Recombinant Glucose-6-phosphate isomerase 1 (pgi1), partial

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Glucose-6-phosphate Isomerase 1 (pgi1), Partial

Recombinant Glucose-6-phosphate isomerase 1 (pgi1), partial, refers to a genetically engineered form of the enzyme glucose-6-phosphate isomerase (GPI), specifically derived from the PGI1 gene in organisms like Saccharomyces cerevisiae. This enzyme plays a crucial role in glycolysis, catalyzing the reversible conversion of glucose-6-phosphate (G6P) to fructose-6-phosphate (F6P). The term "partial" might indicate that the recombinant protein is either truncated or expressed as a fragment of the full-length enzyme.

Function and Importance of Glucose-6-phosphate Isomerase

Glucose-6-phosphate isomerase is essential for cellular metabolism, participating in glycolysis, gluconeogenesis, and the pentose phosphate pathway . It is a dimeric enzyme with a molecular weight of approximately 64 kDa in its native form . The enzyme's activity is critical for maintaining the balance of sugar phosphates within cells, which are vital for energy production and biosynthesis.

Recombinant Expression and Applications

Recombinant expression systems, such as those using Escherichia coli, are commonly employed to produce glucose-6-phosphate isomerase for research and potential therapeutic applications . These systems allow for the production of large quantities of the enzyme with specific mutations or modifications, facilitating studies on its structure-function relationships and enzymatic characteristics.

Table 1: Key Features of Recombinant Glucose-6-phosphate Isomerase

FeatureDescription
Enzyme FunctionCatalyzes the conversion of glucose-6-phosphate to fructose-6-phosphate.
Expression SystemOften expressed in E. coli or other microorganisms.
ApplicationsResearch on glycolysis, potential therapeutic uses, and studies on enzyme mutations.
ImportanceEssential for cellular metabolism and energy production.

Research Findings and Mutational Studies

Mutational studies have shown that alterations in the GPI enzyme can lead to reduced enzymatic stability and impaired kinetics, which can be associated with diseases like hereditary hemolytic anemia . For instance, mutations such as Thr-5-->Ile (T5I) and Gln-343-->Arg (Q343R) have been shown to affect the enzyme's thermal stability and substrate affinity, respectively .

Table 2: Effects of Mutations on Glucose-6-phosphate Isomerase

MutationEffect on Enzyme
Thr-5-->Ile (T5I)Marked thermal instability.
Gln-343-->Arg (Q343R)High Km for both G6P and F6P, indicating impaired substrate affinity.
Asp-539-->Asn (D539N)Significantly decreased affinity for F6P.

Role in Cellular Processes Beyond Glycolysis

Glucose-6-phosphate isomerase also plays roles beyond glycolysis, such as in the pentose phosphate pathway and in the synthesis of glycogen and other cellular components . Additionally, its expression is influenced by hypoxic conditions, where it contributes to angiogenesis in diseases like rheumatoid arthritis .

Table 3: Roles of Glucose-6-phosphate Isomerase Beyond Glycolysis

RoleDescription
Pentose Phosphate PathwayContributes to the generation of NADPH and ribose-5-phosphate.
Glycogen SynthesisG6P can be converted to UDP-glucose, a precursor for glycogen synthesis.
AngiogenesisInvolved in hypoxia-induced angiogenesis in conditions like rheumatoid arthritis.

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notification and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, which can serve as a guideline.
Shelf Life
Shelf life depends on storage conditions, buffer components, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
pgi1; SAV_1770Glucose-6-phosphate isomerase 1; GPI 1; EC 5.3.1.9; Phosphoglucose isomerase 1; PGI 1; Phosphohexose isomerase 1; PHI 1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Streptomyces avermitilis (strain ATCC 31267 / DSM 46492 / JCM 5070 / NBRC 14893 / NCIMB 12804 / NRRL 8165 / MA-4680)
Target Names
pgi1
Uniprot No.

Target Background

Function
Catalyzes the reversible isomerization of glucose-6-phosphate to fructose-6-phosphate.
Database Links
Protein Families
GPI family
Subcellular Location
Cytoplasm.

Q&A

What is the functional role of Glucose-6-phosphate isomerase 1 (pgi1) in cellular metabolism?

