Fact-checked by Grok 2 weeks ago

Metabolic engineering

Metabolic engineering is the science of rewiring cellular through to enhance the production of native metabolites or to enable the synthesis of novel compounds in microorganisms, , or other organisms. This interdisciplinary field integrates principles from biochemistry, , and to optimize metabolic pathways for industrial applications. The origins of metabolic engineering trace back to the late 1980s and early 1990s, building on millennia-old practices of microbial while incorporating modern tools like technology and computational modeling. Pioneering works, such as those by in 1991 and Stephanopoulos and Vallino in 1991, formalized the approach by emphasizing the systematic analysis and redesign of metabolic networks to overcome bottlenecks in product formation. Over the decades, the field has evolved with advances in , enabling the construction of entirely new pathways and the use of iterative design-build-test-learn (DBTL) cycles to refine engineered strains. Key applications of metabolic engineering span the production of biofuels, pharmaceuticals, fine chemicals, and food additives, often achieving dramatic yield improvements through pathway optimization and flux redirection. Notable successes include the over 10,000-fold increase in via targeted strain engineering and the complete biosynthesis of complex opioids like in , demonstrating the potential for sustainable of high-value therapeutics. In recent years, the discipline has expanded to include fixation pathways and pan-genome-scale modeling, supporting greener bioprocesses and enhanced crop productivity in . These developments underscore metabolic engineering's role in addressing global challenges in , , and .

Fundamentals

Definition and Principles

Metabolic engineering is the targeted modification of cellular metabolic pathways in organisms such as microbes, , and animals to optimize the production of specific substances, including biofuels and pharmaceuticals, by altering native pathways or introducing ones. This practice involves optimizing genetic and regulatory processes to enhance the efficiency of cellular toward desired outcomes. Key principles of metabolic engineering include the overexpression of genes encoding enzymes in target pathways to increase toward the product, deletion or downregulation of competing pathways to redirect resources, cofactor balancing to maintain appropriate ratios such as NADPH/NADP+ for redox-dependent reactions, and ensuring the thermodynamic feasibility of engineered reactions to avoid energetic bottlenecks. A central goal is to maximize , defined as the of conversion to product, expressed as
Y = \frac{\text{moles of product}}{\text{moles of substrate}},
achieved by strategically redirecting metabolic through the aforementioned modifications. Metabolic serves as a foundational for quantifying and validating these flux redirections.
As an interdisciplinary field, metabolic engineering integrates biochemistry, genetics, and to systematically redesign metabolic networks. It differs from , which emphasizes the design of genetic circuits and novel biological functions, by primarily focusing on optimizing existing metabolic processes for enhanced productivity. This approach enables the sustainable production of chemicals from renewable resources, thereby reducing dependence on petrochemical feedstocks.

Metabolic Pathways and Networks

Metabolic pathways consist of sequences of enzymatic reactions that convert substrates into products, thereby transforming matter and energy within a . These pathways are essential for cellular functions such as energy production, of biomolecules, and maintenance of . Central examples include , which breaks down glucose to pyruvate to generate ATP (and occurs in both aerobic and anaerobic conditions); the tricarboxylic acid (TCA) cycle, also known as the Krebs cycle, which oxidizes derived from carbohydrates, fats, and proteins to produce reducing equivalents for the ; and the (PPP), which generates NADPH for reductive and ribose-5-phosphate for synthesis. In , for instance, the enzyme (PFK) catalyzes a key committed step, the phosphorylation of fructose-6-phosphate to fructose-1,6-bisphosphate, and serves as a primary regulatory point. Metabolic networks represent the interconnected web of these pathways, forming a where metabolites act as nodes and enzymatic reactions as directed edges linking substrates to products. Within this framework, metabolic denotes the rate at which metabolites flow through reactions, quantified as the turnover rate of a metabolite per unit time, which determines the overall efficiency and capacity of the network. Bottlenecks in these networks often arise from rate-limiting enzymes, whose low activity or restricts through downstream reactions, thereby constraining cellular . Regulation of metabolic pathways and networks occurs at multiple levels to ensure responsiveness to cellular needs. Allosteric control modulates enzyme activity through binding of effectors at sites distinct from the , while inhibition prevents overproduction by end-product repression of upstream enzymes—for example, inhibits threonine deaminase, the first enzyme in its biosynthetic pathway in . further fine-tunes expression of metabolic genes via transcription factors that respond to environmental cues or metabolite levels, such as MarR family proteins that control enzyme-encoding genes in response to ligands. A fundamental mathematical representation of these networks is the stoichiometric matrix S, an m \times n matrix where m is the number of metabolites and n the number of reactions; at , the balance equation S \mathbf{v} = 0 holds, with \mathbf{v} as the flux vector, enforcing across the network. Organism-specific variations in metabolic pathways and networks reflect evolutionary adaptations, particularly in cellular architecture. In prokaryotes like E. coli, metabolism occurs in a single cytoplasmic compartment without membrane-bound organelles, allowing rapid diffusion of metabolites and streamlined flux through interconnected pathways. In contrast, eukaryotes such as (Saccharomyces cerevisiae) exhibit compartmentalization, where pathways are segregated into organelles—for instance, in the , the cycle in mitochondria, and parts of the in both and plastids in plants—enabling specialized regulation and preventing interference between incompatible reactions. This compartmentalization in eukaryotes introduces additional transport steps across membranes, influencing and flux distribution compared to the more unified prokaryotic systems.

Historical Development

Origins and Early Milestones

The origins of metabolic engineering lie in the classical strain improvement techniques of the mid-20th century, which relied on random and selection to enhance microbial production of valuable metabolites. In the 1950s, researchers at Kyowa Hakko Kogyo Co. Ltd. discovered that auxotrophic mutants of Corynebacterium glutamicum could overproduce , establishing the foundation for industrial fermentation and demonstrating how disruptions in metabolic regulation could redirect carbon flux toward product accumulation. By the , iterative rounds of mutagenesis and medium optimization in C. glutamicum had improved L-lysine titers to approximately 50 g/L in industrial fermentations, highlighting the potential and limitations of empirical approaches to pathway manipulation. The field of metabolic engineering emerged in the early as a rational alternative to these random methods, integrating technology with quantitative analysis of networks. In 1991, James E. Bailey coined the term "metabolic engineering" in a seminal paper, defining it as the purposeful alteration of cellular through to achieve desired physiological properties, such as enhanced metabolite overproduction or bioconversion efficiency. Bailey emphasized the need for a systematic that combines kinetic modeling, , and targeted gene modifications to optimize flux through metabolic pathways. Early experimental milestones in the 1990s validated these concepts through targeted genetic interventions. One of the first demonstrations involved the 1991 overexpression of plasmid-encoded enzymes in to amplify metabolite flux, showing how could increase pathway throughput without relying on . In 1995, engineers introduced xylose metabolism genes (from ) into , enabling efficient production from sugars in hydrolysates and achieving yields comparable to glucose (approximately 0.45 g/g ). Key figures like provided theoretical frameworks that guided initial applications, particularly in . These proof-of-concept studies established metabolic engineering as a discipline distinct from traditional , paving the way for more sophisticated pathway optimizations.

