Recombinant Ashbya gossypii Glucose-6-phosphate isomerase (PGI1), partial, refers to a genetically engineered form of the Glucose-6-phosphate isomerase (GPI) enzyme derived from the yeast species Ashbya gossypii . GPI, also known as phosphoglucose isomerase (PGI) or phosphohexose isomerase (PHI), is an enzyme that plays a crucial role in both glycolysis and gluconeogenesis . Specifically, it catalyzes the reversible interconversion of glucose-6-phosphate (G6P) and fructose-6-phosphate (F6P) .
GPI (EC 5.3.1.9) is a dimeric enzyme that facilitates the reversible isomerization of glucose-6-phosphate (G6P) to fructose-6-phosphate (F6P), which is the second step in glycolysis .
2.1. Function
The enzyme carries out multiple functions both inside and outside the cell. Intracellularly, it participates in glycolysis, gluconeogenesis, and the pentose phosphate pathway . Extracellularly, it acts as a neurotrophic factor for spinal and sensory neurons, referred to as neuroleukin . It is also secreted by cancer cells as an autocrine motility factor, which stimulates metastasis . Additionally, extracellular GPI is known to function as a maturation factor .
2.2. Mechanism
The mechanism that GPI uses to interconvert glucose 6-phosphate and fructose 6-phosphate (aldose to ketose) consists of three major steps: opening the glucose ring, isomerizing glucose into fructose through an enediol intermediate, and closing the fructose ring .
Ashbya gossypii is a filamentous fungus known for its ability to produce high concentrations of riboflavin (vitamin B2) . Due to its efficient metabolic pathways, Ashbya gossypii is often used in industrial biotechnology for the production of various enzymes and metabolites .
The "recombinant" aspect of the enzyme indicates that the gene encoding the Ashbya gossypii PGI1 has been cloned and expressed in a host organism using recombinant DNA technology . This approach allows for large-scale production and purification of the enzyme for research and industrial applications .
The term "partial" in "Recombinant Ashbya gossypii Glucose-6-phosphate isomerase (PGI1), partial" likely indicates that the produced enzyme is either:
This might be done intentionally to study specific domains or functions of the enzyme or could be a result of the expression and purification process .
In glycolysis, GPI catalyzes the conversion of glucose-6-phosphate to fructose-6-phosphate, an essential step in the breakdown of glucose for energy production . In gluconeogenesis, the reverse reaction occurs, where fructose-6-phosphate is converted to glucose-6-phosphate, contributing to the synthesis of glucose from non-carbohydrate precursors .
Neuroleukin Activity: GPI acts as a neurotrophic factor promoting the survival of skeletal motor neurons and sensory neurons .
Autocrine Motility Factor: It functions as a tumor-secreted cytokine and angiogenic factor .
In Sugar Homeostasis: PGI plays a key role in maintaining sugar metabolism, cell wall integrity, sclerotia formation, stress responses, and virulence in animal model and crop seeds infection .
GPI Deficiency: Defects in the GPI gene can cause nonspherocytic hemolytic anemia . A severe enzyme deficiency can be associated with hydrops fetalis, immediate neonatal death, and neurological impairment .
Therapeutic Target: PGI is a potential target against cancer or Th17-mediated autoimmune diseases .
Recombinant PGI1 is used in various research applications:
Enzyme Characterization: Studying the enzyme's structure, function, and kinetics .
Metabolic Studies: Investigating its role in glycolysis, gluconeogenesis, and other metabolic pathways .
Drug Discovery: Identifying potential inhibitors or modulators of PGI1 for therapeutic purposes .
Industrial Applications: Utilizing its enzymatic activity in biotechnological processes .
