The reconstruction of metabolic pathways in non-model organisms is a cornerstone of modern synthetic biology, enabling the development of novel microbial cell factories for drug discovery and biomanufacturing.
The reconstruction of metabolic pathways in non-model organisms is a cornerstone of modern synthetic biology, enabling the development of novel microbial cell factories for drug discovery and biomanufacturing. This article provides a systematic guide for researchers and drug development professionals, covering the foundational principles, computational and experimental methodologies, and advanced optimization techniques required to overcome the challenges associated with these non-canonical systems. We explore the unique metabolic capabilities of non-model organisms like Zymomonas mobilis and Streptococcus pneumoniae, detail the use of tools such as CRISPR, genome-scale models, and databases like KEGG and BioCyc, and present rigorous validation frameworks. By integrating insights from comparative analyses of reconstruction tools and emerging machine learning approaches, this resource aims to equip scientists with the strategies needed to harness the biotechnological potential of non-model organisms for biomedical and clinical breakthroughs.
In the landscape of biological research and industrial biotechnology, non-model organisms are emerging as pivotal players. Unlike traditional model organisms such as Escherichia coli or Saccharomyces cerevisiae, non-model organisms are species that lack a comprehensive suite of established genetic tools, databases, and standardized protocols for research [1]. The study of these organisms is driven by the recognition that the vast majority of biological diversity and many industrially valuable traits reside outside the narrow spectrum of traditional model systems [2] [1].
The shift towards investigating non-model organisms is fundamentally altering industrial microbiology. These organisms often possess unique physiological traitsâsuch as exceptional stress tolerance, the ability to consume unconventional feedstocks, or the capacity to synthesize novel compoundsâthat are absent in established model systems [3] [4]. This document, framed within a thesis on metabolic pathway reconstruction, outlines the defining characteristics of non-model organisms, details their industrial advantages, and provides practical protocols for their study.
The term "model organism" has evolved to signify not only an organism that is inherently convenient for studying specific biological questions but also one for which a wealth of tools and resources exists, such as annotated genomes, mutant libraries, and standardized transformation protocols [1]. Consequently, a non-model organism is defined by a relative lack of these research infrastructures. These are often termed "non-model model organisms" (NMMOs) when they are chosen for their exceptional suitability to address a particular biological problem, despite the initial absence of genetic tools [1].
The primary distinctions between model and non-model organisms are summarized in the table below.
Table 1: Key Differentiating Features of Model vs. Non-Model Organisms
| Feature | Model Organisms | Non-Model Organisms |
|---|---|---|
| Genetic Toolkits | Extensive, standardized, and readily available (e.g., CRISPR, libraries of mutants). | Sparse, often need to be developed de novo or adapted from other species. |
| Genomic Resources | High-quality annotated genomes and comprehensive databases (e.g., Ecocyc for E. coli). | Genome sequences may be unavailable, preliminary, or poorly annotated. |
| Physiological Understanding | Well-characterized metabolism and genetics. | Metabolic pathways and genetic regulation are often poorly understood. |
| Research Community | Large, established community facilitating resource sharing. | Often studied by smaller, specialized groups. |
| Inherent Biological Traits | Chosen for convenience and rapid life cycles. | Chosen for unique, extreme, or industrially relevant phenotypes. |
A significant challenge in engineering non-model organisms is recalcitrance, or a natural resistance to genetic manipulation and tissue culture [2] [5]. This can be due to robust defense systems that destroy foreign DNA, complex polyploid genomes, or an inability to regenerate whole plants from single cells in the case of non-model plant species [2] [4].
Non-model organisms are treasure troves of unique biochemistry and robust physiology, making them exceptionally valuable for industrial applications. Their merits span multiple sectors, from the production of sustainable materials to environmental bioremediation.
These organisms often exhibit extraordinary capabilities refined by evolution to thrive in niche or extreme environments.
The unique traits of non-model organisms are being harnessed across various industries, as detailed in the table below.
Table 2: Industrial Applications of Non-Model Organisms
| Application Area | Example Organism(s) | Industrial Merit and Product |
|---|---|---|
| Biofuels & Chemicals | Zymomonas mobilis (bacterium), Oleaginous yeasts | High-yield production of bioethanol and biodiesel from mixed agrowaste hydrolysates [7] [3]. |
| Biomaterials | Corynebacterium glutamicum, Bacillus megaterium | Production of bioplastics such as polyhydroxyalkanoates (PHA) and amino acids for biopolymers [7]. |
| Environmental Remediation | Pseudomonas putida, Stenotrophomonas sp. | Degradation of pollutants including plastics, pesticides, and oil hydrocarbons; wastewater treatment [7] [2]. |
| Pharmaceuticals & High-Value Compounds | Streptomyces sp., Nannochloropsis | Production of antibiotics (e.g., Adriamycin), immunosuppressants (e.g., Cyclosporin A), and novel molecules discovered from unique metabolic pathways [7] [2]. |
Overcoming the recalcitrance of non-model organisms requires a systematic approach, from genomic characterization to the development of custom genetic tools. The following workflow and protocols outline this process.
Objective: To build a computational model that predicts an organism's metabolic capabilities from its genome sequence, guiding metabolic engineering strategies.
Materials:
Method:
gapseq with the genomic FASTA file as input. The software will identify protein-coding sequences and map them to metabolic reactions using homology searches [8].Objective: To develop a functional method for introducing and stably integrating genetic modifications into a recalcitrant non-model organism.
Materials:
Method:
The following table lists essential reagents for working with non-model organisms like Zymomonas mobilis.
Table 3: Key Research Reagent Solutions for Engineering Non-Model Microorganisms
| Reagent / Solution | Function / Application | Example Use Case |
|---|---|---|
| Demethylating E. coli Strain (e.g., Trans110) | Produces plasmids with host-specific methylation patterns, protecting them from degradation by restriction enzymes. | Essential for achieving high transformation efficiency in bacteria with active R-M systems [4]. |
| Endogenous CRISPR-Cas System Components | Provides a host-adapted machinery for programmable DNA cleavage, improving editing efficiency. | Using the native Type I-F system of Z. mobilis for reliable gene knockouts [3] [4]. |
| Temperature-Sensitive Plasmid Backbone | Allows for plasmid replication at a permissive temperature and loss at a non-permissive temperature. | Facilitates marker-free editing and enables multiple rounds of modification in the GW-ICE system [4]. |
| Genome-Scale Metabolic Model (GEM) | Serves as a computational blueprint to predict metabolic flux and identify engineering targets. | iIsor850 model for I. orientalis was used to pinpoint gene knockouts for coupling succinate production to growth [6]. |
| Homology-Directed Repair (HDR) Donor DNA | Serves as a template for precise gene insertions or corrections during CRISPR-Cas editing. | Used alongside CRISPR to introduce heterologous pathways (e.g., 2,3-butanediol pathway in Z. mobilis) [3]. |
| ABIL WE 09 | ABIL WE 09: Silicone Emulsifier for Research | ABIL WE 09 is a silicone-based wetting agent for creating stable water-in-oil emulsions in research settings. This product is For Research Use Only (RUO). |
| DC4 universal | DC4 Universal Reagent |
Non-model organisms represent the next frontier in industrial biotechnology. Their vast, untapped metabolic diversity offers sustainable solutions for producing energy, chemicals, and materials, and for addressing environmental pollution. While significant challenges in genetic recalcitrance remain, the protocols and strategies outlined hereâcentered on robust genomic analysis, sophisticated metabolic modeling, and the development of customized genetic toolkitsâprovide a clear roadmap for their domestication. Integrating these approaches will accelerate the transformation of these enigmatic organisms into efficient microbial cell factories, paving the way for a circular bioeconomy.
The reconstruction of metabolic pathways in non-model organisms represents a frontier in synthetic biology and metabolic engineering. A significant barrier in this field is the presence of dominant native metabolic pathways that effectively compete for central carbon metabolites, severely limiting the flux toward engineered, non-native products. The ethanologenic bacterium Zymomonas mobilis serves as a paradigm for this challenge. This organism possesses an exceptionally efficient native metabolism for ethanol production, where carbon flow through the Entner-Doudoroff (ED) pathway is predominantly directed toward ethanol via the pyruvate decarboxylase (PDC) and alcohol dehydrogenase (ADH) enzymes [3]. This innate metabolic architecture creates a formidable bottleneck for redirecting carbon toward alternative biochemicals, as the native pathway often constitutes over 97% of theoretical yield efficiency on a carbon basis [10]. Overcoming this dominance is not merely a technical hurdle but a fundamental requirement for transforming organisms with ideal industrial characteristics into versatile biorefinery chassis for a sustainable circular bioeconomy [3].
Zymomonas mobilis utilizes the Entner-Doudoroff (ED) pathway anaerobically, a rare characteristic that contributes to its exceptional ethanol production capabilities. The ED pathway generates only one net ATP per glucose molecule, compared to two ATP molecules produced by the more common Embden-Meyerhof-Parnas (EMP) pathway [10]. This lower energy yield results in reduced biomass formation, thereby directing a greater proportion of carbon toward ethanol production. The metabolic journey from glucose to ethanol in Z. mobilis involves several key steps: glucose is first converted to gluconate by glucose-fructose oxidoreductase, then to 2-keto-3-deoxy-6-phosphogluconate (KDPG) by gluconate dehydratase, and finally cleaved into glyceraldehyde-3-phosphate (GAP) and pyruvate by KDPG aldolase. Pyruvate is subsequently decarboxylated by PDC to acetaldehyde, which is then reduced to ethanol by ADH, regenerating NAD+ for glycolytic continuity [3] [10].
Attempts to engineer alternative metabolic routes in Z. mobilis have consistently encountered resistance from its native metabolic network. A particularly illustrative example is the failed attempt to implement the complete EMP pathway by expressing E. coli phosphofructokinase (Pfk I), both alone and in combination with fructose bisphosphate aldolase (Fba) and triose phosphate isomerase (Tpi) [10]. Contrary to predictions, this engineering effort did not establish a functional EMP flux but instead resulted in growth inhibition and mutations in the heterologous pfkA gene. Metabolomic analysis revealed that the homeostatic levels of glycolytic intermediates in Z. mobilis were incompatible with EMP flux, demonstrating how the native metabolomic context constrains potential engineering strategies [10].
Table 1: Failed Metabolic Engineering Attempts Against Dominant Pathways in Z. mobilis
| Engineering Strategy | Target Pathway | Experimental Outcome | Citation |
|---|---|---|---|
| Expression of E. coli Pfk I | EMP glycolysis | Growth inhibition; mutation of heterologous gene; no EMP flux established | [10] |
| Co-expression of Pfk I, Fba, and Tpi | EMP glycolysis | Glycerol production as side product; reverse operation of heterologous reactions | [10] |
| PPi-dependent Pfk expression | EMP glycolysis | No significant metabolic changes; excretion of dihydroxyacetone | [10] |
| Promoter replacement of pdc | Ethanol to lactate shift | Partial redirection; incomplete elimination of ethanol pathway | [3] |
A novel approach termed the Dominant-Metabolism Compromised Intermediate-Chassis (DMCI) strategy has been developed specifically to address the challenge of pathway dominance [3]. Instead of directly engineering the chassis for target biochemical production, this method involves first constructing an intermediate chassis with intentionally compromised dominant metabolism. In Z. mobilis, this was achieved by introducing a low-toxicity but cofactor-imbalanced 2,3-butanediol (2,3-BDO) pathway, which effectively diverted carbon flux from the dominant ethanol production route. This intermediate chassis served as a platform for subsequent engineering, ultimately enabling the construction of a high-efficiency D-lactate producer capable of achieving remarkable titers of >140 g/L from glucose and >104 g/L from corncob residue hydrolysate with yields exceeding 0.97 g/g glucose [3].
The implementation of sophisticated genome-scale metabolic models (GEMs) has proven indispensable for navigating the constraints imposed by dominant native metabolism. The development of enzyme-constrained models like eciZM547 represents a significant advancement over traditional stoichiometric models [3]. By integrating enzyme kinetic parameters and accounting for proteome limitations, these models can more accurately simulate flux distributions and identify potential bottlenecks before experimental implementation. For Z. mobilis, the eciZM547 model successfully predicted the shift from glucose-limited growth to proteome-limited growth at high substrate uptake rates and more accurately simulated carbon distribution between acetate and acetoin under aerobic conditions compared to previous models [3]. This predictive capability is crucial for designing effective strategies to overcome innate metabolic dominance.
Principle: Introduce a metabolic pathway with lower toxicity than the target product but sufficient carbon drain to weaken the dominant native pathway, creating an intermediate chassis for further engineering [3].
Materials:
Procedure:
Technical Notes: The 2,3-BDO pathway serves as an effective intermediate due to its NADH/NAD+ cofactor imbalance, which naturally limits its full dominance while sufficiently draining carbon from ethanol production.
Principle: Utilize enzyme-constrained genome-scale metabolic models (ecGEMs) to predict flux distributions and identify proteome limitations before experimental implementation [3] [12].
Materials:
Procedure:
Technical Notes: The enzyme-constrained model will show proteome-limited growth at high substrate uptake rates (>71 mmol/gDW/h for glucose in Z. mobilis), which is not predicted by traditional GEMs.
Diagram 1: Central carbon metabolism and engineering targets in Z. mobilis. The dominant native ethanol pathway (red) competes with engineered pathways (green) for pyruvate. Key enzymes: ZWF (glucose-6-phosphate dehydrogenase), GFOR (glucose-fructose oxidoreductase), GAD (gluconate dehydratase), EDA (KDPG aldolase), PDC (pyruvate decarboxylase), ADH (alcohol dehydrogenase), LDH (lactate dehydrogenase), ALS (acetolactate synthase), ALDC (acetolactate decarboxylase), BDH (butanediol dehydrogenase).
Diagram 2: DMCI strategy workflow. The approach involves creating an intermediate chassis with compromised dominant metabolism before introducing the target product pathway. ecGEM: enzyme-constrained genome-scale metabolic model; TEA: techno-economic analysis; LCA: life cycle assessment.
Table 2: Key Research Reagents for Engineering Non-Model Organisms
| Reagent/Category | Specific Examples | Function/Application | Experimental Context |
|---|---|---|---|
| Genome Editing Systems | CRISPR-Cas12a, Endogenous Type I-F CRISPR-Cas, MMEJ repair | Precise genome modification; essential for pathway integration and gene knockout | [3] [11] |
| Metabolic Modeling Software | COBRA Toolbox, AutoPACMEN (kcat prediction), MEMOTE (model evaluation) | Pathway simulation; prediction of flux distributions and enzyme limitations | [3] [12] |
| Analytical Chemistry | HPLC (product quantification), GC-MS (metabolite profiling), RNA-Seq (transcriptomics) | Validation of metabolic changes; systems biology analysis | [3] [13] [14] |
| Specialized Growth Media | ZRMG (standard growth), Modified ZYMM (N2-fixing conditions), CRH (lignocellulosic hydrolysate) | Physiological studies; industrial-relevant condition simulation | [10] [15] |
| Pathway Enzymes | 2,3-BDO pathway (alsS, alsD), D-LDH (D-lactate dehydrogenase), XI (xylose isomerase) | Metabolic pathway reconstruction; substrate utilization expansion | [3] [16] [14] |
| GC Lining Cement | GC Lining Cement, CAS:102087-30-9, MF:C26H44S8 | Chemical Reagent | Bench Chemicals |
| TRAGACANTHIN | Tragacanthin | Bench Chemicals |
The challenge of dominant native metabolism in non-model organisms like Zymomonas mobilis represents a significant but surmountable barrier in metabolic pathway reconstruction. The development of sophisticated strategies such as the DMCI approach, coupled with advanced modeling techniques and precise genome editing tools, has demonstrated that even exceptionally efficient native pathways can be redirected toward alternative products. The successful production of D-lactate at titers exceeding 140 g/L with yields >0.97 g/g glucose from Z. mobilis provides compelling evidence that these strategies can achieve commercial viability, as further supported by techno-economic analysis and life cycle assessment [3]. As the field progresses, the integration of multi-omics data, machine learning-assisted pathway design, and dynamic regulation systems will further enhance our ability to engineer non-model organisms with complex metabolic networks, ultimately expanding the repertoire of microbial chassis available for sustainable biochemical production.