Modern Advances

In the 2000s, metabolic engineering expanded through its integration with , enabling a more holistic understanding of cellular via genome-scale reconstructions and predictive modeling. This synergy facilitated the shift from targeted genetic modifications to network-level optimizations, where computational simulations guided experimental designs to enhance metabolite production. A pivotal advancement was the development of the iJR904 genome-scale metabolic model for in 2003, which incorporated 904 genes and 931 reactions, allowing for predictive engineering of metabolic fluxes and identification of bottlenecks in industrial strains. This model laid the groundwork for iterative design-build-test-learn cycles, improving yields in biofuels and pharmaceuticals. A application emerged in 2006 with the engineering of to produce artemisinic acid, a precursor to the antimalarial drug , through a semisynthetic pathway developed by UC Berkeley and researchers, achieving titers of 100 mg/L and demonstrating the feasibility of heterologously expressing complex plant pathways in microbial hosts. The marked a deepening synergy with , particularly through modular pathway assembly techniques that standardized the construction of multi-gene cassettes for plug-and-play metabolic engineering. These approaches, such as and variants, enabled rapid prototyping of pathways by treating genetic elements as , reducing design times from years to months and accelerating optimization for . A notable commercial milestone was Genomatica's 2013 achievement of microbial 1,4-butanediol production in E. coli, reaching 18 g/L in fermenters and marking the first bio-based route to this precursor at industrial scale, which displaced processes and highlighted the economic viability of engineered microbes. Entering the 2020s, innovations like CRISPR-Cas9 have revolutionized precise genome editing in metabolic engineering, with multiplex strategies enabling simultaneous modifications across pathways to fine-tune flux without off-target effects. In plant systems, 2025 developments include the PULSE optogenetic system, which uses light-inducible promoters to control transgene expression in Marchantia polymorpha, expanding tools for dynamic regulation in non-model organisms. Artificial intelligence has further transformed design phases, as seen in the 2024 ecFactory pipeline, a computational tool that predicts optimal gene knockout targets to enhance production of 103 diverse chemicals in E. coli using enzyme-constrained metabolic models, achieving up to 10-fold yield improvements in silico validations. Key milestones underscore the field's maturation, including the 2020 Nobel Prize in Chemistry awarded to and for CRISPR-Cas9, which has exponentially accelerated metabolic pathway refactoring by democratizing high-throughput editing tools. Commercial successes, such as DuPont's engineered E. coli for production, scaled to approximately 45,000 metric tons annually since the mid-2000s, exemplify how metabolic engineering has integrated into global supply chains for bio-based polymers.

Engineering Methods

Genetic and Molecular Tools

Genetic and molecular tools form the cornerstone of metabolic engineering, enabling precise manipulation of cellular pathways through targeted alterations at the DNA and RNA levels. These techniques allow engineers to introduce, amplify, or eliminate genes to redirect metabolic fluxes toward desired products, such as biofuels or pharmaceuticals. Key methods include plasmid-based expression systems for gene overexpression, recombineering and genome editing for knockouts, and seamless DNA assembly for constructing multi-gene pathways. Advanced regulatory elements further refine control over gene expression dynamics. Gene overexpression is commonly achieved using plasmid-based systems, which facilitate high-level production of target proteins without altering the host genome. In , the vector series, developed by F. William Studier, utilizes the strong T7 promoter to drive expression via T7 , enabling up to several grams per liter of recombinant protein in optimized conditions. For stable, long-term expression, chromosomal integration via Red recombineering offers an alternative, where linear products with homology arms are electroporated into cells expressing the phage-derived Red alpha, beta, and gamma proteins, achieving integration efficiencies of 10^4 to 10^6 transformants per microgram of DNA. This method, introduced by Datsenko and Wanner, minimizes plasmid copy number variability and burden on the host. The T7 promoter, specific to T7 , provides tight and high transcription rates—up to eight times faster than E. coli —preventing leaky expression in uninduced states. Gene knockout and deletion are essential for eliminating competing pathways and redirecting carbon flux. Traditional homologous recombination relies on host RecA-mediated repair but suffers from low efficiency in wild-type strains; enhancements using lambda Red systems improve this by promoting single-strand annealing, allowing scarless deletions. The advent of CRISPR-Cas9 has revolutionized multiplex knockouts, with a 2013 protocol by Jiang et al. enabling simultaneous editing of up to three bacterial loci using a single plasmid expressing Cas9 and guide RNAs, achieving efficiencies exceeding 90% for targeted deletions. For instance, knocking out the ldhA gene encoding lactate dehydrogenase in E. coli prevents lactate formation from pyruvate, redirecting flux toward succinate or other products; this modification, combined with additional knockouts like ptsG and pykA, has increased the succinic acid yield ratio by up to 11.5-fold under anaerobic conditions, as reported in studies on glucose metabolism. Recent advances include prime editing, which enables precise insertions, deletions, and base substitutions without double-strand breaks, improving efficiency for complex metabolic edits (as of 2024). Pathway assembly techniques enable the construction of complex, heterologous metabolic routes by joining multiple DNA fragments. The Gibson assembly method, reported by Gibson et al. in 2009, uses a one-pot reaction with T5 exonuclease, Phusion polymerase, and Taq ligase to create seamless overlaps, allowing assembly of 5-10 fragments up to 100 kb in length with efficiencies over 80% for multi-part joins. Complementing this, Golden Gate cloning employs type IIS restriction enzymes like BsaI to generate unique overhangs, facilitating modular, directional assembly of 20 or more parts in a hierarchical manner without internal restriction sites. These tools are pivotal for heterologous pathway transfer, such as inserting plant-derived terpenoid genes (e.g., amorphadiene synthase from Artemisia annua) into yeast chromosomes, enabling de novo production of antimalarial precursors like artemisinic acid at titers of 25 g/L in engineered Saccharomyces cerevisiae. Advanced tools provide finer post-transcriptional and enzymatic control. Riboswitches, elements that sense metabolites to modulate , have been engineered for dynamic in metabolic pathways; for example, synthetic theophylline-responsive riboswitches in E. coli achieve up to 100-fold induction of downstream genes upon ligand binding, enabling dynamic control of in metabolic pathways. enhances performance, using error-prone to introduce random mutations at rates of 1-5 per kb, followed by screening for improved kinetics; this approach has optimized keto acid decarboxylases in E. coli, increasing kcat/Km by over 100-fold to elevate production from glucose.

