . This promoter works well for metabolic enzymes that benefit from continuous expression. For higher expression levels or when temporal control is desired, the inducible Pcrg1 promoter (activated by arabinose and repressed by glucose) provides an excellent alternative, allowing for separated growth and production phases. The Petra promoter (tetracycline-responsive) offers tight regulation and dose-dependent expression, which may be advantageous for fine-tuning squalene synthase levels when excessive enzyme activity might create metabolic imbalances. When implementing these promoter systems, researchers should consider integrating expression constructs at specific genomic loci such as the ip locus (for single copy integration) or the corresponding native gene locus (if replacing the endogenous squalene synthase). It's advisable to include a purification tag (such as His6 or Myc) and verify expression through Western blot analysis before proceeding with activity assays or metabolite analysis .
For efficient expression of recombinant squalene synthase in U. maydis, several vector systems and transformation approaches have been validated. Self-replicating plasmids based on the autonomously replicating sequence (ARS) from U. maydis can be used for initial expression testing, though they may show plasmid instability without selection pressure. For stable integration, vectors containing homologous flanking sequences (typically 1kb upstream and downstream) facilitate targeted genomic insertion through homologous recombination. The pUMa vectors developed specifically for U. maydis provide modular cloning capabilities with various selection markers (hygromycin, carboxin, nourseothricin, or phleomycin resistance). For transformation, protoplast-mediated transformation yields the highest efficiency, requiring enzymatic digestion of the cell wall followed by PEG-mediated DNA uptake. A typical protocol includes growing cells to mid-log phase, digesting with lysing enzymes from Trichoderma harzianum or Novozyme 234 for 2-3 hours, gently pelleting and resuspending protoplasts in STC buffer (1 M sorbitol, 10 mM Tris-HCl pH 7.5, 100 mM CaCl2), mixing with linearized DNA and PEG solution, followed by regeneration on selective media . When selecting transformants, it's advisable to verify correct integration by PCR and confirm protein expression via Western blot before proceeding with functional analyses. For multiple genetic modifications, marker recycling using the FLP/FRT system enables sequential transformations without accumulating multiple resistance markers .
Codon optimization is a critical consideration when expressing heterologous squalene synthase genes in U. maydis, particularly if the source organism has significantly different codon usage patterns. Successful heterologous expression in U. maydis has been demonstrated with codon-optimized genes, such as the phytoene synthase from Agrobacterium aurantiacum (AaCrtB) . For squalene synthase from evolutionarily distant organisms (e.g., plants, mammals, or distant fungi), codon optimization should consider several U. maydis-specific parameters: favoring codons with abundant corresponding tRNAs, adjusting the GC content to match U. maydis preferences (approximately 54%), eliminating rare codons that might cause translational pausing, and removing any cryptic splice sites or transcription termination sequences. Additionally, researchers should consider adding a Kozak-like sequence (CAAA) before the start codon to enhance translation initiation. When designing the codon-optimized sequence, it's also prudent to introduce unique restriction sites at strategic positions to facilitate subsequent cloning procedures, while ensuring these modifications don't alter the amino acid sequence. Several commercial services and free online tools can assist with U. maydis-specific codon optimization, though custom adjustments may be necessary based on the specific characteristics of the squalene synthase gene being expressed. Following synthesis of the optimized sequence, verification of full-length protein production using Western blot analysis with tagged constructs is essential before proceeding to functional studies .
