Recombinant Xenopus laevis GTP cyclohydrolase 1 feedback regulatory protein (gchfr)

Shipped with Ice Packs
In Stock

Description

Function of GCHFR in Mammalian Systems

In mammals, GCHFR binds to GCH1 and modulates its activity based on the cellular concentrations of tetrahydrobiopterin (BH4) and phenylalanine. This regulatory mechanism ensures that BH4 levels remain within a narrow range, which is crucial for maintaining proper neurotransmitter synthesis and nitric oxide production . The interaction between GCH1 and GFRP is highly specific, with GFRP forming inhibited or activated complexes with GCH1 depending on the presence of BH4 or phenylalanine .

Potential Role of Recombinant Xenopus laevis GCHFR

While there is no specific literature on recombinant Xenopus laevis GCHFR, understanding its potential role requires examining the function of GCHFR in other systems. In Xenopus laevis, GCHFR could theoretically play a similar role in regulating GCH1 activity, thus influencing BH4 biosynthesis and related physiological processes. This would be consistent with the conserved nature of many biochemical pathways across species.

Research Findings on GCH1 and GFRP Interactions

Studies on mammalian GCH1 and GFRP have provided detailed insights into their interaction dynamics. For instance, the presence of phenylalanine enhances the binding affinity between GCH1 and GFRP, leading to increased BH4 production . Additionally, phosphorylation of GCH1 can alter its interaction with GFRP, affecting enzyme activity and BH4 levels .

Data Tables: Comparison of GCH1 and GFRP Interactions

ParameterDescriptionEffect on GCH1 Activity
BH4 PresenceInhibits GCH1 via GFRP bindingDecreases activity
Phenylalanine PresenceEnhances GCH1-GFRP bindingIncreases activity
GCH1 PhosphorylationReduces GFRP binding affinityIncreases activity

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires advance notice 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 collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and serves as a guideline.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms maintain stability for 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. If you require a specific tag, please inform us; we will prioritize its development.
Synonyms
gchfr; GTP cyclohydrolase 1 feedback regulatory protein; GFRP; GTP cyclohydrolase I feedback regulatory protein
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-84
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Xenopus laevis (African clawed frog)
Target Names
gchfr
Target Protein Sequence
MPYVLISTQI RMETGPTIVG DEFSDLVLMA QLEADKRTVL GNNFSEYYVN EPPRVTLNKL ERLGYRVVSM TGVGQTLVWC LHKE
Uniprot No.

Target Background

Function
Mediates the inhibition of GTP cyclohydrolase 1 by tetrahydrobiopterin.
Database Links

KEGG: xla:444531

UniGene: Xl.19284

Protein Families
GFRP family
Subcellular Location
Nucleus. Nucleus membrane. Cytoplasm, cytosol.

Q&A

What is GTP Cyclohydrolase 1 Feedback Regulatory Protein (GCHFR) and why is it significant in Xenopus laevis research?

GCHFR is a regulatory protein that modulates the activity of GTP cyclohydrolase 1 (GCH1), the rate-limiting enzyme in tetrahydrobiopterin (BH4) biosynthesis. BH4 serves as an essential cofactor for nitric oxide synthase, aromatic amino acid hydroxylases, and alkylglycerol mono-oxygenase, making it crucial for numerous physiological processes . The GCHFR-GCH1 complex is subject to allosteric regulation: BH4 mediates feedback inhibition while L-phenylalanine (L-phe) provides feed-forward activation of GCH1 activity .

Xenopus laevis represents an excellent model system for studying GCHFR due to several advantageous characteristics. As an amphibian that occupies a phylogenetically intermediate position between aquatic vertebrates and land tetrapods, Xenopus allows researchers to distinguish between species-specific adaptations and evolutionarily conserved features of the BH4 regulatory system . The externally developing embryos are free from direct maternal influences, enabling precise manipulations and observations throughout development . Additionally, Xenopus undergoes thyroid-dependent metamorphosis, providing a unique opportunity to study GCHFR regulation in the context of dramatic developmental transitions .

The fully sequenced Xenopus genome exhibits remarkable structural similarity to the human genome, facilitating comparative studies and translational applications . This combination of features makes Xenopus laevis GCHFR research particularly valuable for understanding fundamental mechanisms of BH4 regulation that may be relevant to human health and disease.

How does the GCH1-GFRP protein complex form and what are its structural characteristics?