Computational Approaches

Computational approaches in metabolic engineering leverage mathematical modeling, , and optimization techniques to predict and metabolic network behaviors without relying solely on trial-and-error experimentation. These methods integrate genomic, biochemical, and physiological data to reconstruct and analyze cellular , enabling the identification of engineering targets for improved production of biofuels, pharmaceuticals, and other biomolecules. By simulating steady-state or dynamic fluxes, computational tools facilitate the rational of microbial strains, reducing time and costs in workflows. Genome-scale metabolic models (GEMs) form the cornerstone of these approaches, representing comprehensive reconstructions of an organism's entire , including thousands of reactions, s, and gene-protein-reaction associations. Tools like the Constraint-Based Reconstruction and Analysis () toolbox automate GEM reconstruction by incorporating constraints such as and thermodynamic feasibility, allowing users to simulate network-wide responses to genetic perturbations. For instance, the iJR904 model for , comprising 931 reactions and 904 genes, has been widely used to predict growth rates and yields under varying conditions. Flux balance analysis (FBA), a key constraint-based method within the framework, optimizes metabolic fluxes under steady-state assumptions to predict maximal theoretical yields or growth rates. Formulated as a problem, FBA solves for the objective function \max Z = c^T v subject to the stoichiometric constraints S v = 0, flux bounds lb \leq v \leq ub, and steady-state conditions, where S is the stoichiometric matrix, v is the flux vector, and c defines the objective (e.g., production). This approach excels in identifying strategies or pathway bottlenecks for enhancing product titers, as demonstrated in optimizing production pathways. For capturing time-dependent behaviors, such as transient responses to environmental changes or , dynamic modeling employs (ODE)-based simulations. Software like COPASI integrates these models by solving systems of the form \frac{dX}{dt} = N v(X), where X represents concentrations, N is the stoichiometric , and v(X) denotes rate laws (e.g., Michaelis-Menten ). This enables the simulation of oscillatory fluxes or regulations in engineered networks, providing insights into and control mechanisms. Recent advancements incorporate to enhance predictive accuracy, particularly in engineering and pathway design. For example, AlphaFold-inspired models predict protein structures to guide mutations that improve efficiency or specificity in metabolic pathways, as seen in redesigning polyketide synthases for novel production. Additionally, tools like OptFlux support , balancing trade-offs such as growth rate versus product yield through evolutionary algorithms and analysis, streamlining the selection of viable engineering candidates. Emerging trends include the use of large language models for metabolic pathway prediction and design (as of 2025).

Metabolic Flux Analysis

Modeling and Setup

In metabolic flux analysis (MFA), modeling and setup involve constructing a quantitative representation of the to enable steady-state flux predictions, typically through constraint-based approaches like (FBA). This preparatory phase focuses on defining the network's structure and boundaries to reflect biological realism while preparing for computational solving. The process begins with selecting relevant pathways and culminates in defining constraints that bound the feasible flux space, ensuring the model aligns with physiological conditions. Pathway selection is the initial step, where target metabolic pathways are identified based on the biological objective, such as for production in , and system boundaries are delineated to include relevant reactions while excluding extraneous ones. Incomplete networks often arise due to gaps in annotation, such as missing reactions or transporters; these are addressed through gap-filling algorithms that propose minimal additions to restore network functionality, for instance, enabling production under defined media. Tools like ModelSEED automate this by integrating genomic data with biochemical databases to predict and fill gaps, reducing manual curation needs and improving model completeness across diverse organisms. Stoichiometric modeling follows, constructing the stoichiometric matrix \mathbf{S} from reaction databases like , where rows represent metabolites and columns denote , with entries indicating stoichiometric coefficients (positive for products, negative for reactants). This matrix encodes the network's topology via the steady-state constraint \mathbf{S} \cdot \mathbf{v} = 0, where \mathbf{v} is the . To incorporate thermodynamics, group contribution methods estimate standard changes (\Delta_r G^{\circ}) for by summing contributions from molecular groups, enabling feasibility for flux directions and preventing thermodynamically implausible solutions. Jankowski et al. (2008) demonstrated that this approach can estimate \Delta_r G^{\circ} for 93%-97% of biochemical in databases like , with a of approximately 2 kcal/mol. Constraints are then established to define the solution space. The biomass equation serves as the primary objective function in FBA, representing the weighted of cellular components (e.g., proteins, ) from precursors, maximized to predict growth yield. Nutrient uptake rates provide environmental bounds, such as glucose limited to 10 mmol/gDW/h in E. coli models to mimic aerobic conditions. For eukaryotes, compartmentalization is modeled by assigning reactions to organelles (e.g., mitochondria, ) and including transport reactions across membranes, as in models where peroxisomal beta-oxidation is segregated to reflect spatial organization. Software tools facilitate automated reconstruction and setup. The RAVEN Toolbox streamlines genome-scale model building by parsing annotations, filling gaps, and generating the stoichiometric matrix, supporting both prokaryotic and eukaryotic networks. For instance, the iMM904 model of Saccharomyces cerevisiae was set up with 904 genes, 1,226 metabolites, and 1,577 reactions, incorporating compartmentalization for 8 intracellular compartments and a biomass objective tuned to experimental growth data.