Streptococcus pneumoniae is a significant global health concern, being a leading cause of community-acquired pneumonia, meningitis, and septicemia [17] [18]. This Gram-positive pathogen poses a substantial threat to young children, the elderly, and immunocompromised individuals, with an estimated one million child deaths annually attributed to pneumococcal disease [17]. The challenge in managing S. pneumoniae infections is compounded by the escalating prevalence of antimicrobial resistance, with over 40% of strains exhibiting resistance to penicillin and frequently demonstrating co-resistance to other antibiotics such as macrolides and tetracyclines [17] [19]. The World Health Organization has recognized this threat by adding S. pneumoniae to its updated Bacterial Priority Pathogens List as a medium-priority pathogen [17].
In the context of metabolic pathway reconstruction for non-model organisms, subtractive genomics represents a powerful computational approach for identifying novel therapeutic targets. This methodology leverages the growing availability of genomic data to systematically identify essential pathogen-specific proteins that are absent in the host, thereby facilitating the development of targeted therapies with minimal side effects [17] [20]. By focusing on non-host homologous genes involved in distinct metabolic pathways crucial for pathogen survival, this approach enables researchers to disrupt pathogen function while preserving host biology [17]. This case study details the application of subtractive genomics for identifying potential drug targets in S. pneumoniae, providing a comprehensive protocol for researchers engaged in metabolic pathway reconstruction and drug discovery.
The complex etiology of Streptococcus pneumoniae infection poses significant challenges in elucidating the molecular mechanisms underlying its pathogenesis [18]. With over 100 recognized serotypes, this pathogen exhibits remarkable genetic variability, with different serotypes demonstrating varying degrees of invasiveness and pathogenicity [17] [21]. Current vaccine strategies, including the 13-valent pneumococcal conjugate vaccine (PCV13) and the 23-valent pneumococcal polysaccharide vaccine (PPSV23), target specific capsular polysaccharide serotypes but face limitations due to emerging non-vaccine serotypes and the phenomenon of capsular switching [17] [19].
The genomic plasticity of S. pneumoniae enables rapid adaptation through competence-dependent horizontal gene transfer, facilitating the dissemination of resistance traits and pathogenic factors [17]. Recent genomic surveillance studies in Indian adult populations have revealed a high prevalence of multidrug resistance (observed in 70% of isolates) and the continuous emergence of novel sequence types through recombination events [19]. This dynamic evolutionary landscape underscores the critical need for novel therapeutic strategies that target essential metabolic pathways conserved across diverse strains.
Metabolomic analyses of S. pneumoniae infections have identified significant alterations in host metabolic profiles, with activation of pathways including galactose metabolism, the hypoxia-inducible factor-1 (HIF-1) signaling pathway, the citrate cycle, the pentose phosphate pathway, and glycolysis/gluconeogenesis [18]. These pathway perturbations represent potential vulnerabilities that can be exploited through targeted therapeutic interventions.
The subtractive genomics approach follows a systematic pipeline to filter and identify potential drug targets from the complete proteome of S. pneumoniae. The stepwise methodology is outlined below and visualized in Figure 1.
The complete genome assembly of S. pneumoniae (GCF002076835.1ASM207683v1protein.fasta) was retrieved from the National Center for Biotechnology Information (NCBI) database [17] [22]. The human proteome (GCF000001405.40GRCh38.p14protein.fasta) was similarly obtained for comparative analysis.
Redundancy elimination was performed using CD-HIT (version 4.8.1) with a 90% sequence identity threshold to cluster and remove duplicate protein sequences, ensuring only unique sequences were retained for subsequent analysis [17].
Protein sequences in S. pneumoniae lacking homologs in human proteins were identified using a BLASTp search against the Homo sapiens genome with an E-value cut-off of 10â5 [17] [20]. Sequences with significant similarity to human proteins were excluded to minimize potential cross-reactivity and host toxicity in subsequent drug development stages.
Table 1: Summary of Proteome Filtering Steps in Subtractive Genomics
| Filtering Stage | Proteins Remaining | Reduction Percentage | Tools/Databases Used |
|---|---|---|---|
| Initial S. pneumoniae Proteome | 2,027 | - | NCBI |
| After Redundancy Elimination | ~2,000 | 1.3% | CD-HIT (90% identity) |
| Non-Homologous to Human | ~2,000 | 0% | BLASTp (E-value: 10â»âµ) |
| Essential Genes | 48 | 97.6% | Database of Essential Genes (DEG) |
| After Gut Microflora Consideration | 21 | 56.3% | BLASTp against gut microbiome |
Essential genes for S. pneumoniae survival were identified using the Database of Essential Genes (DEG), which catalogs genes indispensable for bacterial survival under laboratory conditions [17] [22]. To further refine target selection and minimize disruption to beneficial microbiota, these essential genes were compared against the human gut microbiome proteome using BLASTp with the same E-value threshold, eliminating those with significant matches [17].
The resulting set of potential targets was subjected to Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis to identify metabolic pathways critical for bacterial survival [18] [20]. Additionally, subcellular localization predictions were performed to prioritize targets with accessible subcellular locations, particularly focusing on cytoplasmic membrane proteins that may be more readily targetable [23].
For targets lacking crystal structures, homology modeling was employed to generate three-dimensional structural models [17]. These models were then subjected to structure-based virtual screening of FDA-approved compound libraries to identify potential repurposing candidates, using molecular docking and molecular dynamics simulations to evaluate binding stability and interactions [17] [22].
Figure 1. Workflow for subtractive genomics analysis of S. pneumoniae. The pipeline systematically filters the bacterial proteome to identify potential drug targets that are essential for pathogen survival but absent in the host and beneficial microbiota.
Application of the subtractive genomics pipeline to S. pneumoniae yielded promising results for target identification. From an initial proteome of 2,027 proteins, approximately 2,000 were identified as non-homologous to human proteins [17]. Essential gene analysis identified 48 genes crucial for bacterial survival, which was further refined to 21 potential targets after considering preservation of human gut microflora [17] [22].
Key hub genes identified through protein-protein interaction analysis included gpi (glucose-6-phosphate isomerase), fba (fructose-bisphosphate aldolase), rpoD (RNA polymerase sigma factor), and trpS (tryptophan--tRNA ligase) [17]. These targets were associated with 20 distinct metabolic pathways essential for bacterial survival, with particular enrichment in carbohydrate metabolism and amino acid biosynthesis pathways.
Metabolomic studies of S. pneumoniae infections have revealed significant alterations in host metabolic pathways, providing additional context for target prioritization [18]. Comparative analysis of metabolic profiles between infected individuals and normal controls identified 418 metabolites that significantly contributed to group differentiation [18].
Table 2: Key Metabolic Pathways Altered in S. pneumoniae Infection
| Metabolic Pathway | Role in Pathogenesis | Potential for Therapeutic Targeting |
|---|---|---|
| Galactose Metabolism | Energy production and cell wall biosynthesis | High - Essential for bacterial growth |
| HIF-1 Signaling Pathway | Host immune response to infection | Medium - Host-pathogen interaction |
| Citrate Cycle (TCA Cycle) | Central energy metabolism | High - Essential for bacterial survival |
| Pentose Phosphate Pathway | Nucleotide synthesis and antioxidant defense | High - Essential for replication |
| Glycolysis/Gluconeogenesis | Carbohydrate metabolism and energy production | High - Primary metabolic pathway |
The identified metabolites were categorized into various groups, including amino acids, fatty acids, and phosphatidylcholine, with these metabolic alterations being implicated in the immune response to infection [18]. This comprehensive analysis of the metabolic network provides a foundational framework for targeting pathogen-specific metabolic vulnerabilities.
Virtual screening of 2,509 FDA-approved compounds against the prioritized targets identified Bromfenac as a leading repurposing candidate [17] [22]. This nonsteroidal anti-inflammatory drug exhibited a binding energy of -26.335 ± 29.105 kJ/mol against selected targets in molecular docking studies [22]. Bromfenac, particularly when conjugated with AuAgCu2O nanoparticles, has demonstrated antibacterial and anti-inflammatory properties against Staphylococcus aureus, suggesting potential efficacy against S. pneumoniae pending experimental validation [17].
Objective: To identify essential, non-host homologous proteins in S. pneumoniae as potential drug targets.
Materials:
Procedure:
Objective: To identify potential repurposing candidates against prioritized targets.
Materials:
Procedure:
Table 3: Essential Research Reagents and Computational Tools
| Tool/Resource | Category | Specific Application | Access Information |
|---|---|---|---|
| CD-HIT | Bioinformatics Tool | Sequence clustering and redundancy removal | https://github.com/weizhongli/cdhit |
| BLAST+ | Bioinformatics Tool | Sequence homology searches | https://ftp.ncbi.nlm.nih.gov/blast/executables/blast+/ |
| Database of Essential Genes (DEG) | Database | Essential gene identification | http://origin.tubic.org/deg/public/index.php |
| KEGG Pathway | Database | Metabolic pathway analysis and visualization | https://www.genome.jp/kegg/ |
| AutoDock Vina | Molecular Docking | Structure-based virtual screening | http://vina.scripps.edu/ |
| GROMACS | Molecular Dynamics | Simulation of biomolecular interactions | https://www.gromacs.org/ |
| ModelSEED | Metabolic Modeling | Reconstruction of genome-scale metabolic models | https://modelseed.org/ |
| Sometribove | Sometribove | Sometribove is a recombinant bovine somatotropin for research. This RUO product studies metabolic pathways and lactation biology; not for human or veterinary use. | Bench Chemicals |
| versutoxin | Versutoxin | Versutoxin is a site 3 neurotoxin that slows VGSC inactivation. This spider venom peptide is for research use only (RUO). Not for human consumption. | Bench Chemicals |
The reconstruction of metabolic networks in non-model organisms like S. pneumoniae provides critical insights for drug target identification. Genome-scale metabolic models (GSMMs) integrate genes, metabolic reactions, and metabolites to simulate metabolic flux distributions under specific conditions [24]. For Streptococci, these models have been valuable in linking metabolic regulation and pathogenicity [24].
The iNX525 model of Streptococcus suis, a related species, exemplifies this approach, containing 525 genes, 708 metabolites, and 818 reactions [24]. Similar principles can be applied to S. pneumoniae to systematically analyze metabolic genes associated with virulence factor formation and identify targets affecting both virulence and cell growth [24].
Figure 2. Key metabolic pathways and potential drug targets in S. pneumoniae. Essential enzymes identified through subtractive genomics (fba, gpi, trpS) are highlighted in red, showing their positions in central metabolism and connections to virulence factor production.
The application of subtractive genomics to S. pneumoniae has demonstrated considerable promise in identifying novel therapeutic targets. By systematically filtering the pathogen's proteome, this approach addresses the critical challenge of antibiotic resistance by focusing on essential pathogen-specific pathways [17] [20]. The identification of 21 high-priority targets, including key hub genes such as gpi, fba, rpoD, and trpS, provides a foundation for future drug development efforts [17].
The integration of multi-omics data represents the future of target identification in pathogenic bacteria. Combining genomic, metabolomic, and transcriptomic datasets can provide a more comprehensive understanding of pathogen vulnerability [18] [20]. As demonstrated in metabolomic studies of S. pneumoniae infections, the activation of specific metabolic pathways in response to infection provides additional layers of information for target prioritization [18]. Furthermore, the successful identification of Bromfenac as a repurposing candidate highlights the potential for accelerating therapeutic development through computational approaches [17] [22].
Future directions in this field should emphasize the experimental validation of computationally identified targets through in vitro and in vivo studies [20]. Additionally, the incorporation of artificial intelligence and machine learning approaches will enhance the predictive power of these analyses, enabling more accurate target prioritization and binding affinity predictions [20]. As genomic sequencing technologies continue to advance and become more accessible, subtractive genomics approaches will play an increasingly important role in addressing the global challenge of antimicrobial resistance.
Metabolic pathway reconstruction for non-model organisms is a fundamental challenge in systems biology and metabolic engineering. Without the extensive biochemical characterization available for model organisms, researchers must rely heavily on computational predictions derived from curated reference databases. The Kyoto Encyclopedia of Genes and Genomes (KEGG), BioCyc, and MetaCyc represent three essential knowledge bases that enable scientists to infer metabolic capabilities from genomic sequences. These databases employ different curation philosophies and provide complementary tools for pathway prediction, analysis, and visualization. Within the context of non-model organism research, understanding the relative strengths and applications of each resource is crucial for accurate metabolic reconstruction, which in turn drives discoveries in synthetic biology, drug target identification, and understanding of microbial ecology. This article provides a detailed comparison of these databases and protocols for their effective application in non-model organism studies.
Table 1: Comparative analysis of KEGG, MetaCyc, and BioCyc database content and scope.
| Feature | KEGG | MetaCyc | BioCyc Collection |
|---|---|---|---|
| Primary Focus | Integrated knowledge of biological systems, diseases, and drugs [25] | Reference database of experimentally elucidated metabolic pathways and enzymes [26] | Collection of >20,000 organism-specific Pathway/Genome Databases (PGDBs) [27] |
| Pathway Content | Manually drawn pathway maps (e.g., ko, ec) and modules [28] | 3,264 metabolic pathways (as of 2025) [29] | Varies by organism; includes computationally inferred and curated pathways [27] |
| Reaction Content | 8,692 reactions (2012 data) [30] | 20,039 reactions (as of 2025) [29] | Propagated from MetaCyc and organism-specific curation [31] |
| Compound Content | 16,586 compounds (2012 data) [30] | 20,490 compounds (as of 2025) [29] | Propagated from MetaCyc and organism-specific curation [31] |
| Curation Philosophy | Manual pathway maps with automated genome annotation | Heavy manual curation of individual pathways and reactions [30] | Tiered system (Tier 1: heavily curated, Tier 3: fully computational) [31] |
| Taxonomic Scope | Universal | 3,542 organisms (pathway sources) [29] | 20,080 organisms (as of 2025) [32] |
| Key Strengths | Broad biological scope including diseases and drugs; conserved orthologs (KOs) [28] [25] | High-quality curated metabolic data; supports metabolic engineering [30] [26] | Scalable platform for organism-specific metabolic reconstruction [27] [31] |
The databases employ fundamentally different conceptualizations of metabolic pathways. A systematic comparison found that KEGG pathways contain 3.3 times as many reactions on average as MetaCyc pathways, reflecting their more inclusive, "map"-like nature [30]. KEGG organizes its content into manually drawn "map" pathways and higher-level "module" pathways, whereas MetaCyc distinguishes between base pathways and super-pathways that combine multiple base pathways [30].
The curation scope also differs significantly. MetaCyc contains a broader set of database attributes than KEGG, including regulatory information, identification of spontaneous reactions, and the expected taxonomic range of metabolic pathways [30]. MetaCyc also includes more balanced reaction equations, facilitating metabolic modeling approaches such as flux-balance analysis [30]. Each database also contains unique pathway content: MetaCyc includes more pathways from plants, fungi, metazoa, and actinobacteria, while KEGG contains more pathways for xenobiotic degradation, glycan metabolism, and metabolism of terpenoids and polyketides [30].
Purpose: To generate an organism-specific Pathway/Genome Database (PGDB) from genomic annotation using the PathoLogic component of Pathway Tools.
Applications: Creation of draft metabolic networks for non-model organisms with sequenced genomes, enabling subsequent analysis and curation [26] [31].