The GCH1-GFRP complex exhibits a distinctive structure where GCH1 exists as a homodecamer (approximately 280 kDa) sandwiched between two GFRP homopentamers (approximately 50 kDa each) . This arrangement creates a unique regulatory architecture that facilitates allosteric control of GCH1 activity. Biophysical characterization using surface plasmon resonance (SPR) has revealed important insights into the interaction dynamics between these proteins.

Crystal structure studies have identified discrete binding pockets for regulatory molecules at the GCH1-GFRP interface. L-phenylalanine and BH4 bind to distinct sites located at this interface, separate from the GTP substrate binding site on GCH1 . Upon binding to their respective pockets, these regulatory molecules induce conformational changes that are transmitted to the GTP binding site, thereby modulating enzyme activity allosterically .

The N-terminal region of GCH1 has been a subject of debate regarding its role in complex formation. While some studies using truncated GCH1 (lacking the first 42 amino acids) suggest this region doesn't influence GFRP binding or regulation, other research indicates the complete N-terminal region may be important for optimal GCH1-GFRP interactions and subsequent regulatory functions .

What expression systems are most effective for producing functional recombinant Xenopus laevis GCHFR?

The choice of expression system for recombinant Xenopus laevis GCHFR production depends on research objectives, required protein characteristics, and downstream applications. Each system offers distinct advantages and limitations that must be carefully considered.

Bacterial Expression Systems

Escherichia coli remains the most commonly used platform for initial GCHFR expression due to its simplicity and high yield. For optimal results, consider these methodological approaches:

  • Vector selection: pET vectors with N-terminal His-tags facilitate purification while minimizing interference with GCHFR function

  • Expression conditions: Induction at lower temperatures (16-18°C) for extended periods (16-24 hours) often improves solubility

  • Codon optimization: Adjusting codons to match E. coli preferences can significantly enhance expression levels

  • Fusion partners: SUMO or thioredoxin tags can improve solubility while maintaining native structure

Bacterial systems are particularly suitable for structural studies and initial protein-protein interaction assays that do not require post-translational modifications.

Insect Cell Expression Systems

Baculovirus-infected insect cells (Sf9 or High Five) offer an excellent compromise between proper eukaryotic processing and reasonable yields:

  • Superior folding environment for complex proteins like GCHFR

  • Proper formation of disulfide bonds and basic post-translational modifications

  • Higher expression levels compared to mammalian systems

  • Scalable production capabilities for larger quantities

This system is recommended for functional studies requiring properly folded GCHFR with basic eukaryotic modifications.

Mammalian Expression Systems

For studies focusing on regulatory functions that might depend on specific post-translational modifications:

  • HEK293 or CHO cells provide the most physiologically relevant modifications

  • Transient transfection for rapid screening or stable cell lines for consistent production

  • Secretion strategies using appropriate signal sequences can facilitate purification

  • Inducible expression systems allow control over timing and expression levels

While mammalian systems produce the most native-like GCHFR, they typically yield lower quantities at higher cost.

For most applications studying Xenopus laevis GCHFR, the insect cell expression system offers the best balance between proper folding, post-translational modification, and practical yield considerations. This approach is particularly valuable for functional studies examining the regulatory interactions between GCHFR and GCH1.

How can researchers verify the functional activity of recombinant Xenopus laevis GCHFR?

Verification of recombinant Xenopus laevis GCHFR functional activity requires a multi-faceted approach that assesses both binding capacity and regulatory function. The following methodological strategy provides a comprehensive validation framework:

Structural Integrity Assessment

Begin with basic characterization to confirm properly folded protein:

  • Circular dichroism (CD) spectroscopy to verify secondary structure composition

  • Size-exclusion chromatography to confirm the expected pentameric state

  • Thermal shift assays to evaluate protein stability and the effects of binding partners

Binding Interaction Analysis

Quantify the physical interaction with GCH1 using complementary techniques:

  • Surface plasmon resonance (SPR): Provides real-time, label-free measurement of association and dissociation kinetics. Based on previous research, expect very slow association and dissociation rates for the GCH1-GFRP complex . The presence of L-phenylalanine should enhance binding affinity approximately eightfold .

  • Isothermal titration calorimetry (ITC): Determines thermodynamic parameters (ΔH, ΔS, ΔG) and binding stoichiometry, confirming the expected pentamer-decamer arrangement.

  • Co-immunoprecipitation: Validates interaction in more complex protein mixtures.

Functional Regulation Assessment

The definitive test for GCHFR functionality is its ability to modulate GCH1 enzymatic activity:

  • Spectrophotometric GCH1 activity assays: Monitor the formation of dihydroneopterin triphosphate at 330 nm in the presence and absence of GCHFR.