Analysis Techniques

Steady-state analysis techniques are fundamental to interpreting metabolic fluxes in engineered systems, assuming constant metabolite concentrations over time. (FBA) computes the distribution of intracellular fluxes by solving a problem that maximizes an objective function, such as biomass production, subject to stoichiometric constraints and steady-state conditions (Sv = 0, where S is the and v the flux vector). This method predicts optimal flux distributions without requiring kinetic parameters, making it widely applicable for genome-scale models in metabolic engineering. For more precise flux estimation incorporating experimental data, 13C-metabolic flux analysis (13C-MFA) uses with 13C substrates to measure mass isotopomer distributions via techniques like or NMR. Fluxes are fitted by minimizing the least-squares error between observed and simulated labeling patterns: \min \| D - M(v) \|^2 where D represents measured isotopomer data, M(v) the model-predicted labeling as a function of fluxes v, and the optimization accounts for stoichiometry and carbon transitions. This approach resolves flux ambiguities in central , such as the split between and the , by leveraging the information-rich labeling patterns. Recent advances include boundary flux analysis (BFA), which quantifies large-scale metabolic phenotypes by analyzing changes in extracellular levels across cohorts, and flux potential analysis (FPA), which integrates relative levels with constraint-based modeling to predict flux changes using approaches (as of 2025). These methods enhance scalability for in metabolic engineering. Extensions of FBA address solution non-uniqueness and biological realism. Parsimonious FBA (pFBA) refines predictions by first maximizing growth (as in standard FBA) and then minimizing the sum of absolute among feasible solutions, favoring sparse, efficient distributions that align with minimal usage . For enumerating all possible pathway variants, elementary modes (EFM) decompose the into minimal, non-decomposable steady-state pathways that cannot be simplified further without violating constraints. EFMs reveal routes, such as bypasses or redundant pathways, aiding the identification of engineering targets like bottlenecks. Sensitivity analysis evaluates how fluxes respond to perturbations, highlighting control points in the network. By varying parameters such as , where maximum velocity is given by V_{\max} = k_{\cat} [E] (with k_{\cat} as and [E] as concentration), researchers compute response coefficients to pinpoint reactions with high leverage on overall flux. This perturbation-based approach, rooted in metabolic control analysis, quantifies robustness and identifies key regulatory enzymes without full kinetic models. Software tools facilitate these analyses on prepared models. INCA implements 13C-MFA, including isotopically non-stationary extensions, by simulating labeling dynamics and performing nonlinear least-squares fitting for estimation. OptGene integrates with evolutionary algorithms to infer genetic designs from computed distributions, such as gene knockouts that enhance target . These tools enable scalable computation of steady-state and sensitivities, supporting iterative metabolic engineering workflows.

Optimization and Validation

Optimization strategies in metabolic engineering utilize insights from flux analysis to direct targeted genetic modifications that enhance product formation. OptKnock, a bilevel programming approach, predicts gene deletions that maximize target metabolite by coupling production to biomass growth, thereby identifying minimal interventions for overproduction in microbes like . This method has guided strain redesigns, such as redirecting carbon toward succinate, achieving theoretical yields close to experimental maxima. Complementing this, robustness analysis assesses yield stability against uncertainties in or environmental conditions, employing sampling or interval analysis on flux balance models to prioritize resilient designs. Such evaluations ensure that predicted optimizations withstand real-world variability, as shown in genome-scale pathway studies where robust fluxes maintained over 80% of maximal output under flux perturbations. Experimental validation confirms these predictions by quantifying actual metabolic responses post-engineering. 13C-labeling experiments, where cells are fed position-specific 13C substrates, enable flux reconstruction through GC-MS measurement of isotopomer patterns in or central metabolites, providing intracellular flux maps with errors typically below 10%. Reporter metabolites—central nodes in the network whose connected reactions show coordinated changes in data—pinpoint flux bottlenecks, such as elevated flux through hubs during . Gene-level interventions are verified via qPCR to quantify mRNA levels and Western blots to assess protein abundance, ensuring expression matches intent; for example, transhydrogenase overexpression is routinely confirmed this way to validate balance adjustments. These steps integrate into iterative Design-Build-Test-Learn (DBTL) cycles, where validation data refines models for subsequent rounds. Flux gaps identified in analysis, such as insufficient NADPH for reductive , prompt targeted builds like transhydrogenase (pntAB) overexpression, which redirected reducing equivalents and boosted acetol flux by over 50% in glycerol-fed E. coli. Metrics like specific (g product/gDW/h) , with engineered strains often reaching 0.1–0.5 g/gDW/h for platform chemicals. Carbon yield validation against models, via 13C-MFA, demonstrates close alignment; in engineered for dihydroartemisinic acid (an precursor), experimental fluxes matched model predictions for pentose phosphate and contributions within 20%, confirming low baseline yields (~0.06 mol/100 mol glucose) and guiding further enhancements to approach theoretical maxima of 22 mol/100 mol glucose.

Applications

Industrial Biotechnology

Industrial biotechnology leverages metabolic engineering to enable the large-scale microbial production of biofuels, , and materials from renewable feedstocks, offering sustainable alternatives to processes and reducing . By optimizing metabolic pathways in organisms like and , engineers achieve high titers, rates, and yields that support economic viability, with processes often scaled via fed-batch to maintain nutrient levels and minimize inhibition. These advancements have transformed industries, enabling bio-based products that compete on cost while promoting circular economies through the use of biomass-derived sugars. In biofuel production, metabolic engineering has targeted advanced alcohols like , which can directly replace without engine modifications, enhancing energy density and sustainability. Engineered E. coli strains, incorporating pathways from diverse organisms, have achieved titers up to 22 g/L in shake-flask fermentations, demonstrating 86% of theoretical yield and paving the way for scalable processes. Similarly, , a precursor for and other fuels, has been produced at over 70 g/L using BASF's engineered Basfia succiniciproducens in fed-batch fermentations during the , with productivities exceeding 2.5 g/L/h and yields near 1 g/g glucose, supporting industrial biorefineries that valorize . These metrics underscore the economic impact, as bio-succinic acid reduces reliance on petroleum-derived routes and lowers production costs to below $2/kg. Commodity chemicals represent another cornerstone, with —a key for —produced commercially by via engineered E. coli expressing genes from . This process, commercialized around 2006, reaches titers of approximately 130 g/L in fed-batch mode, with yields of about 0.45 g/g glucose, enabling annual production exceeding 100,000 tons and displacing synthesis that emits significant CO₂. For , essential for nylon-6,6 production, metabolic pathways have been introduced into S. cerevisiae using reverse β-oxidation and enoate reductase enzymes, yielding up to 65 mg/L from glucose and establishing a bio-based route that avoids oxidation's environmental hazards. These developments highlight how pathway enhances by utilizing lignocellulosic feedstocks, potentially capturing a multibillion-dollar market while cutting use by up to 70%. In food and materials sectors, lactic acid production for bioplastics exemplifies industrial success, with NatureWorks' facility utilizing engineered Lactobacillus strains to generate over 150,000 tons annually of polylactic acid (PLA) precursor, achieving titers above 130 g/L and yields of 0.95 g/g in continuous fermentation. This supports biodegradable packaging and textiles, reducing plastic waste and petroleum dependency. Vanillin, the primary flavor in foods, has seen a 10-fold yield increase in engineered S. cerevisiae strains through flux redirection toward phenylpropanoid pathways, reaching 45 mg/L without toxicity issues, offering a natural alternative to synthetic production that dominates 85% of the 20,000-ton market. Scalability relies on fed-batch strategies that balance growth and production, targeting titers >100 g/L, rates >2 g/L/h, and yields >90% theoretical to ensure profitability, as seen in these cases where bio-products now comprise 10-20% of global supply for select chemicals. In 2025, advances in metabolic engineering have further improved titers for next-generation biofuels like butanol derivatives in Clostridium species, achieving over 90% theoretical yields from lignocellulosic feedstocks.