Table 2: Key research reagents and computational tools for pathway reconstruction.
| Research Reagent / Software | Function in Protocol | Access / Requirements |
|---|---|---|
| Pathway Tools Software | Primary software suite for creating, curating, and analyzing PGDBs [33] [34] | Free academic license; runs on Mac, Windows, Linux [33] |
| Annotated Genome File | Input data containing predicted genes and functional assignments (e.g., EC numbers) [31] | Typically in GenBank format or similar |
| MetaCyc Reference DB | Reference metabolic pathway database used for inference [26] | Included with Pathway Tools [33] |
| BioCyc Data Files | Optional comparative data for related organisms [33] | Requires subscription (except EcoCyc) [33] |
Methodology:
Input Preparation: Obtain the completely sequenced and annotated genome of the target non-model organism in a supported format (e.g., GenBank format). Ensure gene annotations include Enzyme Commission (EC) numbers where possible, as these are primary inputs for pathway prediction [31].
PathoLogic Execution:
Output and Validation: The output is a new PGDB containing:
Figure 1: Workflow for *de novo pathway prediction with PathoLogic.*
Purpose: To contextualize metabolomics datasets within the predicted metabolic network of a non-model organism to identify actively used pathways and potential bottlenecks.
Applications: Interpretation of high-throughput metabolomics data; identification of pathway activation under different growth conditions; target identification for metabolic engineering [26].
Methodology:
Data Preparation: Prepare metabolomics data as a tab-delimited file where rows represent metabolites and columns represent experimental conditions or time points. Metabolites should be identified using standard identifiers (e.g., MetaCyc compound IDs, KEGG compound IDs, or standard chemical names) to facilitate mapping.
Data Import and Mapping:
Visualization and Analysis:
Figure 2: Workflow for metabolomics data analysis using a PGDB.
Purpose: To identify conserved and unique metabolic capabilities across multiple non-model organisms by comparing their PGDBs.
Applications: Pan-genome metabolic analysis; identification of taxonomic markers; guiding experimental design by highlighting core and accessory metabolism.
Methodology:
Dataset Establishment: Generate PGDBs for multiple related non-model organisms using Protocol 1. Alternatively, select existing PGDBs from the BioCyc collection for organisms of interest [27].
Comparative Analysis Execution:
Orthology-Based Cross-Referencing with KEGG:
Table 3: Key databases, software, and tools for metabolic pathway research.
| Tool / Resource Name | Type | Primary Function in Research | Access |
|---|---|---|---|
| Pathway Tools [33] [34] | Software Suite | Create, edit, analyze, and visualize PGDBs; predict pathways; omics data analysis. | Free academic license |
| KEGG Mapper [25] | Web Tool Suite | Map user data (genes, compounds) onto KEGG pathway maps and BRITE hierarchies. | Subscription/paid |
| MetaCyc [29] [26] | Reference Database | Curated reference for pathway prediction and enzyme information; educational resource. | Free |
| BioCyc Collection [27] | Database Collection | Access thousands of pre-computed PGDBs for comparative analysis. | Subscription (partial free) |
| KEGG Orthology (KO) [25] | Classification System | Standardized annotation of gene functions for pathway mapping across species. | Subscription/paid |
| SmartTables [27] [34] | Analysis Tool | Create, share, and analyze sets of genes, compounds, etc.; perform enrichment analysis. | Via BioCyc/Pathway Tools |
| BlastKOALA [25] | Annotation Service | Automated KEGG Orthology assignment and pathway mapping for nucleotide/protein sequences. | Web service |
The integration of KEGG, BioCyc, and MetaCyc provides a powerful, multi-faceted framework for tackling the complex challenge of metabolic pathway reconstruction in non-model organisms. While KEGG offers a broad, systems-level view integrated with disease and drug data, MetaCyc provides deep, experimentally-validated metabolic information crucial for accurate prediction, and the BioCyc collection enables scalable, organism-specific reconstruction and comparison. The ongoing curation and expansion of these resourcesâevidenced by MetaCyc's addition of 41 new pathways in its latest releaseâensure they remain at the forefront of biological discovery [29]. For researchers investigating non-model organisms, a strategic approach that leverages the complementary strengths of these databases, combined with the experimental protocols outlined herein, will significantly accelerate the elucidation of metabolic networks, thereby enabling advances in fields ranging from synthetic biology to drug discovery.
Metabolic pathway reconstruction is a foundational step in systems biology, enabling researchers to decipher the biochemical capabilities of an organism from its genomic sequence. For researchers working with non-model organismsâspecies not represented in standard reference databasesâthis process presents a significant challenge. The choice of computational strategy, primarily between reference-based (alignment) and de novo approaches, directly influences the accuracy, completeness, and biological relevance of the resulting metabolic models [35] [36]. Reference-based methods offer efficiency but can overlook novel biology, whereas de novo methods promise discovery at the cost of greater computational complexity. This application note delineates these strategies, provides quantitative performance comparisons, and outlines detailed protocols for their application, specifically within the context of non-model organism research.
The two primary strategies for metabolic pathway prediction differ fundamentally in their philosophy and implementation. Reference-based (or alignment-based) prediction relies on mapping sequencing reads or gene calls to pre-existing databases of known genes, pathways, and genomes. In contrast, de novo prediction reconstructs metabolic pathways directly from sequencing data without relying on reference genomes, often through the assembly of reads into contigs and the subsequent annotation of metagenome-assembled genomes (MAGs) [35].
A recent large-scale comparison of these methods using human gut microbiota data revealed critical differences in their outputs (Table 1) [35].
Table 1: Quantitative Comparison of Reference-Based and De Novo Approaches for Microbiome Analysis
| Performance Metric | Reference-Based (AL) | De Novo (DN) |
|---|---|---|
| Statistical Power | Higher; identified a larger number of statistically significant taxa associated with BMI [35] | Lower; produced a subset of the significant findings from AL [35] |
| Result Sparsity | Lower sparsity of the result matrix [35] | Higher sparsity of the result matrix [35] |
| Sensitivity to Host Factors | Higher explained variance (~8.7%) in PERMANOVA analysis [35] | Lower explained variance in PERMANOVA analysis [35] |
| Archaeal Detection | ~0.4% relative abundance [35] | ~0.9% relative abundance [35] |
| Key Strength | Efficiency and sensitivity for profiling known biology [35] | Discovery of novel taxa, genes, and genomic regions [35] |
| Primary Limitation | Reference database bias; may miss novel elements [35] | High computational resource requirements; expertise needed [35] |
The strategic choice between these methods hinges on the research goal. Reference-based methods are optimal for well-characterized communities or when resources are limited, while de novo approaches are indispensable for exploring true novelty and for generating robust, population-specific genomic resources that serve as a foundation for metabolic reconstruction [35] [36].
Beyond reconstructing existing pathways, a transformative new approach called semantic design now enables the de novo generation of novel functional genetic elements. This method uses a genomic language model, Evo, which learns the "distributional semantics" of gene functionâthe principle that a gene's function can be inferred from the functional context of its genomic neighbors [37].
The model is trained on prokaryotic genomes to perform a genomic "autocomplete." When prompted with a DNA sequence encoding a function of interest (e.g., a toxin gene), the model generates novel, functionally related sequences (e.g., its cognate antitoxin) [37]. This process has been experimentally validated to design functional anti-CRISPR proteins and toxin-antitoxin systems, including proteins with no significant sequence similarity to any known natural protein [37]. This approach is particularly powerful for non-model organisms where characterized genetic parts are scarce, as it allows for the computational design of custom, functional genetic systems from first principles.
gapseq is a tool that provides informed prediction of bacterial metabolic pathways and reconstructs accurate metabolic models. It combines homology searching with a curated reaction database and a novel gap-filling algorithm [8].
Step 1: Software Installation
Step 2: Database Curation gapseq uses a manually curated database derived from ModelSEED biochemistry, comprising 15,150 reactions and 8,446 metabolites. The tool automatically checks for updates to its reference protein sequences from UniProt and TCDB upon execution [8].
Step 3: Pathway Prediction Run the main gapseq pipeline using a genome assembly in FASTA format.
The find command identifies pathways based on sequence homology to a database of 131,207 unique reference sequences [8].
Step 4: Model Reconstruction and Gap-Filling
This step uses a Linear Programming (LP)-based algorithm to resolve network gaps, enabling biomass formation on a specified growth medium. The algorithm also fills gaps for functions supported by sequence homology, reducing medium-specific bias and increasing model versatility [8].
Validation: gapseq has been validated against 14,931 bacterial phenotypes, showing a 53% true positive rate for enzyme activity prediction, outperforming other tools like CarveMe (27%) and ModelSEED (30%) [8].
This protocol outlines the process for reconstructing metabolic pathways directly from metagenomic sequencing reads, culminating in metabolic models for MAGs.
Step 1: Quality Control and Assembly
Step 2: Binning and Metagenome-Assembled Genome (MAG) Curation
Check MAG quality (completeness and contamination) with tools like CheckM.
Step 3: Functional Annotation and Pathway Prediction Annotate the high-quality MAGs using a tool like gapseq, following Protocol 1, but using the MAG as the input genome. This leverages the strength of de novo discovery (MAGs) with the powerful pathway prediction of a reference-based tool [35].
Step 4: Community Metabolic Modeling Reconstruct metabolic models for each MAG and build a community model. The APOLLO resource, for instance, has demonstrated the construction of 14,451 sample-specific microbiome community models to interrogate community-level metabolic capabilities, which can be stratified by body site, age, and disease state [38].
The following diagram illustrates the logical workflow for choosing and applying the appropriate computational strategy for metabolic pathway reconstruction in non-model organisms.
Table 2: Key Computational Tools and Databases for Metabolic Reconstruction
| Tool / Resource | Type | Primary Function | Application Note |
|---|---|---|---|
| gapseq [8] | Software Pipeline | Automated metabolic pathway prediction and model reconstruction from a genome. | Uses a curated reaction database and a novel LP-based gap-filling algorithm. Outperforms others in carbon source utilization prediction. |
| Evo Model [37] | Genomic Language Model | De novo generation of functional genes and systems via semantic design. | Leverages genomic context (e.g., operon structure) to generate novel sequences for targeted functions like anti-CRISPRs. |
| APOLLO Resource [38] | Metabolic Model Database | A resource of 247,092 genome-scale metabolic reconstructions for human microbes. | Enables systems-level modeling of personalized host-microbiome co-metabolism across body sites, ages, and geographies. |
| MetaPhlAn4 [35] | Alignment-based Profiler | Taxonomic profiling of metagenomic samples. | Rapidly maps reads to a database of clade-specific marker genes for efficient community composition analysis. |
| HUMAnN3 [35] | Alignment-based Profiler | Profiling of metabolic pathways in metagenomes. | Quantifies abundance of microbial pathways by mapping reads to a curated database of protein families and metabolic modules. |
| UniProt/TCDB [8] | Protein/Transporter Database | Curated source of protein sequences and transporter classifications. | Forms the core reference database for tools like gapseq to identify homologous genes and predict metabolic functions. |
Genome-scale metabolic models (GEMs) are computational representations of the entire metabolic network of an organism, constructed from its annotated genome sequence [39]. These models mathematically describe the gene-protein-reaction (GPR) associations for all metabolic genes, enabling researchers to simulate metabolic fluxes and predict phenotypic behaviors under various genetic and environmental conditions [40]. The fundamental component of a GEM is the stoichiometric matrix (S matrix), where columns represent reactions, rows represent metabolites, and entries correspond to stoichiometric coefficients [39]. GEMs have become indispensable tools in systems biology and metabolic engineering, particularly through the application of flux balance analysis (FBA), which uses linear programming to predict optimal flux distributions through metabolic networks under steady-state assumptions [39] [41].
The reconstruction of high-quality GEMs for non-model organisms presents both challenges and significant opportunities. While model organisms like Escherichia coli and Saccharomyces cerevisiae have well-established, iteratively refined GEMs, non-model organisms often possess unique metabolic capabilities that make them valuable industrial chassis but lack the comprehensive biological data needed for straightforward model reconstruction [42] [43]. The bacterium Zymomonas mobilis exemplifies this scenarioâit exhibits extraordinary industrial characteristics including high sugar uptake rate, high ethanol yield, and exceptional ethanol tolerance, making it a promising platform for biomanufacturing [42] [43]. However, its development as a biorefinery chassis has been hampered by its dominant ethanol production pathway, which restricts the titer and rate of other valuable biochemicals [42]. This review examines the construction and application of two successive GEMs for Z. mobilisâiZM516 and its enzyme-constrained successor eciZM547âas paradigmatic cases for metabolic pathway reconstruction in non-model organisms.
The iZM516 model was developed to address limitations in existing Z. mobilis GEMs, which suffered from issues such as incorrect ATP generation, missing plasmid gene information, and lack of standard format files [43]. This comprehensive model contains 516 genes, 1,389 reactions, 1,437 metabolites, and 3 cell compartments, achieving the highest MEMOTE evaluation score (91%) among all published Z. mobilis models at the time of its publication [43]. The reconstruction process integrated improved genomic annotation including native plasmid information, experimental data from Biolog Phenotype Microarray studies, and manually curated Gene-Protein-Reaction relationships from multiple databases [43].
A critical advancement in iZM516 was the proper representation of Z. mobilis's unique metabolic characteristics, particularly its utilization of the Entner-Doudoroff (ED) pathway under anaerobic conditionsâa rare capability among known microorganisms [42] [43]. The model accurately simulates the ATP yield from glucose metabolism, correctly representing the production of 1 mol ATP per 1 mol glucose under anaerobic conditions, unlike previous models that generated biologically implausible amounts [43]. When validated against experimental substrate utilization data, iZM516 demonstrated 79.4% accuracy in predicting cell growth, establishing it as a reliable platform for metabolic engineering design [43].
Table 1: Key Characteristics of iZM516 and eciZM547
| Feature | iZM516 | eciZM547 |
|---|---|---|
| Genes | 516 | 547 |
| Reactions | 1,389 | 1,455 |
| Metabolites | 1,437 | 1,455 |
| Compartments | 3 | 3 |
| Constraints Type | Stoichiometric | Enzyme-constrained |
| MEMOTE Score | 91% | Not specified |
| Key Application | Succinate and 1,4-BDO pathway design | D-lactate production via DMCI strategy |
The iZM516 model was subsequently upgraded to eciZM547 through the integration of enzyme constraints that reflect limitations related to protein resources during cell growth [42]. This enzyme-constrained model (ecModel) was developed using ECMpy2 and Kcat values provided by AutoPACMEN, which was determined to be more accurate than other methods such as DLkcat, TurNup, and UniKP [42]. The resulting eciZM547AutoPACMENmean (abbreviated as eciZM547) contains 547 genes, 1,455 metabolites, and represents the enzyme-constrained metabolic network model closest to experimental results [42].
The integration of enzyme constraints fundamentally improved the predictive capabilities of the model. Most notably, eciZM547 revealed a shift from glucose-limited growth to proteome-limited growth when glucose uptake exceeded approximately 71 mmol·gDWâ»Â¹Â·hâ»Â¹ [42]. This constrained simulation predicted a maximum growth rate of 0.50 hâ»Â¹ and a maximum ethanol production rate of 134.76 mmol·gDWâ»Â¹Â·hâ»Â¹, representing more biologically realistic values than the previous model, which highly overestimated these parameters [42]. Additionally, while iZM516 predicted that most carbon sources would be directed toward acetate based on growth criteria when glucose was the sole carbon source, eciZM547 more accurately simulated carbon flux into both acetate and acetoin, aligning with experimental ¹³C-metabolic flux analysis (MFA) data [42].
Diagram 1: GEM reconstruction workflow from iZM516 to eciZM547
The reconstruction of a high-quality GEM for a non-model organism like Z. mobilis requires systematic curation and integration of diverse data sources. The following protocol outlines the key steps employed in developing iZM516:
Draft Reconstruction: Utilize the latest genomic information from NCBI (chromosome: NZ_CP023715.1, plasmids: pZM32, pZM33, pZM36, pZM39) with the Rapid Annotation using Subsystem Technology (RAST) server and the ModelSEED database to automatically generate a draft model [43].