Engineering the mevalonate (MVA) pathway in U. maydis to enhance precursor supply for squalene synthase involves strategic manipulation of key enzymatic steps. The most effective approach combines overexpression of rate-limiting enzymes with targeted modification of competing pathways. Hydroxymethylglutaryl-CoA reductase (Hmg1) represents a primary control point, and expression of a truncated version lacking the N-terminal regulatory domain (Hmg1 N Δ 1–932) has proven effective in increasing flux through the pathway . This modification liberates the catalytic domain from feedback inhibition, resulting in higher activity. Additionally, overexpression of Acetyl-CoA C-acetyltransferase (Aat1) and Hydroxymethylglutaryl-CoA synthase (Hcs1) further enhances precursor formation, creating a push effect toward farnesyl pyrophosphate (FPP) production. To reduce competition for FPP from native pathways, deletion of key enzymes in the carotenoid pathway (such as Car2) redirects metabolic flux toward sterol biosynthesis . A multi-gene expression approach using polycistronic constructs with 2A peptide sequences can ensure balanced expression of pathway enzymes. When implementing these modifications, researchers should maintain marker-free strains through FLP/FRT-mediated marker excision to enable multiple sequential engineering steps. Importantly, the subcellular localization of these enzymes must be considered, as Aat1 localizes to peroxisomes in U. maydis, suggesting that optimizing subcellular trafficking of pathway intermediates may further improve precursor availability for cytosolic or ER-bound squalene synthase .
Balancing recombinant squalene synthase expression with native sterol requirements in U. maydis requires a nuanced approach that maintains sufficient ergosterol biosynthesis for cellular functions while maximizing squalene production. Several strategies can achieve this equilibrium: First, implementing tunable promoter systems, such as the tetracycline-inducible Petra promoter, allows dynamic regulation of squalene synthase expression levels based on growth conditions. Second, a dual-enzyme approach that co-expresses both native and recombinant squalene synthases with distinct regulatory mechanisms can ensure baseline sterol production while enabling overproduction of squalene. Third, temporal separation of growth and production phases using inducible promoters permits robust cell growth before redirecting metabolic flux toward squalene production. Fourth, supplementing growth media with ergosterol or intermediate sterols during high-level squalene synthase expression can reduce dependence on de novo sterol synthesis. Fifth, implementing a feedback-insensitive variant of the native squalene synthase can maintain essential sterol production even when sterol levels increase. When employing these strategies, researchers should carefully monitor growth rates, stress responses, and membrane integrity, as significant disruption of sterol homeostasis can lead to pleiotropic effects on cellular physiology, including altered stress resistance and morphological abnormalities . Additionally, microscopic examination of lipid droplet formation can serve as an indicator of squalene accumulation without disrupting sterol pathways, as excess squalene is often sequestered in these organelles.
Accurate detection and quantification of squalene from recombinant U. maydis strains requires sophisticated analytical approaches due to the complex cellular matrix. Gas chromatography-mass spectrometry (GC-MS) offers high sensitivity and specificity for squalene analysis, typically requiring derivatization with trimethylsilyl reagents to improve volatility. High-performance liquid chromatography (HPLC) with either UV detection (at 210 nm) or evaporative light scattering detection (ELSD) provides another robust approach, particularly when using reversed-phase C18 columns with isocratic methanol or acetonitrile mobile phases. For absolute quantification, isotope dilution mass spectrometry using deuterated squalene as an internal standard enables precise measurements even at low concentrations. Sample preparation significantly impacts analysis quality, with optimal protocols involving cell disruption by glass bead homogenization in the presence of antioxidants (such as BHT) to prevent squalene oxidation, followed by liquid-liquid extraction with hexane or chloroform:methanol mixtures. When analyzing strains with potentially low production levels, solid-phase extraction (SPE) using silica or C18 cartridges can concentrate samples prior to instrumental analysis. Additionally, thin-layer chromatography (TLC) with vanillin-sulfuric acid spray reagent serves as a rapid screening method for transformants, though it offers limited quantitative capabilities. Researchers should validate their analytical methods using squalene standards and establish calibration curves spanning expected concentration ranges, with appropriate quality controls to ensure reproducibility across experimental batches .