| Property | Description |
|---|---|
| Enzyme Name | Glucose-6-phosphate isomerase (GPI) |
| Other Names | Phosphoglucose isomerase (PGI), Phosphohexose isomerase (PHI), Neuroleukin, Autocrine motility factor (AMF) |
| EC Number | 5.3.1.9 |
| Reaction Catalyzed | Reversible interconversion of glucose-6-phosphate (G6P) and fructose-6-phosphate (F6P) |
| Substrates | Glucose-6-phosphate, Fructose-6-phosphate |
| Cofactors | None |
| Molecular Weight | 64 kDa (monomer) |
| Structure | Dimeric enzyme composed of two identical monomers |
| Biological Functions | Glycolysis, Gluconeogenesis, Pentose Phosphate Pathway, Neurotrophic factor, Tumor metastasis |
| Clinical Relevance | GPI deficiency leads to nonspherocytic hemolytic anemia; target for cancer and autoimmune disease therapies |
| Source Organism | Ashbya gossypii (recombinant) |
KEGG: ago:AGOS_AEL249C
STRING: 33169.AAS52436
Ashbya gossypii (syn. Eremothecium gossypii) is an ascomycete fungus naturally known for riboflavin (vitamin B2) production and shares close genetic similarity with Saccharomyces cerevisiae. The significance of A. gossypii in biotechnology extends beyond its traditional role as a riboflavin producer. Recent research has demonstrated its potential as a host for heterologous protein expression, with successful examples including the expression of cellulases from Trichoderma reesei (endoglucanase I and cellobiohydrolase I) and β-galactosidase from Aspergillus niger . The organism possesses several advantageous characteristics for recombinant protein production, including the ability to secrete heterologous enzymes into the culture medium and recognize signal peptides from other organisms as secretion signals, which simplifies downstream processing of produced proteins . Additionally, A. gossypii can perform important post-translational modifications such as N-glycosylation, crucial for biological activity and protein stability .
Glucose-6-phosphate isomerase (PGI1), also known as phosphoglucose isomerase (PGI) or phosphohexose isomerase (PHI), is an essential enzyme in carbohydrate metabolism that catalyzes the reversible isomerization between glucose-6-phosphate and fructose-6-phosphate (EC 5.3.1.9) . This enzyme plays a critical role in both glycolysis and gluconeogenesis, serving as a metabolic bridge between these pathways. In A. gossypii, PGI1 functions within the context of a metabolism that has been extensively re-annotated to reveal significant differences from related yeast species . The enzyme's activity is particularly relevant in A. gossypii due to the organism's unique carbon source utilization patterns and the connection between central carbon metabolism and riboflavin production . Understanding PGI1's function provides insights into the metabolic network that supports both growth and product formation in this industrially important fungus.
A significant advantage of A. gossypii over S. cerevisiae is its tendency to hyperglycosylate heterologous proteins less extensively, particularly regarding N-glycans . This characteristic is beneficial for producing proteins whose properties might be adversely affected by extensive glycosylation . In comparative studies with cellulases expressed in both organisms, A. gossypii demonstrated less extensive glycosylation than observed in S. cerevisiae while still maintaining functionality of the expressed proteins . Additionally, A. gossypii possesses efficient secretory capabilities that allow heterologous proteins to be released into the culture medium, facilitating simpler downstream processing . The fungus also shows versatility in carbon source utilization, though with some differences compared to S. cerevisiae, which may influence cultivation strategies for recombinant protein production . These distinctive features, combined with the extensive genomic knowledge and relatively simple laboratory cultivation requirements, position A. gossypii as a compelling alternative expression system for certain applications.
For efficient expression of recombinant proteins like PGI1 in A. gossypii, researchers have typically adapted expression systems originally developed for S. cerevisiae, given the high gene homology and gene order conservation between these organisms . Based on successful heterologous protein expression studies in A. gossypii, effective promoters include the native strong constitutive promoters such as those from glycolytic pathway genes, which provide robust expression levels . The experimental work with T. reesei cellulases demonstrated that both endogenous A. gossypii promoters and heterologous promoters can be functional in this organism . When designing expression vectors, incorporating appropriate secretion signals is crucial, as A. gossypii has demonstrated the ability to recognize signal peptides from other organisms . For optimal results, expression vectors should contain selectable markers compatible with A. gossypii transformation systems and consider the filamentous growth pattern of this fungus, which differs from the unicellular nature of S. cerevisiae despite their genomic similarities.
Optimal cultivation conditions for recombinant PGI1 production in A. gossypii should account for the organism's filamentous growth pattern and metabolism. Temperature control is critical, with cultivation typically performed between 28-30°C to balance growth rate with protein expression efficiency . Medium composition significantly impacts both biomass accumulation and recombinant protein production, with carbon sources needing careful selection based on A. gossypii's unique metabolic capabilities . The pH of the culture medium should be controlled, as A. gossypii has shown greater sensitivity to low pH than most filamentous fungi . Agitation and aeration parameters must be optimized to prevent damage to the mycelia while ensuring sufficient oxygen transfer, particularly in submerged cultivation systems. For recombinant protein production, a fed-batch cultivation strategy often yields better results than simple batch cultivation, as it allows better control of growth rate and gene expression . Supplementation with specific nutrients that support protein synthesis and secretion, such as amino acids and trace elements, may further enhance PGI1 production yields.