Annotation and ID Conversion: Convert temporary gene IDs from RAST to specific IDs and names of Z. mobilis ZM4 using BLASTp with thresholds set at e-value â¤10â»âµ and identity â¥40% [43].
Biomass Equation Curation: Define biomass composition to include DNA, RNA, proteins, lipids, peptidoglycan, carbohydrates, and small molecules. For Z. mobilis, specifically incorporate the hopane biosynthesis pathway as this is an important membrane component contributing to ethanol tolerance [43].
Manual Curation and Gap Filling: Identify biomass precursors that cannot be synthesized and employ a weight-added pFBA algorithm for gap filling. Set reactions in the draft model with a weight of 1000 and the upper limit of the biomass equation to 0.1 to minimize the number of filling reactions introduced from the ModelSEED database [43].
Validation with Experimental Data: Test the model's predictive accuracy against experimental Biolog Phenotype Microarray results for substrate utilization, with iZM516 achieving 79.4% agreement with experimental growth results [43].
Quality Assessment: Evaluate the model using the standard genome-scale metabolic model test suite MEMOTE, with iZM516 achieving a score of 91% [43].
The transformation of a stoichiometric GEM to an enzyme-constrained model enhances its predictive accuracy by accounting for proteome limitations:
Model Enhancement: Begin with the iZM516 model and incorporate unique genes and reactions from complementary models like iZM4_478 through manual curation to create an enhanced stoichiometric model (iZM547) [42].
Enzyme Constraint Integration: Apply the ECMpy2 computational pipeline to integrate enzyme constraints using Kcat values from the AutoPACMEN tool, which demonstrates superior accuracy compared to alternative methods [42].
Proteome Allocation Modeling: Implement constraints that reflect the trade-off between biomass yield and enzyme usage efficiency, capturing the shift from substrate-limited to proteome-limited growth [42].
Validation with ¹³C-MFA: Compare model predictions with experimental ¹³C-metabolic flux analysis data under relevant conditions (e.g., aerobic growth) to verify accurate prediction of carbon flux distributions [42].
Simulation of Metabolic Phenotypes: Utilize the constrained model to simulate overflow metabolism and identify rate-limiting enzymes in engineered strains [42].
Table 2: Research Reagent Solutions for GEM Reconstruction
| Reagent/Resource | Type | Function in GEM Reconstruction |
|---|---|---|
| RAST Server | Online Tool | Automated genome annotation and draft model generation |
| ModelSEED Database | Database | Biochemical database for reaction and metabolite information |
| MEMOTE Suite | Software | Quality assessment and validation of model structure |
| Biolog Phenotype Microarray | Experimental Assay | Validation of model predictions against experimental growth data |
| COBRA Toolbox | Software Package | MATLAB-based tools for constraint-based reconstruction and analysis |
| ECMpy2 | Computational Pipeline | Integration of enzyme constraints into stoichiometric models |
| AutoPACMEN | Algorithm | Prediction of enzyme Kcat values for constraint implementation |
| MetaCyc Database | Database | Curated database of metabolic pathways and enzymes |
The iZM516 model has served as a powerful computational platform for designing metabolic engineering strategies in Z. mobilis. Through in silico simulations under anaerobic conditions, researchers used iZM516 to design pathways for producing valuable chemicals including succinate and 1,4-butanediol (1,4-BDO) [43]. The model predicted that combinatorial metabolic engineering strategies could achieve yields of 1.68 mol/mol succinate and 1.07 mol/mol 1,4-BDO from glucose, comparable to the performance of established model species like E. coli [43]. These predictions demonstrated the potential of Z. mobilis as a chassis for producing chemicals beyond its native ethanol production.
Additionally, iZM516 enabled the identification of potential endogenous succinate synthesis pathways in Z. mobilis ZM4, providing insights into the native metabolic capabilities of this non-model organism [43]. The model was also used to design and simulate metabolic pathways for various other biochemicals, including 1,3-propanediol (1,3-PDO) from glycerol, butanediol from glucose, xylonic acid, ethylene glycol, glycolic acid, and 1,4-butanediol from xylose [42]. This versatility highlights how high-quality GEMs can expand the biotechnological application range of non-model organisms.
A groundbreaking application enabled by the eciZM547 model was the development of a dominant-metabolism compromised intermediate-chassis (DMCI) strategy to bypass Z. mobilis's innate dominant ethanol production pathway [42]. This approach involved introducing a low-toxicity but cofactor-imbalanced 2,3-butanediol (2,3-BDO) pathway to create an intermediate chassis, rather than directly engineering the chassis for target biochemicals [42]. The compromised chassis could then be more effectively redirected toward high-yield production of target compounds.
This DMCI strategy, guided by predictions from eciZM547, led to the construction of a recombinant D-lactate producer capable of producing more than 140.92 g/L from glucose and 104.6 g/L from corncob residue hydrolysate, with a remarkable yield exceeding 0.97 g/g glucose [42]. Techno-economic analysis (TEA) and life cycle assessment (LCA) further demonstrated the commercial feasibility and greenhouse gas reduction capability of producing D-lactate from lignocellulosic waste, validating the industrial relevance of this model-guided approach [42].
Diagram 2: DMCI strategy for D-lactate production
The development and refinement of genome-scale metabolic models from iZM516 to eciZM547 exemplify the critical role of computational modeling in advancing metabolic engineering of non-model organisms. The iterative enhancement of these modelsâfrom a high-quality stoichiometric foundation to an enzyme-constrained framework capable of predicting proteome-limited growthâdemonstrates how GEMs can evolve to incorporate increasing layers of biological complexity. The successful application of these models to guide metabolic engineering strategies, particularly the innovative DMCI approach for bypassing native regulatory networks, highlights the transformative potential of GEMs in enabling non-model organisms like Z. mobilis to serve as efficient biorefinery chassis for sustainable biochemical production.
Future developments in GEM reconstruction for non-model organisms will likely focus on integrating additional cellular constraints beyond metabolism, including transcriptional regulation, signaling networks, and resource allocation across cellular processes. The integration of machine learning approaches with GEMs, as well as the development of multi-strain and community-level models, will further expand the predictive capabilities and application scope of these computational frameworks [41]. As these tools continue to evolve, they will accelerate the design-build-test-learn cycle in synthetic biology, enabling more efficient engineering of non-model organisms for circular bioeconomy applications. The iZM516 and eciZM547 models for Z. mobilis thus represent both practical tools for metabolic engineers and paradigmatic cases for GEM development in industrially relevant but genetically recalcitrant microorganisms.
The pursuit of sustainable biomanufacturing has catalyzed the exploration of non-model microorganisms as next-generation cellular factories. Unlike their model counterparts, these organisms possess unique and versatile metabolic characteristics, enabling them to thrive on diverse feedstocks, tolerate extreme fermentation conditions, and synthesize novel high-value compounds [44]. However, the full potential of these microbial chassis has been historically locked behind a significant challenge: the lack of efficient genetic tools for precise pathway engineering. The advent of CRISPR-Cas systems has begun to dismantle this barrier, offering a versatile and powerful platform for domesticating non-model bacteria and fungi. This document details specialized application notes and protocols, framed within the broader thesis of metabolic pathway reconstruction, to equip researchers with the methodologies needed to harness non-model organisms for applied biotechnology and drug development.
CRISPR-Cas systems function by utilizing a guide RNA (gRNA) to direct a Cas nuclease to a specific DNA sequence, resulting in a double-strand break (DSB). The cellular repair of this break is then leveraged for genetic edits [45] [46]. The two primary repair pathways are:
A critical consideration for pathway engineering in non-model bacteria and fungi is that NHEJ is often the dominant repair pathway, which can hinder the precise gene integrations required for metabolic engineering [45] [47]. Furthermore, challenges such as low transformation efficiency, the presence of tough cell walls, and the scarcity of species-specific genetic parts like promoters further complicate editing efforts [44] [48]. The protocols that follow are designed to address these specific hurdles.
This protocol is adapted from methodologies successfully applied in Aspergillus and other filamentous fungi for the precise integration of metabolic pathway genes [45] [44].
1. Goal: To integrate a heterologous gene expression cassette into a specific genomic locus of a filamentous fungus.
2. Experimental Workflow:
The following diagram illustrates the key steps for achieving precise gene integration, from design to analysis.
3. Key Reagents and Materials:
4. Detailed Methodology:
Step 1: Design and Synthesis.
Step 2: Vector Construction.
Step 3: Fungal Transformation.
Step 4: Screening and Validation.
This protocol leverages CRISPR-based counterselection to enable scarless, marker-free engineering in bacteria where homologous recombination is inefficient, such as Clostridium and Rhodococcus [44].
1. Goal: To simultaneously knock out multiple genes in a non-model bacterium without leaving selectable markers in the genome.
2. Experimental Workflow:
3. Key Reagents and Materials:
4. Detailed Methodology:
Step 1: Design and Construction.
Step 2: Transformation and Induction.
Step 3: Screening and Validation.
For essential genes whose knockout would be lethal, or for fine-tuning metabolic flux, CRISPRi offers a powerful alternative [44] [48].
1. Goal: To reversibly repress gene expression in non-model bacteria using a catalytically dead Cas9 (dCas9).
2. Experimental Workflow:
3. Key Reagents and Materials:
4. Detailed Methodology:
Step 1: System Design.
Step 2: Transformation and Cultivation.
The table below catalogs key reagents and their critical functions for CRISPR-based metabolic engineering in non-model systems.
Table 1: Essential Research Reagents for CRISPR Pathway Engineering
| Reagent / Solution | Function / Application | Examples & Notes |
|---|---|---|
| Cas9 Nuclease | Creates DSBs for gene knockout or HDR-mediated knock-in. | Use species-specific codon optimization. High-fidelity variants (e.g., SpCas9-HF1) reduce off-target effects [48]. |
| dCas9 (deactivated Cas9) | Serves as a programmable DNA-binding scaffold for CRISPRi/a without cleaving DNA [48]. | Fused to transcriptional repressors (e.g., KRAB) for CRISPRi. |
| Guide RNA (gRNA) | Directs Cas/dCas protein to the specific target DNA sequence via Watson-Crick base pairing. | Can be expressed from a U6 or tRNA promoter. Multiplexed sgRNA arrays enable simultaneous targeting of multiple genes [47] [48]. |
| Donor DNA Template | Serves as a repair template for HDR to enable precise gene insertion or correction. | For fungi, use long homology arms (500-1000 bp). For bacteria, shorter arms may suffice. Can be supplied as a linear dsDNA fragment or circular plasmid [45] [47]. |
| Delivery Vectors | Plasmid-based systems for delivering Cas and gRNA genes into the host. | Include species-specific origins of replication and selectable markers (e.g., antibiotic resistance). All-in-one vectors are preferred [47] [46]. |
| Ribonucleoprotein (RNP) | Pre-complexed Cas9 protein and gRNA. | Direct delivery of RNPs into protoplasts avoids the need for endogenous transcription and can reduce off-target effects and toxicity [46]. |
| Protoplasting Solution | Enzyme mixture to digest the fungal cell wall to create protoplasts for transformation. | Contains lytic enzymes like glucanases and chitinases [46]. |
| Polyethylene Glycol (PEG) | Facilitates the uptake of DNA or RNPs into fungal protoplasts during transformation. | A critical component of PEG-mediated transformation protocols [46]. |
| vitaferro | vitaferro, CAS:115774-15-7, MF:C41H42N4O6 | Chemical Reagent |
| Isobornyl acrylate | Isobornyl acrylate, CAS:111821-21-7, MF:C7H6N2O | Chemical Reagent |
Editing efficiency varies significantly between organisms and protocols. The table below summarizes reported efficiencies to aid in experimental planning.
Table 2: Reported CRISPR Editing Efficiencies in Non-Model Microorganisms
| Organism Group | Species Example | Editing Tool | Edit Type | Reported Efficiency | Key Factors Influencing Efficiency |
|---|---|---|---|---|---|
| Filamentous Fungi | Aspergillus nidulans | CRISPR-Cas9 | Gene Knockout | High (60-100%) [45] | sgRNA design, promoter strength for Cas9/gRNA, NHEJ/HDR balance [45]. |
| Oleaginous Yeasts | Yarrowia lipolytica | CRISPR-Cas9 | Gene Knock-In | Varies (1-20% for HDR) [47] [44] | Length of homology arms, donor DNA concentration/form, suppression of NHEJ [47]. |
| Non-Model Bacteria | Clostridium spp. | CRISPR-Cas9 | Multiplexed Knockout | Achieved in several studies [44] | Efficiency of NHEJ pathway, transformation method, inducible Cas9 expression to avoid toxicity [44]. |
| Cyanobacteria | Synechococcus spp. | CRISPR-Cpf1/Cas12a | Gene Knockout | Efficient editing demonstrated [44] | Choice of Cas nuclease (Cas12a can be more efficient than Cas9 in some strains), PAM availability [44]. |
The CRISPR toolkit has evolved from a simple DNA-cleaving apparatus into a versatile synthetic biology "Swiss Army Knife," enabling researchers to move beyond simple gene knockouts [48]. By applying the detailed protocols and application notes outlined in this documentâranging from precise gene knock-in and multiplexed editing to tunable transcriptional regulationâscientists can systematically overcome the genetic recalcitrance of non-model bacteria and fungi. The continued refinement of these tools, including the adoption of base editors and prime editors for single-nucleotide precision, promises to further accelerate the development of robust microbial cell factories for the sustainable production of drugs, chemicals, and fuels.
The engineering of non-model microorganisms presents a significant opportunity for biotechnology, as these organisms often possess innate, desirable industrial characteristics such as robust stress tolerance and unique metabolic capabilities [3] [2]. However, a central challenge in harnessing these chassis is their frequent possession of a dominant, native metabolic pathway that fiercely competes for central carbon precursors, severely limiting the yield and titer of desired engineered products [3]. The Dominant-Metabolism Compromised Intermediate-Chassis (DMCI) strategy is a novel metabolic engineering approach designed to overcome this fundamental limitation. Instead of directly engineering a target pathway into a wild-type host, the DMCI approach involves first constructing an intermediate chassis where the dominant native metabolism is intentionally compromised by introducing a less toxic, cofactor-imbalanced pathway. This intermediate step effectively "liberates" carbon flux from the dominant pathway, creating a metabolically primed host that is more amenable to the subsequent installation of high-yield production pathways for a wide range of biochemicals [3].
The successful implementation of the DMCI strategy follows a sequence of key stages, integrating computational design, genetic engineering, and fermentation. The overall workflow is depicted in Figure 1.
Objective: To create a high-quality, organism-specific GEM that can accurately simulate metabolic flux and guide pathway design [3] [49].
Protocol:
13C-Metabolic Flux Analysis (13C-MFA) to empirically determine intracellular flux distributions and further validate model predictions [3].Objective: To computationally identify and validate a suitable pathway that effectively diverts carbon from the dominant metabolism without being toxic.
Protocol:
Objective: To genetically engineer the wild-type organism into the intermediate chassis by installing the compromising pathway.
Protocol:
Objective: To engineer the intermediate chassis for high-level production of the target biochemical.
Protocol:
Objective: To evaluate the commercial feasibility of the process.
Protocol:
Figure 1. The DMCI Strategy Workflow. This diagram outlines the key stages in implementing the DMCI strategy, from initial computational modeling to the final high-yield production strain.
The application of the DMCI strategy in the non-model bacterium Zymomonas mobilis for D-lactate production demonstrates its efficacy. The quantitative outcomes are summarized in Table 1.