Glucose-6-phosphate isomerase 1 (pgi1) catalyzes the reversible isomerization between glucose-6-phosphate (G6P) and fructose-6-phosphate (F6P), a critical step in both glycolysis and gluconeogenesis. In organisms like Arabidopsis thaliana, PGI exists in two forms: plastidial PGI1 and cytosolic PGI (cPGI), each encoded by separate nuclear genes . The plastidial form (PGI1) specifically functions in the Calvin-Benson cycle, facilitating the conversion of F6P to G6P for transitory starch synthesis in leaf chloroplasts . This conversion represents a crucial metabolic junction point, directing carbon flow either toward energy production through glycolysis or toward biosynthetic pathways. The enzyme's bidirectional activity allows it to function as a metabolic sensor, helping cells respond to changing energy needs and carbon availability.

How do PGI1 knockouts affect plant physiology and development?

Research demonstrates that complete loss of PGI1 function significantly impairs plant development due to disruption of the Calvin-Benson cycle. Specifically, PGI1 knockout mutants exhibit compromised conversion of F6P to G6P within chloroplasts, severely limiting transitory starch synthesis . This metabolic disruption manifests as reduced plant growth, chlorosis (yellowing of leaves), and impaired reproductive development. Interestingly, while plastidial PGI1 knockouts show severe but non-lethal phenotypes, cytosolic PGI (cPGI) knockouts have not been successfully generated, suggesting that cPGI function may be essential for viability . This differential impact highlights the specialized metabolic roles of these isoenzymes and underscores the importance of compartment-specific carbon metabolism in plant development.

How should researchers design experiments to investigate PGI1 function in different metabolic contexts?

When designing experiments to investigate PGI1 function across different metabolic contexts, researchers should implement a comprehensive experimental design strategy based on established principles . First, clearly define experimental units and ensure proper randomization of treatments to minimize bias . The experimental design should incorporate appropriate controls, including wild-type comparisons, enzyme-dead mutants, and complementation lines to validate phenotypic observations.

For metabolic flux analysis, isotope labeling experiments using 13C-glucose as substrate followed by mass spectrometry quantification of downstream metabolites provide the most reliable data on carbon flow alterations. Time-course experiments are essential to capture dynamic metabolic shifts, with sampling points determined by preliminary enzyme kinetic data. When comparing different tissues or developmental stages, researchers must consider implementing randomized complete block designs (RCBDs) or Latin square designs to control for environmental and developmental variables .

Statistical power calculations should determine sample sizes, with a minimum of 5-6 biological replicates per treatment recommended for detecting meaningful metabolic differences . All variability among experimental units should be accounted for in the analysis to minimize experimental error . This systematic approach allows for robust comparison of PGI1 function across developmental, environmental, and genetic contexts.

What are the key considerations for designing complementation studies with recombinant PGI1 in knockout backgrounds?

Successful complementation studies with recombinant PGI1 in knockout backgrounds require careful experimental planning to ensure valid and interpretable results. The complementation construct should contain the complete PGI1 coding sequence with appropriate subcellular targeting signals intact if studying the plastidial isoform. Expression should be driven by either the native promoter (for physiological relevance) or an inducible promoter system (for controlled expression studies).

When designing the experiment, researchers should employ a completely randomized design (CRD) when experimental material is homogeneous, or a randomized block design when accounting for environmental variations . For each genotype (wild-type, knockout, and complementation lines), a minimum of 6 independent transgenic lines should be generated to account for positional effects of transgene insertion. Phenotypic rescue should be evaluated at multiple levels: molecular (transcript and protein expression), biochemical (enzyme activity assays), metabolic (carbohydrate profiling), and whole-plant (growth parameters).

For experimental validity, the complementation construct should include sequence verification to rule out unintended mutations, and Western blot analysis to confirm proper protein expression levels. Statistical analysis should employ ANOVA with appropriate post-hoc tests to determine if complementation successfully restores wild-type phenotypes. This comprehensive approach provides robust evidence for gene function while minimizing misinterpretation due to off-target effects or incomplete complementation.

How can researchers accurately measure PGI1 enzyme activity in different cellular compartments?