Purification of recombinant PGI1 from A. gossypii cultures requires a multistep approach that preserves the enzyme's structural integrity and activity. Initial clarification of the culture broth should be performed through filtration or centrifugation to separate fungal biomass from the secreted proteins . For PGI1 expressed with affinity tags, affinity chromatography provides an efficient initial capture step, while ion exchange chromatography can be employed for tag-free versions based on the protein's charge properties at specific pH values . Size exclusion chromatography serves as an effective polishing step to achieve high purity (>85% as indicated for commercial preparations) . Throughout the purification process, buffer conditions should be carefully controlled to maintain enzyme stability, with glycerol often added as a stabilizing agent—the recommended final concentration being 5-50% for long-term storage . Post-purification, the enzyme can be stored either in liquid form (with a typical shelf life of 6 months at -20°C/-80°C) or lyophilized (extending shelf life to 12 months at the same temperatures) . Reconstitution of lyophilized PGI1 should be performed in deionized sterile water to a concentration of 0.1-1.0 mg/mL, with glycerol addition recommended for stability .
A. gossypii PGI1, like other glucose-6-phosphate isomerases, likely adopts a dimeric structure with each monomer containing an active site for catalyzing the reversible isomerization between glucose-6-phosphate and fructose-6-phosphate. While the search results don't provide specific structural details for A. gossypii PGI1, analysis can be inferred from the organism's evolutionary relationship with S. cerevisiae and K. lactis . Given that A. gossypii undergoes less extensive protein glycosylation compared to S. cerevisiae, its PGI1 likely exhibits glycosylation patterns that more closely resemble the native enzyme structure . The partial nature of the recombinant PGI1 described in the product information suggests that certain domains may have been excluded in the recombinant form, potentially affecting oligomerization or specific functional aspects . Understanding these structural nuances is important for research applications where enzyme functionality needs to be preserved or optimized. Researchers investigating structural aspects would benefit from techniques such as X-ray crystallography, circular dichroism spectroscopy, or computational modeling to further elucidate the specific characteristics of this enzyme.
Glycosylation of recombinant proteins in A. gossypii represents a critical factor affecting enzyme functionality, stability, and immunogenicity. Based on studies with other recombinant proteins expressed in A. gossypii, this organism demonstrates a tendency to hyperglycosylate less extensively than S. cerevisiae, particularly regarding N-glycans . This characteristic is advantageous for producing enzymes like PGI1, whose catalytic properties might be adversely affected by excessive glycosylation . The specific impact of glycosylation on PGI1 would involve effects on protein folding, thermal stability, resistance to proteolytic degradation, and potentially the fine-tuning of catalytic parameters like Km and Vmax values. Research with recombinant cellulases has shown that while A. gossypii does introduce glycosylation modifications, these are less extensive than those observed with S. cerevisiae, resulting in functional properties more closely resembling the native enzymes . For applications requiring precise control of PGI1 glycosylation, researchers might consider enzymatic deglycosylation treatments or expression system optimization to achieve the desired post-translational modification profile.
The kinetic characterization of recombinant A. gossypii PGI1 involves determining several parameters essential for understanding the enzyme's catalytic efficiency. While specific kinetic data for recombinant PGI1 from A. gossypii isn't directly provided in the search results, the approach to such characterization would typically include assessment of Km values for both glucose-6-phosphate and fructose-6-phosphate substrates, determination of kcat (turnover number), catalytic efficiency (kcat/Km), optimal pH and temperature ranges, and the effects of potential inhibitors or activators. Comparative analysis with native PGI1 would reveal any functional differences resulting from the recombinant expression system or the partial nature of the recombinant form . Based on observations with other recombinant enzymes expressed in A. gossypii, the kinetic properties might be expected to more closely resemble the native enzyme than recombinant versions produced in S. cerevisiae, due to A. gossypii's less extensive hyperglycosylation . This characteristic provides an advantage when functionally equivalent enzymes are required for research applications or when studying the fundamental properties of the enzyme in isolation from cellular contexts.