Table 1: Performance Metrics of the DMCI Strategy for D-Lactate Production in Zymomonas mobilis [3]
| Performance Metric | Wild-Type Chassis (Direct Engineering) | DMCI Chassis | Improvement Factor |
|---|---|---|---|
| D-lactate Titer (g/L) | Not reported / Low | >140.92 g/L (Glucose)>104.6 g/L (Corncob Hydrolysate) | Significant |
| D-lactate Yield (g/g glucose) | Not reported / Low | >0.97 g/g | Significant |
| Ethanol Titer (g/L) | High (Dominant product) | Drastically Reduced | N/A |
| Maximum Growth Rate (hâ»Â¹) | Data from model | ~0.50 hâ»Â¹ (Predicted by eciZM547) | More accurate prediction |
| Ethanol Production Rate (mmol·gDWâ»Â¹Â·hâ»Â¹) | Data from model | ~134.76 (Predicted by eciZM547) | More accurate prediction |
The table shows that the DMCI strategy enabled a dramatic increase in D-lactate production, achieving a near-theoretical yield from glucose. Furthermore, the use of an enzyme-constrained model provided more accurate simulations of microbial growth and metabolism compared to previous models [3].
The core of the DMCI strategy involves a fundamental rewiring of central carbon metabolism. Figure 2 illustrates the key metabolic shifts achieved in the case of engineering Zymomonas mobilis.
Figure 2. Metabolic Flux Re-direction using the DMCI Strategy. The model shows the transition from a native state with a dominant ethanol pathway to a DMCI state where carbon flux is diverted through a compromising 2,3-BDO pathway, enabling high-yield D-lactate production. Abbreviations: ED Pathway (Entner-Doudoroff Pathway); PDC (Pyruvate Decarboxylase); ADH (Alcohol Dehydrogenase); als (Acetolactate Synthase); aldc (Acetolactate Decarboxylase).
Table 2: Key Research Reagent Solutions for Implementing the DMCI Strategy
| Item | Function / Application in DMCI Protocol | Specific Examples / Notes |
|---|---|---|
| Genome-Scale Modeling Software | Platform for constraint-based modeling, simulation, and in silico strain design. | COBRA Toolbox (MATLAB), COBRApy (Python), RAVEN, ModelSEED [49]. |
| Enzyme Kinetics Database | Provides k~cat~ values for integrating enzyme constraints into GEMs, improving predictive accuracy. | AutoPACMEN, DLKcat, SABIO-RK [3]. |
| CRISPR Genome Editing System | Enables precise, marker-free integration of pathway genes into the host chromosome. | CRISPR-Cas12a, Endogenous Type I-F CRISPR-Cas systems [3] [2]. |
| Serine Recombinase Toolkit | Facilitates high-efficiency, site-specific integration of DNA in non-model and undomesticated bacteria. | A versatile tool for organisms where CRISPR tools are not yet optimized [51]. |
| Synthetic Biological Parts | Controls the expression level of pathway genes for balancing metabolic flux. | Strong constitutive promoters, RBS libraries, inducible promoters (e.g., Ptet) [3] [51]. |
| Analytical Chromatography | Quantifies substrate consumption, product formation (e.g., D-lactate, 2,3-BDO, ethanol), and by-products. | HPLC, GC-MS. Essential for validating model predictions and strain performance [3] [50]. |
| 13C-Labeled Substrates | Used with 13C-MFA to empirically determine intracellular metabolic fluxes for model validation. | e.g., [1-13C]-Glucose, [U-13C]-Glucose [3]. |
| Non-Food Feedstock Hydrolysate | Validates the industrial relevance of the engineered strain using low-cost, sustainable carbon sources. | Corncob residue hydrolysate (CRH), lignin hydrolysates [3]. |
| Avanel S 150 | Avanel S 150|Sodium C12-15 Pareth-15 Sulfonate Supplier | Avanel S 150 is a mild, biodegradable anionic surfactant for research. It offers high foaming, emulsifying, and hydrolytic stability. For Research Use Only (RUO). Not for personal use. |
| CM SEPHAROSE | CM Sepharose Fast Flow Cation Exchange Media | CM Sepharose is a weak cation exchanger for preparative protein separation and purification. For Research Use Only. Not for human use. |
The BioCyc collection represents a comprehensive resource for encyclopedic reference, integrating genome data with metabolic reconstructions, regulatory networks, and protein features [27] [52]. It comprises 20,077 Pathway/Genome Databases (PGDBs) as of its 2025 release, providing organism-specific knowledge for model eukaryotes and thousands of microbes [32]. The platform is powered by the Pathway Tools software, an integrated bioinformatics suite that supports metabolic reconstruction, pathway prediction, and multi-omics data analysis [53]. For researchers investigating non-model organisms, BioCyc offers an indispensable framework for generating testable metabolic hypotheses from genomic sequences and interpreting high-throughput experimental data within a biochemical context [52] [31].
The BioCyc database collection is organized into a three-tiered system based on curation level, with Tier 1 databases (e.g., EcoCyc, MetaCyc) receiving the most extensive manual curation (>20 person-years for EcoCyc), Tier 2 undergoing limited curation (<1 person-year), and Tier 3 being entirely computational predictions [52] [31]. This hierarchical structure enables researchers to select the appropriate resource based on their needs for accuracy versus coverage, with Tier 2 and Tier 3 databases being particularly valuable for non-model organisms where curated knowledge is limited.
Table 1: BioCyc Database Collection Growth Over Time
| Year | Number of Genomes | Notable Additions |
|---|---|---|
| 2005 | 376 | Initial collection |
| 2016 | 9,387 | Steady expansion |
| 2021 | 18,030 | Major growth period |
| 2023 | 20,043 | |
| 2025 | 20,077 | Vibrio natriegens, Nostoc/Anabaena sp. PCC 7120 |
Each Pathway/Genome Database (PGDB) within the BioCyc collection describes the complete genome of an organism (chromosomes, genes, sequences), the products of each gene, the metabolic network (pathways, reactions, enzymes, metabolites), and when available, the regulatory network (operons, transcription factors, regulatory interactions) [52]. This integrated architecture allows researchers to traverse seamlessly from genetic elements to their functional manifestations in cellular biochemistry.
The MetaCyc database serves as the foundational reference for metabolic pathways and enzymes across all domains of life, with information curated from more than 76,000 publications [52] [54]. As a Tier 1 database, MetaCyc provides the curated pathway templates used by the PathoLogic component of Pathway Tools to predict organism-specific metabolic networks [53]. The September 2025 release (version 29.1) added 41 new pathways and revised 15 existing pathways, demonstrating the continuous expansion of this knowledge base [32].
The Pathway Tools software provides the computational foundation for both the BioCyc web platform and local installation [53]. Its modular architecture includes:
Pathway Tools is freely available to academic researchers, allowing institutions to create and maintain custom PGDBs for non-model organisms of specific interest [53]. The software can run as both a desktop application and web server, supporting individual research and collaborative projects.
Protocol 3.1: Creating a New Pathway/Genome Database for a Non-Model Organism
Objective: Generate a computationally predicted metabolic network from genomic data to form a foundation for experimental investigation.
Input Requirements:
Methodology:
Validation Steps:
The resulting Tier 3 PGDB provides a preliminary metabolic network that can be refined through manual curation as experimental data becomes available [31]. For the non-model organism researcher, this computationally-generated reconstruction serves as a testable scaffold for designing hypothesis-driven experiments to validate predicted metabolic capabilities.
Protocol 3.2: Community Curation of Organism-Specific PGDBs
Objective: Improve the accuracy and biological relevance of a PGDB through literature-based curation and experimental data integration.
Background: The Nostoc/Anabaena sp. PCC 7120 database exemplifies successful community curation, where researchers contributed information from 444 peer-reviewed publications covering 72 proteins, 5 metabolic pathways, and 28 small regulatory RNAs [32].
Curation Workflow:
Quality Control:
This protocol enables research communities to collectively build authoritative resources for non-model organisms, transforming computational predictions into knowledge-based representations of cellular biochemistry [32] [31].
Diagram 1: Community curation workflow for PGDBs
Protocol 4.1: Visualization of Transcriptomics Data on Metabolic Maps
Objective: Overlay gene expression data onto organism-specific metabolic network diagrams to identify differentially active metabolic subsystems.
Input Requirements:
Methodology:
The Cellular Overview provides a zoomable metabolic map that enables researchers to study local reaction neighborhoods while maintaining context within the full metabolic network [55]. This visualization approach facilitates rapid identification of metabolic bottlenecks, coordinated pathway regulation, and condition-specific metabolic adaptations in non-model organisms.
Protocol 4.2: Comparative Metabolic Analysis Across Multiple Organisms
Objective: Identify metabolic differences and similarities between non-model organisms and reference species to infer specialized metabolic capabilities.
Methodology:
Table 2: BioCyc Analysis Tools and Applications for Non-Model Organisms
| Tool Name | Functionality | Research Application |
|---|---|---|
| Cellular Overview | Zoomable metabolic map | Visualization of omics data on metabolic networks |
| Omics Dashboard | Hierarchical data visualization | Drill-down analysis of functional categories |
| RouteSearch | Path finding in metabolic networks | Identify potential metabolic routes between compounds |
| SmartTables | Set-based analysis of genes/metabolites | Group analysis and data integration |
| Comparative Genome Dashboard | Multi-organism comparison | Identification of metabolic specializations |
| Genome Browser | Visual genome exploration | Positional analysis of genomic features |
Diagram 2: Multi-omics data analysis workflow
The CyanoCyc web portal exemplifies the application of BioCyc resources to a phylogenetically-defined group of non-model organisms [54]. The recent curation of Nostoc/Anabaena sp. PCC 7120 involved collaboration between SRI curators and the cyanobacteria research community, resulting in detailed annotation of specialized metabolic pathways including:
This case study demonstrates how community curation efforts can transform a generic PGDB into an organism-specific knowledge base that captures specialized metabolic adaptations [32].
The incorporation of Vibrio natriegens ATCC 14048 as a Tier 2 curated database showcases BioCyc's utility for organisms with specialized metabolic capabilities [32]. This marine bacterium possesses an exceptionally short doubling time (<10 minutes) and exhibits metabolic versatility that makes it valuable for synthetic biology applications. The curation process included:
This enhanced PGDB provides researchers with a reliable resource for exploiting this non-model organism's unique metabolic capabilities in biotechnological applications.
Table 3: Key Research Reagent Solutions for Metabolic Pathway Analysis
| Reagent/Resource | Function | Application Context |
|---|---|---|
| Pathway Tools Software | PGDB creation and analysis | Generating organism-specific metabolic databases from genomic data |
| MetaCyc Database | Reference metabolic pathway collection | Template for pathway prediction and comparative analysis |
| BioCyc Subscription | Access to curated PGDBs | Reference data for thousands of organisms |
| SmartTables Module | Gene/metabolite set analysis | Management and analysis of omics datasets |
| Cellular Overview Diagrams | Metabolic network visualization | Contextual interpretation of experimental data |
| Omics Dashboard | Hierarchical data exploration | Multi-level analysis of functional datasets |
| RouteSearch Tool | Metabolic path finding | Identification of connections between metabolites |
| Comparative Analysis Tools | Cross-organism comparison | Identification of metabolic specializations |
| NuPro | NuPro® Yeast-Based Nucleotide Supplement | NuPro® is a yeast-derived nutrient rich in nucleotides for animal health and production research. For Research Use Only (RUO). Not for human or veterinary use. |
| Isomolar | Isomolar, CAS:100631-27-4, MF:C6H5Cl2N | Chemical Reagent |
Effective utilization of BioCyc and Pathway Tools requires strategic data management practices. Researchers should establish consistent identifier mapping between their experimental data and BioCyc gene/protein identifiers to enable seamless data integration. For non-model organisms, we recommend implementing a version control system for custom PGDBs to track refinements and additions as knowledge accumulates.
The SmartTables functionality provides a powerful mechanism for integrating diverse datasets including transcriptomics, proteomics, metabolomics, and flux measurements [27]. These tables can be shared among collaborators, enabling team-based analysis while maintaining data integrity.
For researchers developing custom PGDBs for non-model organisms, validation protocols are essential to ensure metabolic model accuracy. We recommend:
The Pathway Tools software includes built-in validation tools that can identify thermodynamically inconsistent reactions, mass-imbalanced equations, and blocked reactions in metabolic networks [53].
The BioCyc platform continues to evolve, with recent developments enhancing its utility for non-model organism research. The September 2025 release introduced significant improvements to HumanCyc, including incorporation of the complete human genome sequence and updated NCBI annotations, demonstrating the platform's commitment to data currency [32]. The addition of new visualization capabilities and expanded omics data integration tools further strengthens the platform's analytical power.
Emerging capabilities in metabolic route search and pathway collages enable researchers to design novel metabolic pathways and visualize custom pathway combinations [27]. These features are particularly valuable for metabolic engineering applications in non-model organisms, where synthetic pathways may be required to achieve desired bioproduction goals.
For the non-model organism researcher, BioCyc and Pathway Tools provide an increasingly essential framework for transforming genomic data into biochemical knowledge, enabling hypothesis-driven investigation of organism-specific metabolic capabilities.
Metabolic pathway reconstruction in non-model organisms presents a powerful frontier in biotechnology, enabling the production of valuable biochemicals beyond traditional ethanol fermentation. While ethanol remains a dominant output in many engineered biosystems, its pathways often compete for carbon flux, limiting the economic viability and product diversity of industrial bioprocesses. Bypassing these dominant ethanol routes requires sophisticated genetic and process engineering strategies to redirect metabolic flux toward alternative target compounds.
This application note details experimental frameworks for reconstructing and optimizing metabolic networks that circumvent ethanol formation in non-model organisms. We provide validated protocols for key steps including pathway design, genetic modification, and analytical verification, with particular emphasis on overcoming the unique challenges posed by non-conventional microbial hosts. These methodologies support the broader thesis that expanding the biosynthetic capabilities of underexplored microorganisms can unlock sustainable production routes for diverse chemical building blocks, pharmaceutical intermediates, and specialty materials.
The inherent preference of many microbial systems for ethanol fermentation via pyruvate decarboxylation creates a significant metabolic engineering challenge. This dominant flux not only limits carbon efficiency for non-ethanol products but also reflects deeply conserved regulatory networks in microbial metabolism. In non-model organismsâwhich often possess advantageous traits like substrate utilization range and stress toleranceâthese native pathways can be particularly resilient to modification.
Recent advances in synthetic biology tools and systems-level metabolic modeling have made it feasible to redesign central metabolism in these challenging hosts. Successful bypass strategies typically involve: (1) knocking out competing pathways to eliminate ethanol formation, (2) introducing heterologous routes for target biochemical synthesis, and (3) implementing dynamic regulatory controls to balance redox and energy cofactors. The resulting engineered strains can convert renewable feedstocks into diverse products such as organic acids, higher alcohols, and polymer precursors with significantly improved yields and titers.
Table 1: Target Biochemicals Accessible Via Ethanol Pathway Bypass
| Biochemical Category | Representative Products | Key Pathway Intermediates | Potential Applications |
|---|---|---|---|
| Organic Acids | Succinate, Lactate, Acetate | Phosphoenolpyruvate, Pyruvate | Biopolymers, Food, Pharma |
| Higher Alcohols | Butanol, Isobutanol | 2-Keto acids, Aldehydes | Biofuels, Solvents |
| Diols | 2,3-Butanediol, 1,3-Propanediol | Dihydroxyacetone phosphate | Polymers, Antifreeze |
| Aromatic Compounds | Cinnamate, Shikimate | Erythrose-4-phosphate | Pharma, Fragrances |
The pyruvate node represents the critical branch point between ethanol formation and alternative biochemical production. Successful bypass of ethanol pathways requires multipronged engineering of pyruvate-utilizing reactions:
Genetic Knockout of Ethanol-Producing Enzymes: Begin by targeting pyruvate decarboxylase (PDC) and alcohol dehydrogenase (ADH) genes responsible for ethanol formation. Use CRISPR-Cas9 systems adapted for your non-model host to create precise deletions of these key enzymes. In parallel, introduce heterologous bypass pathways that consume pyruvate before it can enter ethanol production.
Enhancement of Alternative Pyruvate Sinks: Strengthen native pathways that compete with ethanol formation by overexpressing rate-limiting enzymes such as pyruvate dehydrogenase complex for acetyl-CoA production, pyruvate carboxylase for oxaloacetate generation, or lactate dehydrogenase for lactate synthesis. Implement expression tuning through promoter engineering to optimize flux distribution without creating metabolic imbalances.