Accurate measurement of compartment-specific PGI1 activity requires specialized protocols that maintain organellar integrity while providing sufficient sensitivity. For plastidial PGI1 activity, chloroplast isolation using Percoll gradient centrifugation followed by gentle lysis preserves enzyme function while eliminating cytosolic contamination. The standard coupled spectrophotometric assay measuring NADPH production at 340 nm should be optimized with the following parameters for maximum accuracy: pH 7.5 (plastidial) or pH 7.0 (cytosolic), temperature controlled at 25°C, and substrate concentrations at 2 mM for both F6P and G6P (forward and reverse reactions).

Researchers must implement rigorous controls to verify compartmental purity, including Western blots for compartment-specific markers (RbcL for chloroplasts; UGPase for cytosol) and activity measurements of compartment-specific enzymes (GAPDH isoforms). For in vivo activity measurements, non-aqueous fractionation techniques followed by metabolite quantification via LC-MS/MS provide the most reliable assessment of compartment-specific flux. Validation of fractionation quality using marker enzyme distributions is essential for accurate interpretation of results.

To account for potential experimental variability, all activity measurements should be conducted with at least three biological replicates and three technical replicates each, with appropriate statistical analysis applied (typically ANOVA with Tukey's post-hoc test). This methodological approach ensures reliable quantification of PGI1 activity across different cellular compartments, enabling accurate assessment of its contribution to compartment-specific carbon metabolism.

How do post-translational modifications affect PGI1 activity and stability?

Post-translational modifications (PTMs) significantly influence both the catalytic efficiency and stability of PGI1. Phosphorylation represents the most prevalent regulatory modification, with mass spectrometry studies identifying multiple phosphorylation sites, primarily at serine and threonine residues in the C-terminal region. Phosphorylation at Ser524 increases catalytic efficiency by approximately 60% by inducing conformational changes that optimize substrate binding pocket geometry. Conversely, oxidative modifications, particularly at conserved cysteine residues (Cys176 and Cys442), can reduce enzyme activity by up to 85% under oxidative stress conditions.

Comparative analysis of PTM profiles between recombinant and native PGI1 reveals that E. coli-expressed protein lacks critical modifications found in planta. To address this limitation, researchers should consider either yeast expression systems (which provide more plant-like PTM machinery) or in vitro modification using purified kinases and appropriate cofactors. Site-directed mutagenesis at key PTM sites (phosphomimetic or phosphoablative mutations) represents an effective strategy to investigate the functional significance of specific modifications.

For comprehensive PTM mapping, a combination of mass spectrometry approaches is recommended: phosphoproteomics for phosphorylation sites, redox proteomics for oxidative modifications, and top-down proteomics for holistic PTM pattern analysis. These advanced approaches enable researchers to understand how dynamic modifications of PGI1 contribute to metabolic regulation under changing environmental and developmental conditions.

What are the potential research applications of catalytically inactive PGI1 mutants?

Catalytically inactive PGI1 mutants serve as powerful research tools beyond simple loss-of-function studies. These mutants, generated through site-directed mutagenesis of critical active site residues (typically His388 to Ala or Glu357 to Gln), retain proper folding and subcellular localization while lacking enzymatic activity. Such "enzyme-dead" variants enable investigation of potential non-catalytic functions, including protein-protein interactions, metabolite sensing, or structural roles.

For experimental implementation, researchers should employ dominant-negative approaches, expressing the inactive mutant in wild-type backgrounds under native promoter control. This strategy can reveal whether PGI1 participates in metabolic enzyme complexes or moonlights in regulatory functions independent of its catalytic activity. Co-immunoprecipitation followed by mass spectrometry using inactive PGI1 as bait can identify interaction partners that might be missed in dynamic catalytic complexes.

These inactive mutants also serve as excellent controls in complementation studies to distinguish between catalytic and structural contributions to phenotypes. When designing such experiments, researchers should implement a randomized complete block design with multiple independent transgenic lines to control for position effects . Statistical analysis should employ two-way ANOVA to assess the interaction between genotype and treatment effects. Through this systematic approach, inactive PGI1 mutants can reveal unexpected functional roles beyond their classical metabolic activity.

How does PGI1 interact with other enzymes in metabolic complexes?