Determining enzyme kinetics of recombinant squalene synthase in U. maydis extracts requires careful consideration of assay conditions and potential interfering factors. A robust approach involves preparing microsomal fractions through differential centrifugation, as squalene synthase is membrane-associated. Cell disruption should be performed using gentle methods (such as enzymatic spheroplasting followed by osmotic lysis) to preserve enzymatic activity. The standard assay measures the conversion of radiolabeled farnesyl pyrophosphate (³H-FPP or ¹⁴C-FPP) to squalene in the presence of NADPH and detergent (typically Triton X-100 at 0.1%) to solubilize the enzyme while maintaining activity. Reaction products are extracted with hexane or petroleum ether and quantified by scintillation counting or radiochromatography. For non-radioactive alternatives, LC-MS/MS can be employed using synthetic FPP substrates with subsequent detection of squalene formation. To determine Km and Vmax values, substrate concentrations should span at least 0.2-5 times the Km (typically 1-20 μM for FPP), with appropriate enzyme dilutions ensuring linear reaction rates. When analyzing recombinant strains, parallel assays with wild-type extracts allow normalization for background activities. Potential complications include product inhibition by squalene, presence of phosphatases degrading FPP, and competing FPP-utilizing enzymes, necessitating appropriate controls. Including phosphatase inhibitors (sodium fluoride, sodium orthovanadate) and selective inhibition of competing pathways improves specificity. For tagged squalene synthase variants, immunoprecipitation prior to activity assays can isolate the recombinant enzyme from native activities .
Transcriptomics and proteomics approaches provide powerful tools for systematically optimizing recombinant squalene synthase expression and activity in U. maydis by identifying bottlenecks and unexpected interactions. RNA-Seq analysis comparing wild-type and recombinant strains can reveal compensatory changes in gene expression, particularly in sterol biosynthesis, stress response pathways, and membrane homeostasis genes. This information helps identify potential limiting factors or toxic intermediates affecting cellular physiology. Time-course transcriptomics during induction of squalene synthase expression can pinpoint optimal harvesting times and reveal temporal adaptation mechanisms. Proteomics using LC-MS/MS with isobaric labeling (TMT or iTRAQ) enables quantitative comparison of protein abundance changes, while phosphoproteomics can identify regulatory mechanisms affecting pathway enzymes. Combining these data with metabolomics creates a systems biology perspective revealing how metabolic flux is redistributed upon recombinant expression. For practical optimization, these approaches can guide several strategies: identifying limiting factors in the precursor pathway amenable to overexpression, revealing unexpected stress responses requiring media optimization, discovering co-factors or partner proteins that might enhance squalene synthase folding or activity, and detecting bottlenecks in secretion or protein processing machinery. When designing such studies, researchers should include appropriate controls, such as strains expressing non-functional squalene synthase variants, to distinguish effects specific to enzyme activity versus protein expression burden. Database resources specific to U. maydis, combined with pathway analysis tools, facilitate interpretation of the complex datasets generated by these omics approaches .
High-level functional expression of recombinant squalene synthase in U. maydis faces several challenges that require systematic troubleshooting. First, protein misfolding may occur, particularly with heterologous squalene synthases from evolutionarily distant organisms. This can be addressed by co-expressing molecular chaperones (such as Hsp70 or Hsp90) or implementing low-temperature expression strategies that slow protein synthesis and allow proper folding. Second, membrane insertion issues may arise with the C-terminal transmembrane domain of squalene synthase. Creating chimeric proteins with the transmembrane domain from a native U. maydis membrane protein can improve insertion efficiency. Third, metabolic toxicity from imbalanced precursor flux or squalene accumulation may inhibit growth. Implementing inducible expression systems with careful titration of induction levels can mitigate these effects, as can temporal separation of growth and production phases. Fourth, translation efficiency might be suboptimal despite codon optimization. Introducing a synthetic 5' untranslated region with optimized ribosome binding features can enhance translation initiation. Fifth, proteolytic degradation of heterologous squalene synthase may occur. Testing various purification tags (His6, FLAG, Strep) at both N- and C-termini can identify constructs with improved stability, while co-expression of protease inhibitors or using protease-deficient strains provides additional protection. Additionally, challenges with precursor availability can be addressed through the metabolic engineering strategies previously discussed, including overexpression of MVA pathway enzymes and reduction of competing pathways . When implementing these solutions, a combinatorial approach testing multiple strategies simultaneously in a small-scale expression screen often identifies optimal conditions most efficiently.