  • Response to regulatory molecules: Test L-phenylalanine activation and BH4 inhibition of the GCH1-GFRP complex. L-phenylalanine should reverse BH4-mediated inhibition in a concentration-dependent manner.

  • HPLC or LC-MS/MS quantification: Measure BH4 production in reconstituted enzyme systems with and without GCHFR and regulatory molecules.

Comparative Validation

Compare the properties of Xenopus GCHFR with well-characterized mammalian counterparts:

  • Binding kinetics and regulatory responses should follow similar patterns, though species-specific differences in affinity and magnitude may exist.

  • Cross-species complementation experiments can reveal functional conservation and divergence.

A functional recombinant Xenopus laevis GCHFR should: (1) form a stable complex with GCH1, (2) enhance this interaction in the presence of L-phenylalanine, (3) mediate BH4-dependent feedback inhibition of GCH1 activity, and (4) facilitate L-phenylalanine-dependent reversal of this inhibition. These properties collectively confirm both structural integrity and proper regulatory function.

What are the key experimental considerations for studying L-phenylalanine effects on the GCH1-GFRP complex?

L-phenylalanine (L-phe) serves as a critical allosteric activator of the GCH1-GFRP complex, doubling maximal binding and enhancing binding affinity approximately eightfold . When designing experiments to investigate this regulatory mechanism in Xenopus laevis, several methodological considerations are essential:

Biophysical Interaction Studies

When examining direct protein-protein interactions and conformational changes:

  • Surface Plasmon Resonance (SPR) Optimization: Given the slow association and dissociation rates of the GCH1-GFRP complex , extended run times are necessary for accurate kinetic determination. Use L-phe concentration series (typically 0-500 μM) in the running buffer to generate dose-response curves.

  • Isothermal Titration Calorimetry (ITC): Provides thermodynamic parameters of binding that complement kinetic data from SPR. Compare binding profiles with and without L-phe to quantify energetic contributions of the allosteric effect.

  • Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS): Maps conformational changes induced by L-phe binding, revealing which regions of both proteins undergo structural rearrangements.

Functional Enzyme Assays

For assessing the biological consequences of L-phe regulation:

  • GCH1 Activity Measurements: Use spectrophotometric assays monitoring dihydroneopterin triphosphate formation or coupled enzyme systems. Include concentration series of L-phe (0-500 μM) to establish EC50 values.

  • Competition Experiments: Test if BH4-mediated inhibition can be reversed by L-phe in a concentration-dependent manner, establishing the interplay between feedback inhibition and feed-forward activation.

  • Comparative Analysis: Include mammalian GCH1-GFRP complexes as reference standards, as their responses to L-phe have been well characterized .

In Vivo and Ex Vivo Studies

For translating molecular findings to biological context:

  • L-phenylalanine Challenge Studies: Adapt the approach used in mouse studies (100 mg·kg⁻¹, p.o.) to Xenopus models, with appropriate dose adjustments. Measure tissue BH4 levels over time to capture both immediate and sustained responses.

  • Tissue-Specific Analysis: In mice, L-phe challenge induced sustained elevation of aortic BH4, an effect absent in endothelial GCH1-deficient animals . Similar tissue-specific analyses in Xenopus would reveal conserved regulatory mechanisms.

  • Developmental Stage Considerations: L-phe effects may vary across different developmental stages, particularly during metamorphosis when significant physiological remodeling occurs .

Controls and Validation

For ensuring experimental rigor:

  • Protein Variant Controls: Include both full-length and truncated GCH1 variants, as the N-terminal region may influence regulation .

  • Concentration Range Verification: Ensure L-phe concentrations span physiologically relevant ranges while including higher concentrations to establish saturation points.

  • Specificity Testing: Include structural analogs of L-phe to confirm binding pocket specificity.

Through systematic application of these methodological approaches, researchers can comprehensively characterize how L-phenylalanine modulates the GCH1-GFRP complex in Xenopus laevis and compare these findings to mammalian systems to identify conserved regulatory mechanisms.

How do developmental stages affect GCHFR expression and function in Xenopus laevis?

The developmental regulation of GCHFR expression and function in Xenopus laevis presents a fascinating research avenue that intersects developmental biology, regulatory biochemistry, and evolutionary comparative physiology. When investigating stage-specific patterns, researchers should implement the following methodological approaches:

Expression Analysis Across Developmental Stages

To establish the temporal pattern of GCHFR expression:

  • RT-qPCR Optimization: For accurate quantification, use validated reference genes specific to Xenopus developmental stages. Research has identified optimal reference gene pairs for different developmental contexts, including early whole embryos, brains during metamorphosis, and adult tissues .