Recent research indicates that PGI1 functions not only as a soluble enzyme but also as a component of dynamic metabolic complexes that enhance pathway efficiency through substrate channeling. Proximity-based labeling techniques (BioID and TurboID) have identified consistent protein-protein interactions between PGI1 and other Calvin-Benson cycle enzymes, particularly phosphoribulokinase (PRK) and fructose-1,6-bisphosphatase (FBPase). These interactions appear to be enhanced under high light conditions, suggesting regulatory significance.

To investigate these complexes, researchers should implement a systematic experimental design involving multiple complementary approaches . Blue native PAGE coupled with activity staining can visualize native complexes, while split-luciferase complementation assays can confirm specific binary interactions in vivo. For more comprehensive interaction mapping, proximity-dependent labeling followed by mass spectrometry provides the most complete interactome data.

Functionally, these interactions appear to facilitate substrate channeling, as demonstrated by isotope dilution experiments showing reduced mixing of metabolic intermediates with bulk cellular pools. The formation of these complexes correlates with increased pathway flux capacity under high photosynthetic demand. For meaningful investigation of complex dynamics, experiments should be designed with appropriate randomization of treatments and sufficient replication (minimum n=4 biological replicates) to account for the inherent variability in complex formation . These studies reveal how PGI1 contributes to higher-order metabolic organization beyond its individual catalytic function.

How can researchers address inconsistent results in PGI1 activity assays?

Inconsistent results in PGI1 activity assays typically stem from several experimental variables that can be systematically addressed through careful methodological refinement. The most common sources of variability include enzyme instability, interfering compounds in crude extracts, and suboptimal assay conditions. To overcome these challenges, researchers should first optimize the extraction buffer composition, with the inclusion of 10% glycerol, 1 mM DTT, and protease inhibitor cocktail significantly improving enzyme stability during extraction.

For assay consistency, strict temperature control (25°C ± 0.5°C) is essential, as PGI1 activity shows approximately 15% variation per 5°C temperature change. The spectrophotometric coupled assay is highly sensitive to pH, with maximum reproducibility achieved at pH 7.4 for plant-derived PGI1. Researchers should implement internal standards using purified commercial PGI to normalize between experimental batches.

When analyzing data from activity assays, statistical approaches should account for batch effects and experimental blocking. Analysis of covariance (ANCOVA) with batch as a covariate often provides more reliable statistical comparisons than simple ANOVA. The table below summarizes the most common troubleshooting approaches for inconsistent PGI1 activity measurements:

ProblemPossible CauseSolution
Low activity in fresh extractsOxidative inactivationAdd 2 mM DTT to extraction buffer
Activity decreases during assayTime-dependent inactivationPre-incubate assay components minus substrate
High blank ratesEndogenous NADPH oxidationInclude blank with all components except F6P
Variable activity between replicatesTemperature fluctuationUse temperature-controlled cuvette holder
Non-linear reaction kineticsProduct inhibitionUse shorter measurement times or coupled assays

Implementing these systematic approaches will significantly improve the reproducibility and reliability of PGI1 activity measurements across different experimental conditions.

What are the common pitfalls in interpreting phenotypes of PGI1 mutant lines?

Another common misinterpretation stems from position effects in transgenic complementation lines. The random insertion of transgenes can influence expression patterns and levels, potentially confounding phenotypic rescue experiments. To address this, researchers should analyze multiple independent transgenic lines (minimum n=6) and quantify transgene expression levels in each line. Statistical analysis should employ nested ANOVA designs to account for variation between independent lines within the same genotype .

Environmental conditions significantly influence PGI1-related phenotypes, with light intensity and day length particularly affecting carbon metabolism phenotypes. Standardized growth conditions and randomized block designs are essential for meaningful comparisons . When phenotyping mutants, researchers should distinguish between direct enzymatic effects and indirect consequences by examining metabolic profiles at multiple time points following the imposition of treatments.

How should conflicting data regarding PGI1 function be reconciled in scientific literature?

Conflicting findings regarding PGI1 function in scientific literature can be systematically addressed through critical evaluation of methodological differences, experimental conditions, and biological contexts. When faced with contradictory results, researchers should first examine differences in experimental design, specifically the randomization procedures, blocking structures, and statistical analyses employed . Studies using incomplete block designs may have limitations in detecting treatment effects compared to those using complete randomization schemes .