Troubleshooting product toxicity when overexpressing squalene synthase in U. maydis requires a multifaceted approach addressing both direct toxicity mechanisms and indirect metabolic perturbations. First, researchers should implement regulated expression systems, such as the arabinose-inducible Pcrg1 or tetracycline-regulated Petra promoters, to precisely control expression levels and determine the toxicity threshold. Titration experiments with varying inducer concentrations can identify optimal expression levels that balance production with cellular health. Second, growth media optimization can mitigate toxicity effects, with supplementation of ergosterol or intermediates potentially reducing feedback inhibition of essential pathways. Third, engineering the native lipid droplet formation machinery through overexpression of proteins involved in neutral lipid storage (such as acyltransferases) can sequester excess squalene and reduce membrane disruption. Fourth, implementation of efflux pumps or transporters capable of exporting squalene from cells provides another detoxification strategy, potentially creating a two-phase fermentation system where squalene partitions into an organic overlay. Fifth, adaptive laboratory evolution under gradually increasing squalene synthase expression can select for strains with improved tolerance mechanisms. When diagnosing toxicity, researchers should examine multiple cellular parameters including growth rates, membrane integrity (using propidium iodide staining), reactive oxygen species formation (with H2DCFDA), and subcellular ultrastructure changes (via transmission electron microscopy). Additionally, transcriptome analysis comparing wild-type and recombinant strains can identify specific stress response pathways activated by squalene accumulation, providing molecular targets for further engineering to enhance tolerance .
Addressing poor growth or decreased viability in recombinant U. maydis strains expressing squalene synthase requires a comprehensive approach targeting multiple potential causes. First, researchers should implement balanced pathway engineering rather than focusing solely on squalene synthase overexpression. Co-expression of downstream enzymes such as squalene epoxidase can prevent squalene accumulation while maintaining metabolic flux through the pathway. Second, media composition optimization, particularly carbon source selection, can significantly impact strain performance. Complex carbon sources like sucrose or maltose often provide better energy balance than glucose for strains with high metabolic burden. Third, supplementation with sterols or precursors can alleviate deficiencies caused by pathway imbalances. Ergosterol supplementation at 20-50 μg/ml can support growth when native sterol synthesis is compromised. Fourth, two-stage fermentation strategies separating growth and production phases allow biomass accumulation under non-producing conditions before initiating squalene synthase expression. Fifth, cultivation parameters including temperature reduction (to 25°C instead of 30°C), increased aeration, and pH stabilization can reduce metabolic stress and improve viability. Additionally, genetic modifications to enhance stress tolerance, such as overexpression of antioxidant enzymes (superoxide dismutase, catalase) or membrane lipid composition alterations, can improve strain robustness. When implementing these strategies, researchers should use appropriate controls such as strains expressing non-catalytic squalene synthase variants to distinguish between toxicity from enzyme activity versus protein expression burden. Continuous monitoring of growth parameters through automated systems provides early detection of viability issues, allowing timely intervention with optimized conditions .