Table 2: Key Enzymes for Pyruvate Redirection Strategies
| Engineering Approach | Target Enzymes | Effect on Metabolic Flux | Common Host Systems |
|---|---|---|---|
| Ethanol Pathway Knockout | PDC, ADH | Eliminates ethanol formation | Yeast, Zymomonas |
| Acetyl-CoA Diversion | PDH, ACS, ACL | Increases acetyl-CoA supply | Bacteria, Fungi |
| C4 Acid Production | PYC, PEPC, MDH | Redirects to TCA cycle | Actinobacteria |
| Redox-Balanced Routes | LDH, ALS, ALD | Maintains cofactor balance | Engineered E. coli |
A primary challenge in bypassing ethanol pathways is maintaining redox homeostasis when eliminating this NAD+-regenerating route. Implement these complementary strategies:
Transhydrogenase Systems: Introduce soluble or membrane-bound transhydrogenase enzymes to enable flexible cofactor interchange between NADH and NADPH pools. This approach supports pathways requiring different cofactor specificities without ethanol formation.
Synthetic NADH Sinks: Engineer synthetic electron transport chains or NADH-oxidizing pathways such as water-forming NADH oxidases to regenerate NAD+ without ethanol production. Couple these systems with your target product pathway to create metabolic valves that prevent redox imbalance.
This protocol enables simultaneous disruption of multiple ethanol pathway genes in non-model organisms using a CRISPR-Cas9 system adapted for your specific host.
Materials and Reagents
Procedure
Troubleshooting Notes
This method quantifies changes in carbon flux after ethanol pathway disruption using isotopic labeling and metabolic flux analysis.
Materials and Reagents
Procedure
Expected Outcomes Successful ethanol pathway knockout should show:
Table 3: Essential Research Reagent Solutions for Ethanol Pathway Bypass
| Reagent/Category | Specific Examples | Function in Pathway Engineering | Implementation Notes |
|---|---|---|---|
| Genetic Tools | CRISPR-Cas9 systems, Broad-host-range plasmids | Enable targeted gene knockout and heterologous pathway insertion | Must be adapted for specific non-model hosts; consider replicon compatibility |
| Enzyme Assay Kits | Pyruvate decarboxylase activity assay, Alcohol dehydrogenase activity assay | Quantify success of ethanol pathway disruption | Use cell-free extracts; normalize to total protein content |
| Analytical Standards | Ethanol-d6, [13C3]pyruvate, [13C2]acetate | Enable accurate quantification and isotopic tracing | Essential for GC-MS and LC-MS based flux analysis |
| Culture Media | Defined minimal media, Carbon source libraries | Support reproducible fermentation studies | Must exclude interfering compounds for metabolite analysis |
| Pathway Assembly | Gibson assembly master mix, Golden Gate modular cloning system | Streamline construction of complex metabolic pathways | Enables rapid testing of enzyme variants and expression levels |
| TRIGLYCERYL STEARATE | Triglyceryl Stearate | Triglyceryl Stearate for research applications. This product is For Research Use Only (RUO), not for human consumption. | Bench Chemicals |
| Comspan | Comspan|High-Purity Reagent for Research | Bench Chemicals |
Figure 1: Metabolic Engineering Strategy for Bypassing Dominant Ethanol Pathways. The diagram illustrates key intervention points for redirecting carbon flux from ethanol production toward diversified biochemical outputs. Red arrows indicate native ethanol pathway targets for disruption, while green arrows show engineered routes for product diversification.
Figure 2: Integrated Workflow for Developing Ethanol-Bypass Production Strains. The flowchart outlines the iterative process from initial design to validated strain, emphasizing the characterization phase that combines fermentation studies with advanced analytics.
The protocols and strategies outlined herein provide a comprehensive framework for bypassing dominant ethanol pathways to unlock diverse biochemical production in non-model organisms. Success in this endeavor requires systematic integration of multiple engineering approaches: genetic disruption of competing routes, careful balancing of redox metabolism, and precise analytical validation of flux redistribution.
Future directions in this field will likely involve dynamic pathway regulation using biosensors and feedback controls, as well as machine learning-assisted design of optimal pathway configurations. As synthetic biology tools continue to advance for non-model hosts, the scope of accessible products will expand significantly, moving industrial biotechnology toward more sustainable and economically viable manufacturing paradigms beyond conventional ethanol fermentation.
The application of CRISPR-Cas technologies in polyploid and recalcitrant species represents a frontier in metabolic pathway reconstruction for non-model organisms. Polyploid species, which contain multiple sets of chromosomes, are of immense agricultural importance, constituting a substantial proportion of the world's primary food and cash crops [56]. Similarly, recalcitrant speciesâthose resistant to genetic transformation and regenerationâinclude many horticulturally and industrially valuable plants characterized by high water content in tissues and limited totipotency during in vitro regeneration [57]. While polyploidy can confer enhanced agronomic traits and improved productivity, the genetic redundancy presented by multiple homologous gene copies necessitates simultaneous editing at multiple lociâa significant challenge for conventional genome editing approaches [58] [56].
The reconstruction of metabolic pathways in non-model organisms demands precise genetic manipulations that often require multiplexed editing systems. Fortunately, CRISPR-based genome editing possesses a distinct advantage in the assembly of multiplexed gRNA cassettes, making it particularly suitable for simultaneous modification of multiple gene copies in polyploid genomes [56]. Nevertheless, technical bottlenecks persist, including delivery of editing reagents, low transformation efficiency, somatic chimerism, and challenges in detecting complex editing outcomes across homologous loci [58] [59] [60]. This application note synthesizes recent advances in CRISPR tool development and experimental protocols specifically designed to overcome these barriers, with particular emphasis on applications for metabolic engineering in challenging species.
Engineering polyploid and recalcitrant species presents interconnected technical hurdles that impede efficient genome editing. In polyploids, genetic redundancy requires concurrent modification of multiple homologous genes, while their complex genomes often exhibit structural variations that complicate gRNA design and mutation detection [58] [56]. Recalcitrant species, particularly perennial crops and woody species, frequently demonstrate limited regenerative capacity, high heterozygosity, long generation times, and resistance to Agrobacterium-mediated transformation [59] [57]. These limitations are compounded by the inability to segregate transgenes through conventional breeding in vegetatively propagated species, creating a demand for transgene-free editing approaches [59].
Recent advances in CRISPR platform development have yielded specialized tools to address species-specific challenges. Table 1 summarizes the key innovative tools and their applications for overcoming barriers in polyploid and recalcitrant species.
Table 1: Advanced CRISPR Tools for Polyploid and Recalcitrant Species
| Tool/Strategy | Key Features | Applications | References |
|---|---|---|---|
| Multiplex CRISPR Systems | Simultaneous expression of multiple gRNAs; tRNA/gRNA arrays; polycistronic cassettes | Addressing genetic redundancy in polyploids; polygenic trait engineering; gene family characterization | [58] |
| CRISPR-Combo Platform | Combines genome editing with gene activation systems | Accelerates plant regeneration by activating morphogenic genes (e.g., WUS, WOX11); improves transformation efficiency | [56] |
| Viral Delivery Systems | Engineered plant viruses (e.g., SYNV, CLCrV) for reagent delivery; fusion with mobile FT RNA | Circumvents tissue culture; potential for meristem invasion; heritable mutations | [56] |
| Dominant-Metabolism Compromised Intermediate-Chassis (DMCI) | Attenuates native dominant metabolic pathways | Redirects carbon flux in non-model microbes for enhanced production of target biochemicals | [3] |
| Nodal Culture Regeneration | Utilizes immature nodal explants with high meristematic activity | Improves regeneration in recalcitrant horticultural crops; reduces contamination | [57] |
| Enzyme-Constrained Genome-Scale Models (ecGEMs) | Integrates enzyme kinetics with metabolic models | Predicts flux distribution; identifies rate-limiting steps in metabolic pathways | [3] |
| CELLULOSE ACETATE | CELLULOSE ACETATE, CAS:9004-35-7, MF:[C6H7O2(OH)3-m(OOCCH3)m], m = 0~3 | Chemical Reagent | Bench Chemicals |
| Cellaburate | Cellaburate, CAS:9004-36-8, MF:N/A | Chemical Reagent | Bench Chemicals |
Non-model microorganisms possess unique metabolic capabilities that make them attractive candidates for industrial biotechnology, yet the lack of efficient genetic tools has historically limited their development. CRISPR systems have been extensively developed to domesticate these non-model microbes, enabling metabolic pathway engineering for biosynthesis of target products [61] [62]. A paradigm established in Zymomonas mobilis demonstrates the effectiveness of a Dominant-Metabolism Compromised Intermediate-Chassis (DMCI) strategy for redirecting carbon flux from native ethanol production to high-value biochemicals [3].
In this approach, the innate dominant ethanol pathway was first compromised by introducing a low-toxicity but cofactor-imbalanced 2,3-butanediol pathway, creating an intermediate chassis that could subsequently be engineered for D-lactate production exceeding 140 g/L from glucose [3]. This strategy successfully bypassed the metabolic bottleneck posed by efficient native pyruvate decarboxylase (PDC) and alcohol dehydrogenases (ADHs), enabling the non-model bacterium to function as an efficient biorefinery chassis. The workflow was guided by an improved enzyme-constrained genome-scale metabolic model (eciZM547), which provided superior predictive accuracy for flux distribution compared to previous models [3].
Many agronomic traits are controlled by quantitative trait loci (QTLs) rather than single genes, presenting both a challenge and opportunity for genome editing in polyploid species. Editing of cis-regulatory elements has emerged as an effective approach to modulate gene expression and generate continuous variation in quantitative traits [56]. Successful applications include promoter editing of the VERNALIZATION 1 (VRN-1) gene in wheat, where an 8 bp deletion in the promoter region shortened head emergence time by 2-3 days without complete gene knockout [56]. Similarly, genome editing of upstream open reading frames (uORFs) enables precise manipulation of gene translation, creating a wide range of variation in crop plants [56].
Table 2: Quantitative Data on Editing Efficiencies in Polyploid Species
| Species | Target | Ploidy | Editing System | Efficiency Range | Outcome | Reference |
|---|---|---|---|---|---|---|
| Arabidopsis thaliana | 12 genes | Diploid | Cas9 with 24 individual Pol III promoters | 0-94% per locus | Successful multigene knockout; some transgene-free lines | [58] |
| Cucumis sativus | 3 MLO genes | Diploid | Cas9 with tRNA-gRNA array | Not specified | Full powdery mildew resistance | [58] |
| Triticum aestivum (wheat) | VRN-A1 promoter | Hexaploid | CRISPR/Cas9 | Not specified | 2-3 day earlier heading time | [56] |
| Allotetraploid tobacco | Somatic editing | Allotetraploid | SYNV-delivered Cas9 | High frequency somatic mutations | Limited heritability | [56] |
| Zymomonas mobilis | Ethanol pathway | Polyploid | CRISPR-Cas12a & endogenous systems | Not specified | >140 g/L D-lactate production | [3] |
The following diagram illustrates a comprehensive workflow for implementing multiplex CRISPR editing in polyploid species, integrating computational design, reagent delivery, and regeneration strategies:
Title: tRNA-gRNA Array Assembly for Multiplex Editing in Polyploid Species
Background: This protocol describes the assembly of a multiplex gRNA expression system using tRNA-processing systems for simultaneous editing of multiple homologous genes in polyploid species, addressing genetic redundancy [58].
Materials:
Procedure:
gRNA Design and Synthesis:
Vector Assembly:
Plant Transformation:
Regeneration:
Technical Notes:
Title: Enhanced Regeneration via Nodal Culture for CRISPR-Edited Recalcitrant Crops
Background: This protocol addresses the regeneration bottleneck in recalcitrant horticultural crops by utilizing immature nodal explants with high meristematic activity, significantly improving transformation efficiency [57].
Materials:
Procedure:
Explant Preparation and Sterilization:
Inoculation and Shoot Regeneration:
Root Induction:
Acclimatization:
Technical Notes:
The following diagram compares different approaches for delivering CRISPR reagents while avoiding transgene integration:
Table 3: Essential Research Reagents for Advanced CRISPR Applications
| Reagent/Category | Specific Examples | Function/Application | Considerations for Polyploid/Recalcitrant Species |
|---|---|---|---|
| CRISPR Nucleases | Cas9, Cas12a, Base Editors, Prime Editors | Creating DSBs, base conversions, precise edits | Cas12a recognizes T-rich PAMs, advantageous for some genomes; base editors enable precise single-base changes without DSBs |
| gRNA Expression Systems | tRNA-gRNA arrays, ribozyme-gRNA arrays, Pol II/III systems | Multiplexed gRNA expression; compact vector design | tRNA systems enable processing of multiple gRNAs from single transcript; useful for targeting gene families |
| Delivery Vectors | Agrobacterium binary vectors, viral vectors (SYNV, CLCrV), nanoparticle complexes | delivering editing reagents to cells | Viral vectors bypass tissue culture but may have limited cargo capacity; nanoparticles enable DNA-free delivery |
| Regeneration Enhancers | WUSCHEL (WUS), BABY BOOM (BBM), SHOOT MERISTEMLESS (STM) | Improving transformation efficiency in recalcitrant species | Co-expression with CRISPR systems boosts regeneration; CRISPR-Combo platform enables simultaneous editing and regeneration enhancement |
| Selection Systems | Antibiotic resistance, fluorescence markers, regeneration-enabling edits | Identifying successfully transformed cells | For transgene-free editing, visual markers or regeneration advantages enable selection without antibiotic resistance |
| Analytical Tools | Long-read sequencers (Oxford Nanopore, PacBio), enzyme-constrained GEMs | Detecting complex edits; predicting metabolic outcomes | Long-read sequencing essential for detecting structural variations; ecGEMs predict flux redistribution in engineered strains |
| LEMix | LEMix, CAS:102510-99-6, MF:C13H15N3O | Chemical Reagent | Bench Chemicals |
| AMBERLITE MB-150 | AMBERLITE MB-150, CAS:100915-96-6, MF:NULL | Chemical Reagent | Bench Chemicals |
The continuing evolution of CRISPR-based technologies is progressively overcoming the fundamental challenges associated with engineering polyploid and recalcitrant species. The integration of multiplex editing systems, advanced delivery methods, and enhanced regeneration protocols creates a powerful toolkit for metabolic pathway reconstruction in non-model organisms. As these technologies mature, they promise to unlock the vast biotechnological potential of previously intractable species, enabling the development of novel traits and expanding the repertoire of organisms available for industrial and agricultural applications. Future directions will likely focus on improving spatiotemporal control of editing, enhancing prediction of complex phenotypic outcomes from multiplex edits, and developing increasingly sophisticated DNA-free delivery systems to streamline regulatory approval and commercialization.
The reconstruction of metabolic pathways in non-model organisms presents a significant challenge for researchers in metabolic engineering and drug development. Unlike well-characterized model species, non-model organisms lack comprehensive biochemical annotations and organism-specific data, making the construction of high-quality Genome-Scale Metabolic models (GEMs) particularly difficult [63]. These mathematical representations of cellular metabolism are powerful tools for predicting physiological states and metabolic fluxes, yet their predictive accuracy is often hampered by knowledge gaps and missing reactions in the metabolic network [64]. The integration of machine learning (ML) techniques with GEMs has emerged as a transformative approach to overcome these limitations, enabling more predictive bioengineering and accelerating the development of microbial cell factories for biotechnological applications [65] [66]. This protocol details practical methodologies for leveraging ML to optimize metabolic pathways and enzyme activity within the framework of GEMs, with particular emphasis on applications for non-model species.
Context: Draft GEMs, especially for non-model organisms, frequently contain gaps resulting from incomplete genomic and functional annotations. Traditional gap-filling methods often require experimental phenotypic data, which may be unavailable for less-studied species [64]. CHESHIRE (CHEbyshev Spectral HyperlInk pREdictor) is a deep learning method that predicts missing reactions in GEMs using solely topological features of the metabolic network, requiring no experimental data input [64].