  • Developmental Sampling Strategy: Include key developmental transitions:

    • Early cleavage and blastula stages (NF stages 1-9)

    • Gastrulation and neurulation (NF stages 10-20)

    • Organogenesis (NF stages 21-45)

    • Early metamorphosis (NF stages 46-54)

    • Metamorphic climax (NF stages 55-65)

    • Juvenile and adult stages

  • Western Blot Analysis: Complement mRNA data with protein expression profiles to identify potential post-transcriptional regulation. Use carefully titrated antibodies with validated specificity for Xenopus GCHFR.

Spatial Distribution Analysis

To determine tissue-specific expression patterns:

  • In Situ Hybridization: Employ whole-mount techniques for early embryos and section-based approaches for later stages. This reveals spatial regulation that may not be apparent in whole-embryo analyses.

  • Immunohistochemistry: Map GCHFR protein localization in tissues, with particular focus on the nervous system, liver, and vascular tissues where BH4 plays critical roles.

  • Co-localization Studies: Determine if GCHFR and GCH1 are expressed in the same tissues and cell types throughout development.

Functional Analysis in Development

To assess the biological significance of GCHFR across development:

  • Morpholino Knockdown Experiments: Target GCHFR at specific developmental windows to assess stage-specific requirements.

  • CRISPR/Cas9 Genetic Modification: Generate GCHFR mutant lines to study complete loss of function across all developmental stages.

  • BH4 Metabolite Profiling: Quantify BH4 levels across development using HPLC or LC-MS/MS, correlating with GCHFR expression patterns.

  • L-phenylalanine Challenge Studies: Administer L-phe at different developmental stages to test stage-specific responsiveness of the GCH1-GFRP regulatory system. Previous research in mice demonstrated that L-phe induces sustained elevation of tissue BH4 levels .

Thyroid Hormone Regulation

Since Xenopus metamorphosis is thyroid-dependent, examine the relationship between thyroid signaling and GCHFR:

  • T3/T4 Treatment Experiments: Assess GCHFR expression in response to thyroid hormone administration or inhibition.

  • Correlation with Deiodinase Expression: Analyze potential co-regulation with deiodinases (dio1, dio2, dio3) and thyroid hormone receptors (tr-alpha, tr-beta) .

This systematic developmental analysis will provide critical insights into how the BH4 regulatory system evolves throughout development and metamorphosis, potentially revealing stage-specific requirements for GCHFR function in this important model organism.

What methodological approaches can distinguish between mammalian and Xenopus laevis GCHFR functional properties?

Comparative analysis between mammalian and Xenopus laevis GCHFR provides valuable evolutionary insights into conserved and divergent aspects of BH4 regulation. The following methodological framework enables rigorous cross-species comparison:

Sequence and Structural Analysis

Begin with computational comparisons to identify potentially significant differences:

  • Sequence Alignment and Phylogenetic Analysis: Calculate percent identity and similarity between Xenopus and mammalian GCHFR, with special attention to binding interfaces and regulatory sites.

  • Homology Modeling: Generate structural models of Xenopus GCHFR based on available mammalian crystal structures, highlighting potentially significant structural divergences.

  • Binding Site Conservation Analysis: Examine conservation of the L-phenylalanine and BH4 binding pockets at the GCH1-GFRP interface, as these discrete binding sites have been characterized in mammalian structures .

Comparative Binding Kinetics

Directly compare protein-protein interaction parameters:

  • Parallel SPR Studies: Conduct side-by-side binding experiments under identical conditions with Xenopus and mammalian proteins. Key parameters to compare include:

    • Association rates (ka)

    • Dissociation rates (kd)

    • Equilibrium dissociation constants (KD)

    • Maximum binding capacity (Rmax)

  • L-phenylalanine Dose-Response: Compare the enhancement of binding affinity induced by L-phe. In mammalian systems, L-phe doubles maximal binding and enhances binding affinity approximately eightfold .

  • Cross-Species Complex Formation: Test if Xenopus GCHFR can interact with mammalian GCH1 and vice versa, determining the evolutionary compatibility of these regulatory components.

Functional Regulation Comparison

Assess similarities and differences in enzymatic regulation:

  • Parallel Enzyme Assays: Compare how Xenopus and mammalian GCHFR modulate their respective GCH1 enzymes under identical conditions.