Biomedical Applications

Metabolic engineering has transformed biomedical applications by enabling the precise of high-value therapeutics and nutraceuticals through engineered , addressing challenges in supply, purity, and scalability for health-related molecules. Unlike bulk industrial chemicals, these efforts target low-volume, high-potency compounds such as pharmaceuticals and bioactive metabolites, often navigating stringent regulatory requirements for therapeutic and . Key strategies involve reconstructing complex biosynthetic pathways in microbial or hosts to yield precursors or active agents, with optimizations focusing on enhancement and mitigation to achieve clinically relevant titers. In pharmaceutical production, metabolic engineering has facilitated the semisynthesis of antimalarial by engineering to produce artemisinic acid, a direct precursor, at titers of 25 g/L through multi-gene pathway integration and fermentation optimization. Similarly, the taxol () biosynthetic pathway, a complex diterpenoid anticancer agent, has been partially reconstructed in using multi-gene cassettes to overproduce the early intermediate taxadiene at approximately 1 g/L, marking a 15,000-fold improvement over native levels and enabling scalable precursor supply. Another landmark example is the engineering of for shikimic acid production, a critical precursor for the (Tamiflu), where pathway modifications in a 2006 strain achieved yields of 57 g/L, providing an alternative to plant extraction during supply shortages. For nutraceuticals and therapeutics, metabolic engineering has boosted resveratrol production—a polyphenol with antioxidant and potential anti-aging benefits—in plants by introducing stilbene synthase genes, resulting in a 5-fold increase in transgenic tobacco upon elicitation. Engineered microbes have also been pivotal for insulin precursors, with Saccharomyces cerevisiae and E. coli strains optimized via promoter tuning and secretion signals to produce recombinant human insulin at industrial scales, comprising over 90% of global supply through proinsulin processing. Links to gene therapy highlight metabolic engineering's role in enhancing cellular therapies, such as modifying chimeric antigen receptor (CAR) T cells in the 2020s to improve persistence by rewiring glycolysis and oxidative phosphorylation pathways, thereby sustaining antitumor activity in immunosuppressive tumor microenvironments. Recent advances include 2025 efforts in plant metabolic engineering to elevate anti-cancer alkaloids like benzylisoquinolines (e.g., berberine analogs) in transient expression systems such as Nicotiana benthamiana, achieving up to 10-fold yield increases for chemotherapeutic precursors through pathway elucidation and co-expression modules.

Challenges and Future Directions

Technical and Biological Challenges

Metabolic engineering faces significant biological hurdles that limit the efficiency of engineered pathways. One major challenge is product , where accumulated metabolites inhibit cellular growth and ; for instance, concentrations exceeding 10% v/v can severely impair viability by disrupting membrane integrity and osmotic balance. Pathway imbalances often lead to the accumulation of toxic byproducts, as uneven expression or flux distribution diverts carbon toward unintended side reactions, reducing overall yields and stressing the host . Cofactor limitations further exacerbate these issues, particularly in systems where ATP scarcity restricts energy-intensive conversions, constraining flux through redox-dependent pathways like those involving NAD(P)H. Technical challenges compound these biological constraints, notably low enzyme stability in heterologous hosts, where many introduced proteins exhibit half-lives under 1 hour due to proteolytic degradation or suboptimal folding, leading to rapid loss of catalytic activity during production. Scalability from laboratory shake flasks to industrial 1000 L fermenters introduces inefficiencies, such as oxygen transfer limitations in aerated systems, which hinder high-density cultures and cause heterogeneous nutrient distribution, often resulting in reduced titers by orders of magnitude. Engineering complex pathways presents additional obstacles, especially in elucidating specialized like composite pathways exceeding 20 enzymatic steps, where incomplete knowledge of regulatory interactions and intermediate toxicities hampers reconstruction in microbial . C1 pathways, such as those converting to chemicals, suffer from inefficiencies, often resulting in low carbon yields due to thermodynamic barriers in formaldehyde fixation and high energy demands for carbon incorporation. Quantifying these challenges is complicated by cellular heterogeneity, where subpopulations within engineered cultures exhibit varying metabolic states, leading to inconsistent flux distributions and reduced predictability in production outcomes. Integration of omics data reveals further gaps, as transcriptomic profiles often fail to correlate with actual metabolic fluxes, owing to post-transcriptional regulation and environmental perturbations that decouple gene expression from enzymatic activity. The integration of (AI) and is revolutionizing metabolic engineering by accelerating the design-build-test-learn (DBTL) cycles essential for strain optimization. In 2025, the ecFactory platform emerged as a computational tool that predicts targets for enhancing of over 100 valuable chemicals in , incorporating constraints into genome-scale metabolic models to identify flux-rewiring opportunities with high accuracy. High-throughput in biofoundries now enable the screening of thousands of genetic variants daily, facilitating rapid iteration in DBTL workflows and reducing development timelines from years to months for industrial strains. Fusions with are expanding the scope of metabolic engineering through the creation of pathways for non-natural , enabling the production of novel biomolecules from simple carbon sources via multi-enzyme cascades. For instance, engineered strains have been optimized to biosynthesize branched-chain β,γ-diols and other non-canonical compounds, broadening applications in pharmaceuticals and . Optogenetic tools, such as the Plant-Usable Light Switch Elements () system, provide dynamic control over in , allowing light-inducible regulation of metabolic fluxes for enhanced production as demonstrated in 2025 applications. Sustainability prospects in metabolic engineering focus on engineering microbes to utilize C1 feedstocks like CO2, with modifications to improving carboxylation efficiency and reducing to boost carbon fixation rates. These advances support integrations, where waste streams such as plastics are upcycled into high-value chemicals via engineered pathways in , converting polyethylene terephthalate () hydrolysates into platform molecules without competing with food resources. Globally, metabolic engineering is poised to address challenges through advanced biofuels, with projections indicating that biofuels could account for up to 12% of demand by 2030 in net-zero scenarios, driven by engineered microbial strains for sustainable . In personalized medicine, engineered are emerging as targeted therapeutics, with modifications enabling gut modulation for treating metabolic disorders like through precise delivery of bioactive compounds.