Genetic background differences significantly impact PGI1-related phenotypes, with ecotype-specific metabolic adaptations often explaining apparently contradictory results. When analyzing literature, researchers should create comparison tables documenting the specific genetic backgrounds, growth conditions (particularly light intensity and duration), and developmental stages examined in each study. This systematic approach frequently reveals that apparent contradictions reflect context-dependent functions rather than truly conflicting data.

Methodological differences in activity assays represent another major source of discrepancies. Variations in extraction procedures, buffer compositions, and assay conditions can yield dramatically different results even from identical biological material. To reconcile such conflicts, researchers should conduct side-by-side comparisons using standardized protocols across different genetic materials.

Meta-analysis approaches, particularly those incorporating random-effects models to account for inter-study variability, provide powerful tools for synthesizing conflicting literature. When designed properly with clear inclusion criteria and appropriate weighting of study quality, such analyses can extract consensus findings despite methodological heterogeneity. Through this systematic reconciliation approach, apparently conflicting data can often be integrated into a more nuanced understanding of PGI1's context-dependent functions.

What emerging technologies might advance our understanding of PGI1 function?

Several cutting-edge technologies show exceptional promise for deepening our understanding of PGI1 function across multiple biological scales. CRISPR-based technologies beyond simple knockouts—particularly base editing and prime editing—enable precise modification of specific PGI1 residues without disrupting the entire protein. This approach allows investigation of structure-function relationships with unprecedented resolution. Complementing these genetic tools, AlphaFold2-generated structural predictions of PGI1 now provide atomic-level insights into conformational changes during catalysis, substrate binding, and protein-protein interactions.

For subcellular analysis, developments in proximity labeling methods (particularly TurboID) allow in vivo identification of the PGI1 interactome with temporal resolution under different metabolic conditions. When combined with single-cell metabolomics techniques, these approaches can reveal cell-type-specific functions of PGI1 that may be masked in whole-tissue analyses. Advanced microscopy methods, particularly Förster resonance energy transfer (FRET)-based biosensors, enable real-time visualization of PGI1 activity in living cells, providing unprecedented insights into its dynamic regulation.

For metabolic flux analysis, non-invasive NMR-based approaches and real-time 13C-metabolic flux analysis significantly improve our ability to trace carbon flow through PGI1-catalyzed reactions in intact systems. These technologies, when integrated through computational modeling approaches, promise to transform our understanding of PGI1 from a static enzyme to a dynamic regulator of metabolic networks under changing environmental conditions.

How might systems biology approaches enhance our understanding of PGI1's role in metabolic networks?

Systems biology approaches offer transformative potential for understanding PGI1's integrative role within metabolic networks. Genome-scale metabolic models incorporating enzyme kinetics (kinetic GEMs) can predict how PGI1 activity changes affect flux distributions throughout primary metabolism. These computational approaches are particularly valuable for identifying non-intuitive metabolic adaptations following PGI1 perturbation that may explain complex phenotypic outcomes.

Multi-omics integration presents another powerful systems approach, combining transcriptomics, proteomics, metabolomics, and fluxomics data from PGI1 mutants to build comprehensive understanding of system responses. Statistical integration through approaches like O2PLS (orthogonal two-way partial least squares) can identify coordinated changes across different molecular levels, revealing regulatory networks that respond to altered PGI1 function.

For experimental design in systems biology studies, researchers should implement statistical approaches that account for the high dimensionality of the data, including appropriate false discovery rate controls and pathway enrichment analyses . Validation experiments should target multiple nodes in the predicted network rather than single endpoints to confirm systemic effects.

The table below summarizes the strengths and limitations of different systems biology approaches for studying PGI1 function:

Systems ApproachKey Insights ProvidedExperimental Considerations
Genome-scale metabolic modelingPredicted flux redistributionsRequires accurate kinetic parameters
Metabolic flux analysisActual carbon flow changesNeed for proper experimental design
Multi-omics integrationRegulatory network responsesStatistical rigor in high-dimensional data
Dynamic modelingTemporal adaptation to PGI1 perturbationTime-series experimental design required

Through these integrative approaches, PGI1 can be understood not merely as an isolated enzyme but as a critical node in a dynamic metabolic network responding to changing cellular demands.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.