Recombinant A. gossypii PGI1 represents a valuable tool for metabolic engineering studies focused on understanding and optimizing carbon flux distribution. Researchers can employ the purified enzyme in in vitro reconstruction of glycolytic and gluconeogenic pathways to study reaction kinetics and regulatory mechanisms outside the complexity of cellular systems. Within metabolic engineering contexts, modulated expression of PGI1 through genetic manipulation can redirect carbon flux between glycolysis and the pentose phosphate pathway, potentially enhancing riboflavin production—A. gossypii's primary industrial application . The enzyme can serve as a control point for engineering efforts aimed at optimizing glucose utilization efficiency or redirecting metabolism toward specific valuable products . Additionally, isotope labeling experiments using 13C-glucose combined with recombinant PGI1 variants can help elucidate carbon flux distribution patterns and identify limiting steps in engineered pathways. The manipulation of PGI1 expression or activity could be particularly relevant in A. gossypii strains designed for heterologous protein production, balancing the metabolic requirements for growth with those needed for recombinant protein synthesis and secretion .
Several analytical methods provide robust assessment of recombinant PGI1 activity across diverse experimental settings. The standard spectrophotometric assay couples PGI1 activity with glucose-6-phosphate dehydrogenase, measuring NADPH formation at 340 nm as glucose-6-phosphate is oxidized. This method offers high sensitivity and is adaptable to plate reader formats for high-throughput screening. For more complex biological samples, activity staining following native PAGE separation can localize PGI1 activity while distinguishing it from endogenous isomerases. Advanced techniques include isothermal titration calorimetry (ITC) for detailed kinetic characterization and binding studies, and surface plasmon resonance (SPR) for real-time analysis of substrate interactions. In cellular contexts, metabolic flux analysis using 13C-labeled substrates can quantify carbon flux through the PGI1-catalyzed reaction. Differential scanning fluorimetry proves valuable for stability assessments under varying conditions. When selecting an analytical approach, researchers should consider sample complexity, required sensitivity, and the specific research questions being addressed . These methodologies collectively enable comprehensive characterization of recombinant PGI1 from fundamental enzymology to complex metabolic engineering applications.
Researchers working with recombinant PGI1 expression in A. gossypii may encounter several challenges that require systematic troubleshooting approaches. Low expression yields represent a common issue potentially attributable to codon usage differences, as demonstrated in comparative studies with heterologous proteins from T. reesei . Implementing codon optimization for the PGI1 gene sequence can significantly improve translation efficiency. Protein misfolding or aggregation may occur, especially with partial PGI1 constructs , necessitating optimization of growth temperatures (typically lowering to 25-27°C during induction) or co-expression of chaperone proteins. Proteolytic degradation poses another challenge, particularly given A. gossypii's secretory capabilities . Addressing this may require adding protease inhibitors to culture media, engineering protease-deficient host strains, or modifying culture conditions to minimize protease activation. Inconsistent glycosylation patterns can affect PGI1 activity and stability; researchers should consider characterizing glycosylation profiles using mass spectrometry and potentially employ glycosylation site mutagenesis if specific modifications prove detrimental . Finally, difficulties in protein purification may arise due to the partial nature of some PGI1 constructs , requiring optimization of purification protocols with alternative chromatography approaches or affinity tag systems specifically validated for A. gossypii-expressed proteins.
Optimizing storage and handling conditions for recombinant A. gossypii PGI1 is essential for maintaining enzymatic activity during research applications. According to product specifications, liquid formulations of recombinant PGI1 typically maintain stability for approximately 6 months when stored at -20°C/-80°C, while lyophilized preparations extend this shelf life to 12 months under the same temperature conditions . Prior to opening, vials should be briefly centrifuged to bring the contents to the bottom, minimizing protein loss . For reconstitution of lyophilized PGI1, deionized sterile water should be used to achieve a concentration of 0.1-1.0 mg/mL, with addition of glycerol (recommended final concentration 5-50%) as a cryoprotectant for long-term storage . Repeated freeze-thaw cycles significantly diminish enzyme activity and should be avoided; instead, preparing small working aliquots for storage at 4°C (stable for approximately one week) provides a practical approach for ongoing experiments . Buffer composition represents another critical factor, with phosphate buffers (pH 7.0-7.5) containing stabilizing agents like dithiothreitol (DTT) or β-mercaptoethanol helping maintain the enzyme's native conformation by preventing oxidation of cysteine residues. Activity assays should be performed periodically to monitor stability during long-term storage under various conditions.