References

  1. [1]
  2. [2]
    Metabolic Engineering: Methodologies and Applications
    ### Encyclopedia Intro: Metabolic Engineering
  3. [3]
    Metabolic engineering of microorganisms for carbon dioxide utilization
    This review covers the engineering of endogenous CO2 fixation pathways, the construction of novel synthetic pathways, and strategies to optimize metabolic flux ...
  4. [4]
    Metabolic Engineering in Plants: Advancing Crop Productivity and ...
    Jul 18, 2024 · This comprehensive review explores cutting-edge strategies for manipulating primary and secondary metabolic pathways in plants, utilizing advanced genetic ...
  5. [5]
    Metabolic Engineering and Synthetic Biology - NIH
    Mar 4, 2019 · Metabolic engineering seeks for the optimization of cellular processes, endemic to a specific organism, to produce a compound of interest from a substrate.
  6. [6]
    Metabolic Engineering: Past and Future - Annual Reviews
    Mar 27, 2013 · We present here a broad overview of the field of metabolic engineering, describing in the first section the key fundamental principles that ...
  7. [7]
    Genome-Scale Consequences of Cofactor Balancing in Engineered ...
    Nov 4, 2011 · Moreover, model guided cofactor balancing has emerged to help the metabolic engineering of microorganisms to fuels production. We used a ...
  8. [8]
    Thermodynamics of Metabolic Pathways - Wiley Online Library
    Jun 4, 2021 · The thermodynamic feasibility of a reaction is governed by its Gibbs free energy. This free energy can be calculated from the standard Gibbs ...
  9. [9]
    A mathematical framework for yield (vs. rate) optimization in ...
    Here, the nonlinear yield Y P / S = r P / r S has to be maximized explicitly since the substrate uptake rate at the maximum product yield is not known. In ...
  10. [10]
    Metabolic Pathway - an overview | ScienceDirect Topics
    Metabolic pathways are a series of biochemical reactions that convert substrates into products, transforming matter and energy within a cell.
  11. [11]
    Glycolysis: A multifaceted metabolic pathway and signaling hub
    Glycolysis is a highly conserved metabolic pathway responsible for the anaerobic production of adenosine triphosphate (ATP) from the breakdown of glucose ...Missing: definition | Show results with:definition
  12. [12]
    Energy metabolism in health and diseases - Nature
    Feb 18, 2025 · The TCA cycle is a central pathway for the complete oxidation of carbohydrates, fats, and proteins (amino acids) and represents a pivotal link ...
  13. [13]
    Pentose Phosphate Pathway - an overview | ScienceDirect Topics
    The pentose phosphate pathway (PPP) is a metabolic pathway that generates NADPH and ribose-5-phosphate, and produces biosynthetic reductant and precursor ...
  14. [14]
    Phosphofructokinase - an overview | ScienceDirect Topics
    As a regulatory enzyme of glycolysis, PFK is negatively inhibited by ATP and citrate and positively regulated by ADP. ATP serves as an allosteric inhibitor for ...
  15. [15]
    Metabolic networks: enzyme function and metabolite structure
    Connections between biochemical reactions via substrate and product metabolites create complex metabolic networks that may be analyzed using network theory.Missing: bottlenecks | Show results with:bottlenecks
  16. [16]
    Metabolic Flux - an overview | ScienceDirect Topics
    Metabolic flux is defined to be the turnover rate of a metabolite through a biochemical reaction (Nielsen, 2003; Stephanopoulos, 1999). Thus, a set of metabolic ...
  17. [17]
    Optimizing microbial networks through metabolic bypasses
    In fact, carbon metabolism can present bottlenecks that might be difficult to overcome by simply increasing the number of rate-limiting enzyme(s).Research Review Paper · 2. Cell Factories... · 2.1. Achieving...<|control11|><|separator|>
  18. [18]
    Cooperative binding of the feedback modifiers isoleucine and valine ...
    Control of the regulatory enzyme threonine deaminase from Escherichia coli is achieved by isoleucine inhibition and valine activation.
  19. [19]
    Regulation of Metabolic Pathways by MarR Family Transcription ...
    This review focuses on the role of ligand-responsive MarR family transcription factors in controlling expression of genes encoding metabolic enzymes.
  20. [20]
    NetRed, an algorithm to reduce genome-scale metabolic networks ...
    Theory and methods. A metabolic network at steady state is balanced as follows: S ⋅ v = 0. The stoichiometric matrix S is obtained from network ... metabolic ...
  21. [21]
    Designing microbial cell factories for programmable control of ...
    Compartmentalization of metabolic pathways in yeast mitochondria improves the production of branched-chain alcohols. Nat Biotechnol, 31 (2013), pp. 335-341.Missing: differences | Show results with:differences
  22. [22]
    METABOLIC COMPARTMENTATION - Annual Reviews
    There are several structural compartments in eukaryotes involved in the different aspects of carbohydrate metabolism. Glycolytic enzymes are cytosolic; Krebs ...
  23. [23]
    Lysine Fermentation: History and Genome Breeding - PubMed
    Lysine fermentation by Corynebacterium glutamicum was developed in 1958 by Kyowa Hakko Kogyo Co. Ltd. (current Kyowa Hakko Bio Co. Ltd.) and is the second ...
  24. [24]
    Industrial production of amino acids by coryneform bacteria
    The history of the species Corynebacterium as amino acid producer started in the 1950s when Dr Kinoshita was the first to discover that C. glutamicum is a ...
  25. [25]
    Toward a Science of Metabolic Engineering
    Toward a Science of Metabolic Engineering. James E. BaileyAuthors Info ... METABOLIC ENGINEERING IN METABOLITE OVERPRODUCTION, SCIENCE 252: 1675 (1991).
  26. [26]
    Metabolic Engineering of a Pentose Metabolism Pathway ... - Science
    The ethanol-producing bacterium Zymomonas mobilis was metabolically engineered to broaden its range of fermentable substrates to include the pentose sugar ...
  27. [27]
    An expanded genome-scale model of Escherichia coli K-12 (iJR904 ...
    Aug 28, 2003 · An expanded genome-scale metabolic model of E. coli (iJR904 GSM/GPR) has been reconstructed which includes 904 genes and 931 unique biochemical reactions.
  28. [28]
    Metabolic engineering of Escherichia coli for direct production of 1,4 ...
    May 22, 2011 · The organism produced BDO from glucose, xylose, sucrose and biomass-derived mixed sugar streams. This work demonstrates a systems-based ...Missing: 2013 | Show results with:2013
  29. [29]
    Multiplexed CRISPR technologies for gene editing and ... - Nature