Protocol: Implementing CHESHIRE for Metabolic Network Completion
The following diagram illustrates the CHESHIRE workflow for predicting missing reactions:
Context: Kinetic models traditionally used to predict metabolic pathway dynamics are difficult to develop and rely heavily on domain expertise. A machine learning approach can instead learn the dynamics governing a pathway directly from time-series multiomics data (e.g., proteomics and metabolomics), producing accurate predictions capable of guiding bioengineering efforts [67].
Protocol: ML-Based Prediction of Pathway Dynamics
The workflow for this dynamic modeling approach is depicted below:
Table 1: Essential reagents, tools, and datasets for integrating Machine Learning with GEMs.
| Item Name | Type/Format | Function in the Protocol | Critical Parameters for Success |
|---|---|---|---|
| Genome Annotation File | Data File (e.g., GFF, GBK) | Provides the initial gene set and functional predictions required for draft GEM reconstruction. | Completeness and accuracy of functional assignments. |
| Biochemical Reaction Database | Database (e.g., MetaCyc, KEGG) | Serves as a universal reaction pool for gap-filling algorithms to propose missing metabolic functions [64]. | Comprehensive coverage of metabolic transformations across all kingdoms of life. |
| Time-Series Multiomics Dataset | Dataset (Proteomics, Metabolomics) | Provides the training data for ML models learning pathway dynamics; includes protein and metabolite concentrations over time [67]. | High temporal resolution, technical reproducibility, and precise quantification. |
| CHESHIRE Software Package | Software/Algorithm | A specific deep learning tool for topology-based prediction of missing reactions in a metabolic network [64]. | Proper hyperparameter tuning and adequate negative sampling during training. |
| Stoichiometric Matrix | Mathematical Matrix (S) | The core mathematical representation of a GEM, defining the mass balance constraints for all reactions in the network. | Accurate reaction stoichiometry and correct assignment of reaction directionality. |
| Meliodent | Meliodent, CAS:120366-88-3, MF:C10H22N2 | Chemical Reagent | Bench Chemicals |
| gelucire 44-14 | gelucire 44-14, CAS:121548-04-7, MF:C9H14N2 | Chemical Reagent | Bench Chemicals |
Applying these ML-augmented methods to non-model organisms like Atlantic cod (Gadus morhua) requires a structured workflow [63]. The process begins with generating a draft model from available annotation data, which is often sparse. This draft model is subsequently refined using a topology-based ML method like CHESHIRE to propose and add missing reactions, thereby improving network connectivity and functionality without immediate need for experimental data [64]. The curated GEM can then be used to simulate metabolic fluxes. For more dynamic predictions, especially for a heterologous pathway, time-series multiomics data can be collected and used to train an ML model as described in Section 2.2, allowing for the prediction of metabolite concentration changes over time and the identification of potential bottlenecks [67]. This integrated approach, which combines the ML-augmented GEM with learned dynamics, facilitates the final step of proposing and prioritizing genetic interventions (e.g., enzyme engineering or regulatory element modifications) to optimize the desired metabolic output [65].
The following diagram summarizes this comprehensive, multi-stage research program:
High-throughput screening (HTS) has become an indispensable methodology in the development of microbial cell factories, enabling researchers to rapidly evaluate vast libraries of microbial strains or genetic variants. In the context of metabolic pathway reconstruction in non-model organisms, HTS addresses a critical bottleneck in the Design-Build-Test-Learn (DBTL) cycle by allowing for the rapid phenotypic evaluation of engineered strains [68] [69]. Traditional strain screening methods, primarily based on colony plate assays, lack the capacity for detailed phenotypic screening and are limited by low throughput, delayed feedback, and an inability to address cellular heterogeneity [68]. Contemporary HTS platforms investigate hundreds of thousands of compounds or genetic variants per day, dramatically accelerating the discovery and optimization of strains for industrial applications [69]. This application note details established and emerging HTS platforms, provides validated protocols, and outlines essential tools specifically framed within metabolic engineering of non-model organisms.
The selection of an appropriate HTS platform is paramount to the success of any strain development campaign. The table below summarizes the core characteristics of current HTS platforms relevant to metabolic engineering.
Table 1: Comparison of High-Throughput Screening Platforms for Strain Development
| Platform/Technology | Throughput | Key Principle | Resolution | Primary Applications in Strain Development |
|---|---|---|---|---|
| Digital Colony Picker (DCP) [68] | 16,000 microchambers | AI-powered imaging of picoliter-scale microchambers with contact-free export | Single-cell | Growth and metabolic phenotyping (e.g., lactate production, stress tolerance) |
| Acoustic-Droplet-Ejection Mass Spectrometry (ADE-MS) [70] | Seconds per sample | Acoustic ejection of samples directly into MS ionization source | Population | Ultrahigh throughput screening of metabolite production (e.g., from industrial strains) |
| Microtiter Plate-Based Screening [71] [69] | 96-/384-well formats | Colorimetric or fluorometric assays in standardized plates | Population | Enzyme activity (e.g., isomerase), substrate utilization, and tolerance screening |
| Colorimetric Assays (e.g., Seliwanoff's reaction) [71] | 96-well format | Chemical reaction producing a visible color change correlated with activity | Population | Specific detection of metabolic activity (e.g., D-allulose depletion by L-rhamnose isomerase) |
Choosing the correct platform depends on the specific goals and constraints of the project. AI-powered Digital Colony Pickers (DCP), like the one described by [68], represent a cutting-edge approach. This platform uses a microfluidic chip with 16,000 addressable picoliter-scale microchambers to compartmentalize individual cells. An AI-driven image analysis system dynamically monitors single-cell morphology, proliferation, and metabolic activities. Target clones are subsequently exported via a laser-induced bubble technique, all without physical contact [68]. This platform is ideal for screening based on complex growth and metabolic phenotypes at single-cell resolution.
For extreme speed in sample analysis, Acoustic-Droplet-Ejection Mass Spectrometry (ADE-MS) enables fully automated sample pretreatment and analysis, processing samples in seconds [70]. This technology is particularly valuable when direct quantification of a wide array of metabolites is required at an ultrahigh throughput level.
Conversely, well-established microtiter plate-based assays remain a robust and accessible workhorse for many laboratories. These can be coupled with colorimetric assays, such as the Seliwanoff's reaction protocol optimized for detecting L-rhamnose isomerase activity by monitoring D-allulose depletion [71]. These assays are highly reliable for screening specific enzymatic activities or metabolic conversions.
This protocol outlines the procedure for screening Zymomonas mobilis mutants for enhanced lactate production and tolerance using the DCP platform [68].
Research Reagent Solutions:
Methodology:
This protocol, adapted from [71], provides a robust method for screening isomerase variant libraries, such as L-rhamnose isomerase (L-RI).
Research Reagent Solutions:
Methodology:
The reconstruction and analysis of metabolic pathways in non-model organisms present unique challenges, primarily due to gene homology mismatches that can lead to incomplete or inaccurate network models [72]. Tools like the Metabolic Interactive Nodular Network for Omics (MINNO) have been developed to address this. MINNO is a JavaScript-based web application that allows users to create and modify interactive metabolic pathway visualizations for thousands of organisms [72].
Key Features and Workflow:
This hybrid genomics-metabolomics approach is crucial for elucidating the functional metabolic architecture of non-model organisms like Zymomonas mobilis or Borrelia species, guiding subsequent engineering strategies.
Diagram 1: A workflow for reconstructing and validating metabolic pathways in non-model organisms using a hybrid genomics-metabolomics approach, culminating in target identification for engineering.
The non-model bacterium Zymomonas mobilis exemplifies the application of advanced HTS and metabolic engineering strategies. It is an excellent chassis due to its extraordinary industrial characteristics, including high sugar uptake rate and ethanol yield [3]. A major challenge in engineering this organism is overcoming its innate dominant ethanol production pathway.
Engineering Strategy and HTS Application: A Dominant-Metabolism Compromised Intermediate-Chassis (DMCI) strategy was employed to circumvent the dominant ethanol pathway. This involved introducing a low-toxicity but cofactor-imbalanced pathway (2,3-butanediol) to perturb central metabolism before introducing the final D-lactate production pathway [3]. The successful implementation of this strategy relied on HTS to identify strains where the ethanol pathway was sufficiently compromised. The resulting engineered producer was able to generate over 140.92 g/L D-lactate from glucose with a yield exceeding 0.97 g/g [3]. This case demonstrates the critical role of HTS in validating intermediate chassis and isolating successful high-producing mutants from a heterogeneous pool of engineered cells.
Diagram 2: Metabolic engineering strategy in Z. mobilis using a Dominant-Metabolism Compromised Intermediate-Chassis (DMCI) to bypass native ethanol production for high-yield D-lactate production.
Genome-scale metabolic models (GEMs) provide a mathematical representation of an organism's metabolism, connecting genomic information to metabolic phenotypes. For non-model organismsâspecies lacking extensive experimental characterizationâthe reconstruction of these models relies heavily on automated tools [73]. The selection of an appropriate reconstruction tool is therefore a critical first step in research aimed at elucidating the metabolic capabilities of understudied species. This application note provides a comparative analysis of three prominent automated reconstruction toolsâCarveMe, gapseq, and KBaseâfocusing on their underlying methodologies, performance characteristics, and suitability for non-model organism research. Such tools enable the in silico prediction of metabolic network properties, which can guide experimental design in metabolic engineering and drug development [74] [8].
Automated reconstruction tools employ distinct strategies to convert genomic data into functional metabolic models. CarveMe utilizes a top-down approach, starting with a universal model containing all known metabolic reactions and "carving out" those unsupported by genomic evidence [74] [75]. In contrast, both gapseq and KBase employ bottom-up approaches, building models from scratch by mapping annotated genes to biochemical reactions [74] [8]. KBase further distinguishes itself as an integrated platform that combines reconstruction capabilities with various other analysis tools, including metagenomic assembly and RNA-seq analysis [76].
A recent comparative study analyzing marine bacterial communities revealed that these approaches, when applied to the same genomic input, can produce models with significant structural and functional differences [74] [77]. The table below summarizes the key characteristics of each tool.
Table 1: Key Characteristics of Automated Reconstruction Tools
| Feature | CarveMe | gapseq | KBase |
|---|---|---|---|
| Reconstruction Approach | Top-down | Bottom-up | Bottom-up |
| Primary Database | BiGG | ModelSEED, MetaCyc | ModelSEED |
| User Interface | Command-line | Command-line | Web-based platform |
| Output | Ready-to-use model for FBA | Ready-to-use model for FBA | Ready-to-use model for FBA |
| Ideal Use Case | Rapid reconstruction of individual organisms | Comprehensive pathway prediction | End-to-end analysis from sequences to models |
| Gap-Filling Strategy | Context-specific | Informed by pathway topology and homology | Medium-specific during reconstruction |
Quantitative comparisons of models generated from the same metagenome-assembled genomes (MAGs) highlight substantial variations in model content and predictive performance. The following table presents a structural comparison of GEMs reconstructed from identical bacterial genomes using the different tools.
Table 2: Quantitative Structural Comparison of GEMs from Marine Bacterial Communities (adapted from [74])
| Reconstruction Tool | Average Number of Genes | Average Number of Reactions | Average Number of Metabolites | Notable Features |
|---|---|---|---|---|
| CarveMe | Highest | Intermediate | Intermediate | Efficient model generation |
| gapseq | Lowest | Highest | Highest | Lowest false negative rate in enzyme activity prediction [8] |
| KBase | Intermediate | Intermediate | Intermediate | Higher similarity to gapseq models |
| Consensus Approach | High | Highest | Highest | Reduces dead-end metabolites |
Table 3: Essential Research Reagents and Computational Tools
| Item Name | Function/Description | Example/Format |
|---|---|---|
| Genome Sequence | Primary input for reconstruction | FASTA file (.fna, .fa) |
| Biochemical Databases | Source of reaction stoichiometry, metabolite, and enzyme information | BiGG, ModelSEED, MetaCyc |
| COMMIT | Algorithm for community model gap-filling | Python package |
| MetaNetX | Platform for reconciling metabolite and reaction namespaces across databases | Web resource/API |
| COBRA Toolbox | MATLAB package for constraint-based modeling | MATLAB toolbox |
| GEMsembler | Python package for comparing and building consensus models | Python package |
Protocol 1: Standardized Reconstruction Using Individual Tools
Input Requirements: A high-quality genome sequence in FASTA format. For non-model organisms, ensure comprehensive annotation using tools like RAST or Prokka, which are also available within the KBase platform [76] [78].
gapseq Reconstruction Procedure:
gapseq draft -genome genome.fna -o output_dir.CarveMe Reconstruction Procedure:
pip install carveme.carve genome.fna -o model.xml. This carves the universal model based on genome annotation.--media flag to refine the gap-filling process for particular environmental conditions.KBase Reconstruction Procedure:
Evidence suggests that consensus models, which integrate reconstructions from multiple tools, can capture a more comprehensive view of an organism's metabolic potential while reducing tool-specific biases [74] [75]. These models typically encompass a larger number of reactions and metabolites while concurrently reducing the presence of dead-end metabolites [74].
Protocol 2: Generating a Consensus Model Using GEMsembler
Principle: The GEMsembler pipeline converts models from different tools to a unified namespace, combines them into a "supermodel," and generates consensus models with features present in a user-defined subset of the input models [75].
Diagram 1: Workflow for building a consensus metabolic model. The process begins with a single genome file, which is used to reconstruct models via different tools. These models are then unified and combined into a consensus model using GEMsembler.
Research on non-model organisms presents unique challenges that directly impact metabolic reconstruction. The following technical considerations are paramount:
Database Biases: Automated tools rely on biochemical databases (BiGG, ModelSEED, MetaCyc) that are inherently biased toward well-studied model organisms. This can lead to incomplete or inaccurate networks for novel species with unique metabolisms [74] [79]. The consensus approach helps mitigate this by aggregating evidence from multiple databases.
Gap-Filling Pitfalls: Gap-filling is necessary to complete metabolic networks but can introduce reactions without genomic evidence. gapseq's algorithm, which incorporates pathway topology and sequence homology beyond a single growth medium, can yield more versatile models for non-standard environments [8].
Validation with Limited Data: For non-model organisms, experimental data for validation is often scarce. Researchers should leverage any available phenotypic data, such as carbon source utilization or fermentation products, to benchmark model predictions [8] [78]. The AGORA framework, while focused on human microbes, offers a paradigm for standardized model reconstruction and validation [73].
Compartmentalization and Specialized Metabolism: Eukaryotic non-model organisms require careful attention to subcellular compartmentalization. While automated tools are improving, manual curation is often necessary to correctly localize reactions [73]. Furthermore, pathways for unique natural products often require de novo prediction methods, as they are absent from reference databases [79].
CarveMe, gapseq, and KBase each offer distinct advantages for metabolic reconstruction of non-model organisms. CarveMe provides speed and efficiency, gapseq offers comprehensive pathway prediction and accuracy, and KBase delivers an integrated, user-friendly environment. The emerging best practice is to leverage a consensus approach, utilizing tools like GEMsembler to integrate the strengths of individual reconstructions. This strategy provides a more robust and comprehensive foundation for downstream applications in drug target identification and metabolic engineering by minimizing the biases inherent to any single tool or database.
Genome-scale metabolic models (GEMs) serve as powerful computational frameworks for predicting the metabolic capabilities of organisms, with applications ranging from metabolic engineering to drug discovery [64] [80]. For non-model organismsâspecies lacking comprehensive biochemical and genetic characterizationâthe reconstruction of high-quality GEMs presents particular challenges. Automated reconstruction tools such as CarveMe, gapseq, and KBase leverage different biochemical databases and algorithms, resulting in models with varying network structures and functional predictions from the same genomic starting point [74] [80]. This variability introduces significant prediction uncertainty, undermining the reliability of biological insights and practical applications.