Addressing batch-to-batch variability in recombinant A. gossypii PGI1 requires implementing comprehensive quality control procedures and standardized production protocols. Establishing a consistent seed culture preparation method represents a foundational step, with standardized media composition, inoculum size, and growth phase at harvest all contributing to reproducible starting conditions. Fermentation parameters should be precisely controlled through automated bioreactor systems that maintain constant temperature, pH, dissolved oxygen, and agitation profiles across production runs . Implementing real-time monitoring of critical process parameters allows for adaptive control strategies that respond to biological variability. Analytical characterization of each batch should include multiple quality attributes: purity assessment via SDS-PAGE (targeting >85% as specified for commercial preparations) , activity testing using standardized spectrophotometric assays, glycosylation profiling through mass spectrometry, and thermal stability analysis. Reference standards from well-characterized batches enable comparative analysis and calibration of activity measurements. Implementing design of experiments (DoE) approaches helps identify critical process parameters most strongly influencing final product quality, allowing focused optimization efforts. For research applications requiring exceptional consistency, consider implementing more stringent purification procedures that might sacrifice yield for improved homogeneity of the final enzyme preparation.
A. gossypii possesses several distinctive metabolic features that must be considered when designing experiments to study recombinant PGI1 function. The recent genome-wide metabolic re-annotation revealed significant differences between A. gossypii and related yeasts like S. cerevisiae and K. lactis in numerous pathways, including purine metabolism, nitrogen metabolism, and lipid metabolism . These differences directly influence carbon flux distribution patterns through central metabolism, where PGI1 functions as a critical node. A. gossypii's natural riboflavin overproduction capability creates a unique metabolic background, with enhanced flux through the pentose phosphate pathway competing with glycolysis for glucose-6-phosphate—the substrate of PGI1 . When designing in vivo experiments, researchers should account for the absence of certain metabolic routes present in model yeasts, such as the reported absence of the NADP+-dependent glutamate dehydrogenase ammonium assimilation route and the GABA route of glutamate degradation in A. gossypii . Additionally, A. gossypii's filamentous growth pattern introduces spatial considerations not present in unicellular yeasts, potentially creating microenvironments with varying metabolite concentrations that might affect local PGI1 activity . The observed sensitivity to low pH compared to most filamentous fungi further necessitates careful pH control in experimental systems .
Studies using recombinant PGI1 offer valuable perspectives on the evolutionary adaptation of A. gossypii metabolism. The genomic relationship between A. gossypii and S. cerevisiae presents an intriguing case study in evolutionary metabolic rewiring, as these organisms share high gene homology and synteny (91% of A. gossypii's genes are syntenic to S. cerevisiae genes) yet display distinct metabolic capabilities and growth patterns . Biochemical characterization of recombinant PGI1 can reveal how this key glycolytic enzyme may have adapted its kinetic properties to support A. gossypii's filamentous growth and natural riboflavin overproduction . Comparative analysis of substrate specificity, catalytic efficiency, allosteric regulation, and stability between A. gossypii PGI1 and homologs from related species can highlight molecular adaptations that contribute to metabolic specialization. The genome-wide metabolic re-annotation has already identified numerous enzymatic differences between A. gossypii and related yeasts, including several enzymes found exclusively in A. gossypii within purine metabolism—a pathway connected to riboflavin biosynthesis . PGI1's role at the branch point between glycolysis and the pentose phosphate pathway positions it as a potential control point in the metabolic adaptation toward riboflavin production, making it particularly relevant for evolutionary metabolic studies.
Protein engineering offers powerful approaches to enhance or modify recombinant A. gossypii PGI1 for specialized research applications. Rational design approaches, guided by structural knowledge and computational modeling, can target specific amino acid residues to alter substrate specificity, improve catalytic efficiency, or enhance stability under challenging experimental conditions. Site-directed mutagenesis of key catalytic residues might generate PGI1 variants with altered reaction kinetics useful for metabolic engineering applications seeking to redirect carbon flux at the glucose-6-phosphate node . For applications requiring improved thermal stability, consensus-based design approaches can incorporate stabilizing features from thermophilic PGI homologs. Domain-swapping experiments, particularly relevant for the partial PGI1 variants described in the search results, might generate chimeric enzymes with novel functionalities by combining domains from different species' homologs . To address the challenge of protein purification, fusion protein approaches incorporating affinity tags or solubility-enhancing partners can improve expression yield and simplify purification protocols. Directed evolution methodologies, while more labor-intensive, provide a complementary approach for generating and screening PGI1 variant libraries when targeting properties difficult to predict from structural information alone. These protein engineering strategies collectively expand the research utility of recombinant A. gossypii PGI1 beyond its native characteristics.