    Mar 9, 2020 · Multiplexed CRISPR–Cas technologies are frequently used to simultaneously activate and repress multiple genes at once, an approach that has been ...
  30. [30]
    Computational biology predicts metabolic engineering targets for ...
    We developed a computational pipeline, ecFactory, designed to predict optimal gene targets for enhancing the production of 103 valuable chemicals by ...
  31. [31]
    Optogenetic control of transgene expression in Marchantia ...
    Most of the optogenetic tools adapted or developed for plants are incompatible with ... The utility of PULSE as a tool for the metabolic engineering of M.
  32. [32]
    Current advances in microbial production of 1,3‐propanediol
    Jun 14, 2021 · 1,3-Propanediol was mainly produced through two chemical methods developed by Shell and Degussa (now owned by DuPont). Shell uses ethylene oxide ...
  33. [33]
    One-step inactivation of chromosomal genes in Escherichia coli K ...
    It has also recently been shown that the λ Red (γ, β, exo) function promotes a greatly enhanced rate of recombination over that exhibited by recBC sbcB or recD ...
  34. [34]
    Metabolic Engineering of Escherichia coli for Enhanced Production ...
    This in silico analysis predicted that disrupting the genes for three pyruvate forming enzymes, ptsG, pykF, and pykA, allows enhanced succinic acid production.
  35. [35]
    Enzymatic assembly of DNA molecules up to several ... - PubMed
    The method uses a 5' exonuclease, DNA polymerase, and DNA ligase to assemble DNA by first creating single-stranded overhangs, then joining them.
  36. [36]
    Engineering and In Vivo Applications of Riboswitches - PubMed - NIH
    Jun 20, 2017 · In this review, we summarize the methods that have been developed to engineer new riboswitches and highlight applications of natural and ...
  37. [37]
    Genome-scale modeling for metabolic engineering - PMC - NIH
    The available stoichiometric information for a metabolic network is incorporated into a stoichiometric matrix S, in which rows represent metabolites and columns ...
  38. [38]
    High-throughput comparison, functional annotation, and metabolic ...
    One of the first methods, called “pathway hole filling,” involves adding an entire pathway if a gene can be associated with one or more steps of the pathway (44) ...
  39. [39]
    ModelSEED Biochemistry Database for the integration of metabolic ...
    Models can then be applied to automatically identify any gaps that interrupt these pathways and suggest new hypothesis-driven experiments to fill these gaps (1) ...
  40. [40]
    Advanced Stoichiometric Analysis of Metabolic Networks of ...
    The stoichiometric matrix (S) is constructed according to material balance equations. The measured fluxes are used to reduce the possible solution space ...
  41. [41]
    Group contribution method for thermodynamic analysis of complex ...
    This group contribution method is demonstrated to be capable of estimating Delta(r)G'(o) and Delta(f)G'(o) for the majority of the biochemical compounds and ...
  42. [42]
    The Biomass Objective Function - PMC - PubMed Central
    To computationally predict cell growth using FBA, one has to determine the biomass objective function that describes the rate at which all of the biomass ...
  43. [43]
    Identification of flux trade-offs in metabolic networks | Scientific Reports
    Dec 10, 2021 · Trade-offs are inherent to biochemical networks governing diverse cellular functions, from gene expression to metabolism.Missing: bottlenecks | Show results with:bottlenecks
  44. [44]
    Principles and functions of metabolic compartmentalization - PMC
    Oct 20, 2022 · Spatial separation of metabolic pathways enables rapid control of metabolite levels and coordination between pathways and the environment.
  45. [45]
    RAVEN 2.0: A versatile toolbox for metabolic network reconstruction ...
    RAVEN is a commonly used MATLAB toolbox for genome-scale metabolic model (GEM) reconstruction, curation and constraint-based modelling and simulation.
  46. [46]
    Improving the iMM904 S. cerevisiae metabolic model using ...
    In this paper we make use of the automated GrowMatch procedure for restoring consistency with single gene deletion experiments in yeast and extend the procedure ...
  47. [47]
    Stoichiometric flux balance models quantitatively predict growth and ...
    The flux balance model specified by these parameters was found to quantitatively predict glucose and oxygen uptake rates as well as acetate secretion rates.Missing: Analysis paper
  48. [48]
    13C metabolic flux analysis - PubMed
    Metabolic flux analysis using 13C-labeled substrates has become an important tool in metabolic engineering. It allows the detailed quantification of all ...Missing: paper | Show results with:paper
  49. [49]
    Omic data from evolved E. coli are consistent with computed optimal ...
    Jul 27, 2010 · Parsimonious enzyme usage FBA. pFBA is a bilevel linear programming optimization using the genome-scale constraint-based model of E. coli K-12 ( ...Results · Analysis Of Omics Data In... · Parsimonious Enzyme Usage...Missing: paper | Show results with:paper
  50. [50]
  51. [51]
    INCA: a computational platform for isotopically non-stationary ... - NIH
    INCA is the first publicly available software package that can perform both steady-state metabolic flux analysis and isotopically non-stationary metabolic flux ...
  52. [52]
    Evolutionary programming as a platform for in silico metabolic ...
    Dec 23, 2005 · We illustrate the principles and utility of OptGene algorithm by using three interesting metabolic engineering problems with the yeast ...
  53. [53]
    Genome-scale metabolic rewiring improves titers rates and yields of ...
    Oct 23, 2020 · We take the minimal cut set (MCS) approach that predicts metabolic reactions for elimination to couple metabolite production strongly with growth.
  54. [54]
    Efficient estimation of the maximum metabolic productivity of batch ...
    Jan 31, 2017 · This work presents an efficient method for the calculation of a maximum theoretical productivity of a batch culture system using a dynamic optimization ...Methods · Dynamic Flux Balance... · Dynamic Optimization<|control11|><|separator|>
  55. [55]
    Production of Succinic Acid From Basfia succiniciproducens - Frontiers
    Dec 23, 2021 · Basfia succiniciproducens is a facultative anaerobic capnophilic bacterium, isolated from rumen, that naturally produces high amounts of succinic acid.
  56. [56]
    High-flux isobutanol production using engineered Escherichia coli
    Promising approaches to produce higher alcohols, e.g., isobutanol, using Escherichia coli have been developed with successful results.
  57. [57]