A critical manifestation of incomplete metabolic networks is the presence of dead-end metabolites (DEMs)âmetabolites that are produced but not consumed, or consumed but not produced within the network, indicating gaps in metabolic pathways [81]. DEMs represent the "known unknowns" of metabolism, highlighting areas where our understanding of the metabolic network is incomplete [81]. Consensus modeling has emerged as a powerful strategy to mitigate these limitations. By integrating multiple individual reconstructions into a unified model, consensus approaches enhance network completeness and reduce reliance on any single reconstruction method, thereby providing a more robust framework for metabolic analysis in non-model organisms [74].
Different automated reconstruction tools produce GEMs with substantial structural differences, even when based on identical genomic input. A comparative analysis of models reconstructed from 105 metagenome-assembled genomes (MAGs) from marine bacterial communities revealed significant variations in network content and composition across three commonly used tools [74].
Table 1: Structural characteristics of GEMs from different reconstruction approaches
| Reconstruction Approach | Number of Genes | Number of Reactions | Number of Metabolites | Number of Dead-End Metabolites |
|---|---|---|---|---|
| CarveMe | Highest | Moderate | Moderate | Low |
| gapseq | Low | Highest | Highest | Highest |
| KBase | Moderate | Low | Low | Moderate |
| Consensus | High | High | High | Lowest |
The analysis demonstrated that gapseq models typically encompassed more reactions and metabolites compared to CarveMe and KBase models. However, this increased network size came with a trade-off: gapseq models also exhibited a larger number of dead-end metabolites, which can impair network functionality and predictive accuracy [74]. In contrast, consensus models integrated content from multiple approaches, resulting in more comprehensive network coverage while simultaneously reducing dead-end metabolites.
The Jaccard similarity index, which measures the similarity between sets, reveals low overlap between GEMs reconstructed from the same genome using different tools [74].
Table 2: Jaccard similarity between reconstruction approaches (coral-associated bacteria models)
| Comparison | Reaction Similarity | Metabolite Similarity | Gene Similarity |
|---|---|---|---|
| gapseq vs KBase | 0.23 | 0.37 | - |
| CarveMe vs KBase | - | - | 0.42 |
| CarveMe vs Consensus | - | - | 0.75 |
The relatively higher similarity between gapseq and KBase models in terms of reactions and metabolites (attributed to their shared use of the ModelSEED database) and between CarveMe and KBase models in gene composition highlights how database choices and algorithmic approaches differentially shape the resulting reconstructions [74]. The strong similarity between CarveMe and consensus models (0.75 for genes) indicates that consensus approaches effectively preserve and integrate content from individual reconstructions rather than generating entirely novel network structures.
The following diagram illustrates the comprehensive workflow for constructing consensus metabolic models, integrating multiple automated reconstruction tools and refinement steps:
Objective: Generate multiple draft GEMs using different automated reconstruction tools.
Procedure:
carve genome.faa --output model.xmlgapseq find -p all genome.faagapseq draft -b reaction_presence.lstcobrapy or the COBRA Toolbox to facilitate comparison and integration.Objective: Merge individual draft reconstructions into a unified consensus model.
Procedure:
cobrapy checkmassbalance() function or equivalent.Objective: Identify and fill metabolic gaps while validating model functionality.
Procedure:
For enhanced predictive accuracy, incorporate enzyme constraints into consensus models:
Procedure:
The CHESHIRE method provides a deep learning approach for identifying missing reactions in GEMs:
Procedure:
Table 3: Key resources for consensus metabolic model reconstruction
| Category | Resource | Description | Application in Consensus Modeling |
|---|---|---|---|
| Reconstruction Tools | CarveMe | Top-down reconstruction using universal template model | Generates one component of consensus model |
| gapseq | Bottom-up reconstruction with comprehensive biochemical data integration | Provides complementary network perspective | |
| KBase | Web-based platform for automated draft model generation | Enables rapid reconstruction without local installation | |
| Biochemical Databases | BiGG Models | Curated metabolic reconstruction database | Provides standardized namespace for reaction mapping |
| ModelSEED Framework for annotation and model generation | Common database for multiple tools | ||
| BRENDA Comprehensive enzyme information database | Source of enzyme kinetic parameters | ||
| Analysis Software | COBRA Toolbox MATLAB package for constraint-based modeling | Model simulation, gap-filling, and validation | |
| Pathway Tools Bioinformatics software package | Dead-end metabolite identification | ||
| CHESHIRE Deep learning method for reaction prediction | Topology-based gap-filling | ||
| Validation Resources | MEMOTE Open-source test suite for GEM quality assessment | Model quality control and standardization | |
| AutoPACMEN Enzyme constraint prediction tool | kcat value estimation for enzyme constraints |
Consensus modeling represents a paradigm shift in metabolic reconstruction for non-model organisms, directly addressing the critical challenges of prediction uncertainty and dead-end metabolites. By integrating multiple reconstruction approaches, consensus models capture a more complete representation of metabolic capabilities while minimizing tool-specific biases. The structured protocol outlined here provides researchers with a comprehensive framework for implementing this powerful approach, from initial draft reconstruction to advanced machine learning-enhanced gap-filling. As the field progresses, the integration of enzyme constraints, machine learning methods, and expanded biochemical databases will further enhance the predictive power of consensus models, accelerating the exploration of non-model organisms for biomedical and biotechnological applications.
This application note provides a detailed protocol for employing 13C-Metabolic Flux Analysis (13C-MFA) to validate model predictions of metabolic pathway activity, with a specific focus on non-model organisms. We outline a standardized workflow that integrates multi-omics data to construct and refine genome-scale metabolic models, design definitive tracer experiments, and statistically validate flux predictions. The procedures described herein are designed to help researchers overcome the challenges inherent in studying poorly characterized metabolic systems, enabling accurate quantification of intracellular reaction rates in non-model organisms for metabolic engineering and systems biology.
Metabolic pathway reconstruction in non-model organisms is often hindered by incomplete genomic annotation and poor gene homology, leading to gaps in metabolic networks [72]. While in silico models can predict metabolic fluxes, these predictions require empirical validation to accurately represent in vivo physiology. 13C-MFA has emerged as the gold-standard technique for quantifying intracellular metabolic fluxes, providing a direct method for validating model predictions [82] [83]. When integrated with multi-omics datasets, 13C-MFA offers a powerful framework for probing the metabolic architecture of non-model organisms, identifying bottlenecks in biochemical production, and guiding metabolic engineering strategies [84] [3].
This protocol details the application of 13C-MFA within a multi-omics context to validate metabolic predictions, emphasizing experimental design, data integration, and statistical validation specifically tailored for non-model systems where metabolic networks may be incomplete or poorly annotated.
13C-MFA utilizes stable isotope tracers to track the flow of carbon through metabolic networks. Cells are cultured with 13C-labeled substrates, and the resulting labeling patterns in intracellular metabolites are measured via mass spectrometry [83]. These labeling distributions are used with computational models to infer in vivo metabolic reaction rates [84]. The validation of model predictions against experimentally determined fluxes provides a rigorous test of metabolic reconstruction accuracy.
The following workflow diagram illustrates the integrated process for validating metabolic predictions in non-model organisms:
Objective: Develop a functional genome-scale metabolic model (GEM) for flux prediction.
Procedure:
Objective: Acquire high-quality isotopic labeling and extracellular rate data for 13C-MFA.
Procedure:
Objective: Estimate intracellular fluxes and statistically validate the model predictions.
Procedure:
The following table details essential materials and their functions for conducting 13C-MFA validation studies.
Table 1: Essential Research Reagents and Tools for 13C-MFA Validation
| Item | Function / Application in Protocol | Examples / Specifications |
|---|---|---|
| 13C-Labeled Substrates | Serve as metabolic tracers; choice is critical for flux elucidation in specific pathways. | [1,2-13C]Glucose, [U-13C]Glucose; isotopic purity > 99% [84] [83]. |
| Minimal Culture Medium | Provides a defined chemical environment to ensure the tracer is the sole carbon source. | Custom formulations (e.g., M9 for bacteria, DMEM without glucose/glutamine for mammalian cells) [83]. |
| GC-MS / LC-MS System | Analytical instrumentation for measuring Mass Isotopomer Distributions (MIDs) of metabolites. | Systems from Agilent, Thermo Fisher, etc.; GC-MS often requires derivatization (e.g., TBDMS) [84] [85]. |
| 13C-MFA Software | Computational platform for flux estimation, model simulation, and statistical analysis. | INCA, Metran, 13CFLUX2, OpenFLUX2 [84] [86] [83]. |
| Data Integration & Visualization Tools | For multi-omics integration and empirical refinement of metabolic networks for non-model organisms. | MINNO, Escher, Omix [72]. |
Traditional model selection relying solely on the ϲ-test can be sensitive to errors in measurement uncertainty estimates, potentially leading to overfitting or underfitting [86]. For more robust validation:
The integration of 13C-MFA with multi-omics data provides a powerful, empirical framework for validating metabolic predictions in non-model organisms. This protocol outlines a systematic approach from model construction through to statistical validation, enabling researchers to move beyond genomic predictions to a quantitative understanding of in vivo metabolic function. By iteratively applying this cycle, metabolic reconstructions of non-model organisms can be rigorously refined, accelerating their development as robust chassis for biotechnology and providing deeper insights into their unique physiology.
The transition from laboratory-scale success to commercially viable bioprocesses requires rigorous assessment of both economic feasibility and environmental impact. For products derived from metabolic pathway reconstruction in non-model organisms, such as D-lactate, Techno-Economic Analysis (TEA) and Life Cycle Assessment (LCA) provide complementary analytical frameworks that are crucial for research prioritization and investment decisions. TEA evaluates the economic viability of a process by calculating production costs, identifying cost drivers, and establishing minimum selling prices [88]. Concurrently, LCA quantifies environmental impacts across the entire value chainâfrom raw material extraction to end-of-life disposalâenabling researchers to identify and mitigate environmental hotspots [89] [90]. For non-model organisms like Zymomonas mobilis and Komagataella phaffii engineered for D-lactate production, these analyses provide critical data to bridge the gap between metabolic engineering achievements and industrial implementation [3] [91].
TEA employs process modeling and economic calculations to determine the financial viability of bioprocesses. For renewable diesel production from animal waste oil via hydrothermal conversion, researchers demonstrated a minimum fuel selling price (MFSP) of \$0.76/kg, with sensitivity analysis revealing a range of \$0.64â\$0.89/kg based on variations in capital investment, feedstock price, labor costs, and byproduct valuation [88]. This approach is directly applicable to D-lactate production, where similar calculations determine competitiveness against petroleum-derived alternatives.
LCA follows standardized ISO methodologies (ISO 14040:2006 and 14044:2006) to evaluate environmental impacts across multiple categories [89]. The "cradle-to-gate" system boundary encompasses raw material acquisition, production, and processing, while "cradle-to-grave" analyses include product use and end-of-life disposal [90]. For polylactic acid (PLA)âa key derivative of lactateâLCA studies quantify global warming potential (GWP) in kg COâ-equivalent per unit product, along with other impact categories such as water use, land use, and eutrophication potential [92].
Table 1: TEA and LCA Benchmarks for Bio-Based Products
| Product | Feedstock | Minimum Selling Price | GWP Reduction vs. Conventional | Key Cost/Impact Drivers | Source |
|---|---|---|---|---|---|
| Renewable Diesel | Animal Waste Oil | \$0.76/kg | 34% reduction vs. petroleum diesel | Capital investment, feedstock price | [88] |
| D-Lactate | Corncob Residue Hydrolysate | Not specified | Significant GHG reduction capability demonstrated | Feedstock pretreatment, energy consumption | [3] |
| L-Lactate | Methanol (from COâ) | Commercially viable (exact value not specified) | Carbon-negative potential | Methanol metabolism efficiency, cofactor balancing | [93] |
| Polylactic Acid (PLA) | Corn or Sugarcane | Varies by production method | Lower GHG vs. fossil-based plastics | Conversion process energy, feedstock agriculture | [92] |
A comprehensive study on D-lactate production using engineered Zymomonas mobilis demonstrates the integration of TEA and LCA within metabolic pathway reconstruction research. Researchers developed a Dominant-Metabolism Compromised Intermediate-Chassis (DMCI) strategy to circumvent the innate ethanol pathway in this non-model organism [3]. This involved:
The TEA demonstrated commercialization feasibility for lignocellulosic D-lactate, with the corncob residue hydrolysate feedstock substantially reducing raw material costs compared to refined sugars [3]. The high titer and yield achieved through metabolic engineering directly improved process economics by reducing fermentation volume and downstream processing requirements.
The LCA revealed significant greenhouse gas reduction capability for the lignocellulosic D-lactate process [3]. The utilization of agricultural residue (corncob) avoided the agricultural land use and fertilizer impacts associated with food crop feedstocks, while the efficient metabolic pathway minimized energy consumption during fermentation.
Objective: Introduce and optimize D-lactate biosynthesis pathways in non-model chassis organisms.
Materials:
Procedure:
Troubleshooting Tips:
Objective: Enhance D-lactate production traits in engineered strains through UV mutagenesis.
Materials:
Procedure:
Expected Outcomes: Successful mutagenesis should yield strains with 1.5-fold or higher D-lactate production compared to parent strain, as demonstrated by DLacMut2221 strain producing 5.38 g/L D-lactate from methanol [91].
Objective: Quantify environmental impacts of D-lactate production from cradle to gate.
Materials:
Procedure:
Life Cycle Inventory (LCI):
Life Cycle Impact Assessment (LCIA):
Interpretation:
Data Quality Requirements: Prefer primary data for foreground processes; use peer-reviewed secondary data for background processes. Conduct uncertainty analysis when possible.
Table 2: Essential Research Reagents for D-Lactate Pathway Engineering
| Reagent/Category | Specific Examples | Function/Application | Source/Reference |
|---|---|---|---|
| D-LDH Enzymes | Leuconostoc mesenteroides D-LDH, Lactobacillus delbrueckii D-LDH | Catalyzes pyruvate to D-lactate conversion; varying kinetics/cofactor specificity | [91] |
| Expression Vectors | Methanol-inducible (AOX1), Constitutive (GAP), Episomal plasmids, Chromosomal integration systems | Controlled gene expression; stable pathway maintenance | [93] [91] |
| Engineering Tools | CRISPR/Cas9 systems, Homologous recombination, UV mutagenesis | Genome editing; strain improvement | [3] [91] |
| Analytical Methods | HPLC with chiral columns, GC-TOF/MS, LC-MS, SIMDIS | Product quantification; metabolic flux analysis; component identification | [88] [93] |
| Modeling Resources | Genome-scale metabolic models (e.g., iZM516, eciZM547), Enzyme constraint models (ecModels) | Pathway design; flux distribution simulation; prediction of metabolic bottlenecks | [3] |
The integration of TEA and LCA early in the metabolic engineering workflow provides critical guidance for developing commercially viable and environmentally sustainable bioprocesses. For D-lactate production in non-model organisms, key success factors include:
This integrated approach ensures that research on metabolic pathway reconstruction in non-model organisms remains grounded in technical, economic, and environmental realities, accelerating the translation of laboratory innovations to industrial applications that support a circular bioeconomy.
Metabolic pathway reconstruction in non-model organisms has matured from a exploratory endeavor into a disciplined engineering science, pivotal for advancing biomedical research and sustainable biomanufacturing. The synthesis of foundational knowledge, sophisticated computational and CRISPR-based methodologies, robust optimization frameworks, and rigorous comparative validation provides a powerful toolkit for constructing efficient microbial cell factories. Future progress hinges on interdisciplinary collaboration, further development of high-efficiency genome-editing tools for recalcitrant species, and the deeper integration of machine learning with multi-omics data to create predictive, genome-scale models. These advances promise to unlock the vast, untapped metabolic potential of non-model organisms, accelerating the discovery of novel therapeutics, biofuels, and biomaterials with significant implications for clinical and industrial applications.