The next decade of recombinant A. gossypii PGI1 research will likely be transformed by several emerging technologies that enhance our understanding and application of this enzyme. CRISPR-Cas9 genome editing technologies, already revolutionizing fungal biotechnology, will enable precise genomic integration of modified PGI1 variants with minimal disruption to cellular metabolism . This approach will facilitate in vivo structure-function studies and metabolic engineering applications. Single-cell metabolomics techniques will provide unprecedented insights into how PGI1 activity varies across the mycelial network of A. gossypii, addressing questions about metabolic compartmentalization in filamentous fungi that have previously been inaccessible . Artificial intelligence and machine learning approaches will accelerate protein engineering efforts by improving prediction of how specific mutations affect PGI1 properties, enabling more efficient rational design strategies. Microfluidic cultivation systems combined with real-time activity monitoring will allow high-throughput screening of environmental conditions or genetic modifications affecting PGI1 function. Synthetic biology approaches, leveraging modular genetic parts optimized for A. gossypii, will facilitate precise control over PGI1 expression levels and timing . Cryo-electron microscopy advancements will enable higher-resolution structural studies of PGI1 in complex with various binding partners, potentially revealing previously uncharacterized regulatory interactions that influence central carbon metabolism in this biotechnologically important fungus.
Despite progress in understanding A. gossypii metabolism, several significant questions regarding PGI1 function remain unanswered. A fundamental question concerns the potential moonlighting functions of PGI1 beyond its catalytic role in glycolysis and gluconeogenesis. In other organisms, glucose-6-phosphate isomerase has been shown to function extracellularly as a cytokine-like factor—whether A. gossypii PGI1 possesses similar secondary functions remains unexplored. The regulatory mechanisms controlling PGI1 expression and activity in response to changing carbon sources or developmental stages of A. gossypii are poorly characterized, despite their potential importance for riboflavin production optimization . The spatial organization of glycolytic enzymes, including PGI1, within the elongated hyphal cells of A. gossypii represents another open question, with implications for understanding compartmentalized metabolism in filamentous fungi . The complete structure of A. gossypii PGI1 has not been determined, limiting structure-function analyses and rational engineering approaches. The evolutionary significance of subtle sequence variations between A. gossypii PGI1 and homologs from related yeasts remains unclear, particularly regarding their potential role in adapting central metabolism to support riboflavin overproduction . Finally, the potential applications of engineered PGI1 variants in redirecting carbon flux toward valuable products beyond riboflavin have not been systematically explored despite promising preliminary indications from metabolic re-annotation studies .
Advances in A. gossypii PGI1 research have implications extending well beyond this specific enzyme system into broader biotechnological fields. As a key branch-point enzyme between glycolysis and the pentose phosphate pathway, insights from PGI1 engineering could inform general strategies for redirecting central carbon metabolism in various industrial microorganisms . The lessons learned from expressing and characterizing recombinant PGI1 in A. gossypii contribute to our understanding of heterologous protein production in filamentous fungi, supporting broader efforts to develop these organisms as production platforms for industrial enzymes and biopharmaceuticals . A. gossypii's unique position as a natural riboflavin overproducer with potential for expressing heterologous proteins makes it a valuable model system for studying how central metabolism can be engineered to simultaneously support growth and product formation . The comprehensive metabolic re-annotation of A. gossypii, coupled with specific insights about PGI1 function, provides a blueprint for similar systematic analyses of other non-conventional microbial hosts with biotechnological potential . Methodologies developed for characterizing and engineering A. gossypii PGI1 can be adapted for other key metabolic enzymes, building toward integrated approaches for whole-cell biocatalysis applications. Finally, A. gossypii's evolutionary relationship with S. cerevisiae, combined with their distinct metabolic capabilities, makes comparative studies of their PGI1 enzymes valuable for understanding how metabolic networks evolve and adapt to different ecological niches or physiological requirements .
These technical specifications provide essential information for researchers planning experiments with recombinant A. gossypii PGI1, ensuring appropriate handling, storage, and application of this enzyme preparation. The partial nature of the recombinant protein should be carefully considered when designing experimental workflows, as this may affect certain structural or functional properties compared to the full-length native enzyme .