    [PDF] Improving bio-based succinate production with Basfia ...
    The fermentation process must feature high product yields and titers of above 100 g L-1 combined with efficient productivities of at least 2.5 g L-1 h-1, while ...
  58. [58]
    Biosynthetic Pathway and Metabolic Engineering of Succinic Acid
    Mar 8, 2022 · In this review, different succinic acid biosynthesis pathways are summarized, with a focus on the key enzymes and metabolic engineering approaches.
  59. [59]
    Progress in 1,3-propanediol biosynthesis - PMC - PubMed Central
    Nov 29, 2024 · DuPont company developed a commercial biological route through E. coli strain metabolic engineering and fermentation for 1,3-PDO production ...
  60. [60]
    Biocatalytic production of adipic acid from glucose using engineered ...
    Adipic acid is produced from glucose using a three-stage fermentation process in engineered S. cerevisiae, using a yeast-based biocatalytic system.<|separator|>
  61. [61]
    Metabolic engineering strategies to bio-adipic acid production
    Adipic acid is the most industrially important dicarboxylic acid as it is a key monomer in the synthesis of nylon. Today, adipic acid is obtained via a chemical ...
  62. [62]
    Polymers Based on PLA from Synthesis Using D,L-Lactic Acid (or ...
    ... NatureWorks (USA) industrial large-scale PLA production plant, which currently operates with a production capacity of 150,000 metric tons/year [41,69,74,75].
  63. [63]
    Benchmarking two commonly used Saccharomyces cerevisiae ...
    S288c produces 10-fold more vanillin glucoside than CEN.PK in continuous cultivation. •. The higher yield by S288c is achieved during respiratory growth.
  64. [64]
    Metabolic Engineering of Yeast and Plants for the Production ... - NIH
    A 5-fold improvement of the resveratrol production was ... Stilbene Production and Biological Benefits of Resveratrol Synthesis in STS Engineered Plants.
  65. [65]
    Cell factories for insulin production - PMC - PubMed Central
    Oct 2, 2014 · Nowadays, recombinant human insulin is mainly produced either in E. coli or Saccharomyces cerevisiae. Using E. coli expression system, the ...
  66. [66]
    Metabolic engineering for optimized CAR-T cell therapy - PubMed
    Feb 22, 2024 · This Perspective will highlight key foundational studies that examine the relevant metabolic pathways required for effective T cell cytotoxicity ...Missing: 2020s | Show results with:2020s
  67. [67]
    Benzylisoquinoline alkaloid production: Moving from crop farming to ...
    Sep 2, 2025 · Benzylisoquinoline alkaloids (BIAs) are a diverse group of plant secondary metabolites that play a key role as analgesics, anti-cancer, and anti ...
  68. [68]
    A review of metabolic and enzymatic engineering strategies for ... - NIH
    Toxicity of pathway products or intermediates. Many over-produced metabolic products are toxic to the host organism, which is a serious consideration when ...
  69. [69]
    Engineering of Metabolic Pathways by Artificial Enzyme ... - Frontiers
    Metabolic regulation faces many challenges, including avoidance of flux imbalances ... In metabolic engineering where a natural endogenous biosynthetic pathway ...
  70. [70]
    Metabolic Engineering Through Cofactor Manipulation and Its ...
    Although it is generally known that cofactors play a major role in the production of different fermentation products, their role has not been thoroughly and ...
  71. [71]
    Maximizing the stability of metabolic engineering‐derived whole ...
    Jul 18, 2017 · Industry-relevant substrates and products often are often toxic in a concentration-dependent way and act on three different levels, affecting ...
  72. [72]
    Challenges to Ensure a Better Translation of Metabolic Engineering ...
    Metabolic burden is now generally recognized as a major hurdle to be overcome with consequences on genetic stability but also on the intensified performance ...Missing: imbalance | Show results with:imbalance
  73. [73]
  74. [74]
    Seven critical challenges in synthetic one-carbon assimilation and ...
    In this review, we summarize the challenges in establishing synthetic pathways for assimilation of C1 feedstocks in microbes and how to solve these issues.Introduction · Approaches, Tools, And... · Enzyme Engineering As A...
  75. [75]
    Bacterial Metabolic Heterogeneity: Origins and Applications in ...
    Bacteria within an isoclonal population display significant heterogeneity in metabolism, even under tightly controlled environmental conditions.
  76. [76]
    [PDF] Physiological limitations and opportunities in microbial metabolic ...
    Intracellular product accumulation can lead to shift of the equilibrium away from products, metabolic rerouting and increased toxicity168. This obstacle can ...
  77. [77]
    Frontiers in biofoundry: opportunities and challenges
    By leveraging advanced automation and digital technology to accelerate iterative DBTL cycles, biofoundries are capable of generating new high-throughput ...
  78. [78]
    Recent advances in biosynthesis of non-canonical amino acids and ...
    De novo synthesis usually refers to the synthesis of ncAAs from simple carbon sources or compounds through multi-enzyme cascade reactions. In this approach, the ...
  79. [79]
    Optimized Biosynthetic Pathway for Nonnatural Amino Acids: An ...
    Apr 23, 2025 · The EMP pathway, TCA cycle, and the biosynthesis of L-2-ABA are metabolically connected through the PEP-PYR-OAA (oxaloacetic acid). This node ...
  80. [80]
    Advances in analyzing and engineering plant metabolic diversity
    Jul 11, 2025 · The transfer of biosynthetic genes between plant species is a powerful tool for metabolite production and the dissection of metabolic ...
  81. [81]
    Engineering Rubisco to enhance CO2 utilization - PMC
    Dec 27, 2023 · Ribulose-1,5-bisphosphate carboxylase/oxygenase (Rubisco) is a pivotal enzyme that mediates the fixation of CO2.
  82. [82]
    Metabolic engineering of Escherichia coli for upcycling of ...
    Innovative approaches for upcycling PET waste into high-value chemicals can mitigate these issues while contributing to a circular economy. In this study ...<|separator|>
  83. [83]
    Biofuels Market Size, Share & Growth Analysis Report, 2030
    The global biofuels market size was estimated at USD 99.53 billion in 2023 and is projected to reach USD 207.87 billion by 2030, growing at a CAGR of 11.3% ...Missing: metabolic | Show results with:metabolic
  84. [84]
    Engineered probiotics: a new era in treating inflammatory bowel ...
    Nov 4, 2025 · They hold broad potential for treating metabolic abnormalities, chronic inflammation, and rare genetic diseases. Therapeutic mechanisms of ...