CRISPR/Cas9 gene editing offers transformative approaches for optimizing U. maydis strains for squalene production through precise, multiplexed genetic modifications. This technology enables several sophisticated engineering strategies beyond traditional homologous recombination methods. First, simultaneous editing of multiple target genes involved in competing pathways (such as ergosterol biosynthesis regulators and carotenoid pathway enzymes) can redirect metabolic flux without sequential transformation rounds, significantly accelerating strain development. Second, CRISPR-mediated promoter replacement can fine-tune expression levels of native pathway genes, optimizing the balance between essential sterol production and squalene accumulation. Third, precise editing of regulatory regions controlling feedback inhibition can create semi-constitutive expression of rate-limiting enzymes while maintaining basic cellular regulation. Fourth, targeted mutagenesis of the native squalene synthase to modify product specificity or regulation while retaining U. maydis-specific protein interactions presents an alternative to heterologous expression. Fifth, genome-wide CRISPR screens using sgRNA libraries can identify unexpected genes affecting squalene production, revealing novel engineering targets. When implementing CRISPR/Cas9 in U. maydis, researchers should consider using the recently developed U. maydis-optimized Cas9 variants and validated sgRNA design algorithms to maximize editing efficiency while minimizing off-target effects. Ribonucleoprotein (RNP) delivery methods can provide effective editing while avoiding prolonged Cas9 expression. Additionally, CRISPR interference (CRISPRi) and CRISPR activation (CRISPRa) systems adapted for U. maydis would enable reversible and tunable regulation of pathway genes without permanent genetic modifications .
Synthetic biology approaches offer immense potential for creating modular, optimized terpenoid pathways in U. maydis through systematic redesign of metabolic architecture. These approaches extend beyond traditional metabolic engineering by implementing standardized genetic parts and rational design principles. First, developing a comprehensive library of characterized promoters, terminators, and ribosome binding sites specifically for U. maydis enables predictable control over gene expression levels. This parts collection should include constitutive promoters of varying strengths, inducible systems responsive to different signals, and synthetic regulatory elements. Second, implementing polycistronic expression cassettes using viral 2A peptides or internal ribosome entry sites (IRES) ensures stoichiometric expression of multiple pathway enzymes from a single transcript, maintaining pathway balance. Third, synthetic protein scaffolds can co-localize sequential pathway enzymes, creating metabolic channeling that improves flux and reduces intermediate diffusion. Fourth, orthogonal translation systems using expanded genetic codes could produce enzymes with novel amino acids at catalytic sites, potentially enhancing activity or stability. Fifth, genome-scale metabolic models calibrated specifically for U. maydis can predict optimal intervention points and expression levels for maximizing terpenoid production. When designing such systems, researchers should incorporate metabolic sensors that monitor key intermediates (such as FPP or squalene) and link them to reporter outputs, enabling real-time pathway optimization. Additionally, implementing standardized assembly methods like Golden Gate or Gibson Assembly facilitates rapid testing of different pathway configurations. These synthetic biology approaches, combined with the inherent advantages of U. maydis as a production host, could transform terpenoid production by creating predictable, modular systems adaptable to diverse terpenoid targets beyond squalene .
Integrating systems biology approaches with recombinant squalene synthase expression in U. maydis offers a comprehensive framework for understanding complex cellular responses and identifying non-intuitive optimization strategies. This integration enables several sophisticated analytical approaches. First, genome-scale metabolic modeling incorporating thermodynamic constraints can simulate the effects of squalene synthase overexpression on global metabolism, identifying potential bottlenecks and predicting optimal intervention points. These models can be progressively refined using experimental data from engineered strains. Second, multi-omics integration combining transcriptomics, proteomics, metabolomics, and fluxomics data provides a holistic view of cellular adaptation to recombinant expression. Network analysis of these datasets can reveal unexpected regulatory connections between squalene production and seemingly unrelated cellular processes. Third, experimental evolution combined with whole-genome sequencing of adapted strains can identify genetic changes conferring improved productivity or tolerance, revealing targets for rational engineering. Fourth, computational protein design could optimize squalene synthase for improved catalytic efficiency or stability in the U. maydis cellular environment through targeted mutations based on molecular dynamics simulations. Fifth, machine learning approaches trained on experimental datasets from varied strain designs can develop predictive models correlating genetic configurations with squalene production, enabling in silico strain optimization. When implementing these approaches, researchers should develop standardized cultivation and analytical protocols to ensure data comparability across experiments. Additionally, establishing an open data repository for U. maydis omics data would accelerate community-wide progress through collaborative analysis. These systems biology approaches transcend traditional strain engineering by providing mechanistic insights into the complex interplay between recombinant expression and native metabolism .