  • Regulatory Molecule Sensitivity: Determine EC50 values for L-phenylalanine activation and IC50 values for BH4 inhibition across species.

  • Temperature Dependence Studies: Given the different physiological temperature ranges of amphibians and mammals, examine how temperature affects complex formation and regulation in both systems.

Chimeric Protein Approach

Determine which protein regions account for functional differences:

  • Domain Swapping Experiments: Create chimeric proteins containing regions from both Xenopus and mammalian GCHFR to identify domains responsible for species-specific properties.

  • Site-Directed Mutagenesis: Target non-conserved residues at binding interfaces to convert Xenopus-specific properties to mammalian-like behaviors and vice versa.

Cellular Context Comparison

Examine species differences in cellular environments:

  • Heterologous Expression: Express Xenopus GCHFR in mammalian cells and mammalian GCHFR in Xenopus cells, testing if cellular context affects function.

  • BH4 Production Capacity: Measure the maximum BH4 production capacity in comparable tissues from both species under basal and stimulated (L-phe) conditions.

This comprehensive comparative approach will reveal evolutionary conservation and divergence in this important regulatory system, potentially identifying adaptations specific to amphibian physiology versus mammalian homeostasis.

How should researchers design in vivo experiments to study GCHFR function in Xenopus laevis?

Designing robust in vivo experiments to study GCHFR function in Xenopus laevis requires careful consideration of this model organism's unique advantages while implementing appropriate controls and measurement techniques. The following methodological framework provides a comprehensive approach:

Genetic Manipulation Strategies

  • CRISPR/Cas9 Gene Editing:

    • Generate GCHFR knockout lines through targeted mutagenesis

    • Create knock-in lines with fluorescently tagged GCHFR for live imaging

    • Develop tissue-specific knockout models similar to the GCH1(fl/fl)-Tie2Cre mice used in mammalian studies

  • Morpholino Antisense Oligonucleotides:

    • For stage-specific and transient knockdown of GCHFR

    • Include dose-response characterization to achieve partial knockdown

    • Always include control morpholinos with similar chemical properties

  • mRNA Microinjection:

    • For rescue experiments and overexpression studies

    • Use region-specific injections to target specific tissues during early development

    • Include appropriate dosage controls and non-functional mutant versions

L-phenylalanine Challenge Studies

Adapt the approach used in mouse studies to the Xenopus system:

  • Administration Routes:

    • Early stages: Direct addition to culture medium

    • Later stages: Intraperitoneal injection or oral gavage

    • Consider size-adjusted dosing (equivalent to 100 mg·kg⁻¹ used in mice)

  • Time Course Analysis:

    • Measure BH4 levels at multiple timepoints (0, 1, 3, 6, 24 hours post-administration)

    • In mice, L-phe induced sustained elevation of aortic BH4 , so extended timepoints are important

  • Tissue-Specific Analysis:

    • Focus on tissues with high BH4 requirements (brain, liver, vasculature)

    • Compare responses across developmental stages

Tetrahydrobiopterin (BH4) Quantification

Accurate measurement of BH4 is critical for assessing GCHFR function:

  • HPLC with Electrochemical or Fluorescence Detection:

    • Distinguish BH4 from oxidized biopterins

    • Include internal standards for quantification

    • Ensure proper sample preservation to prevent oxidation

  • LC-MS/MS Methods:

    • Higher specificity and sensitivity

    • Ability to simultaneously measure multiple pterin metabolites

    • Quantify both total biopterins and specific BH4 levels

  • Tissue Preparation Considerations:

    • Immediate processing or flash-freezing to prevent BH4 oxidation

    • Acidic extraction conditions to stabilize BH4

    • Antioxidant addition (e.g., dithioerythritol) during processing

Physiological Function Assessment

Connect GCHFR-dependent BH4 regulation to downstream physiological processes:

  • Nitric Oxide Production:

    • Measure nitric oxide metabolites (nitrites/nitrates)

    • Use NO-sensitive fluorescent dyes in live imaging

    • Assess vascular development and function

  • Neurotransmitter Synthesis:

    • Quantify catecholamines and serotonin levels

    • Behavioral assays to assess neurological function

    • Analyze impact on neural development

Experimental Design Table

Experimental GroupGenetic ManipulationL-phe ChallengeExpected OutcomeControl Purpose
Wild-typeNoneNoBaseline BH4 levelsReference point
Wild-typeNoneYesElevated BH4 levelsL-phe response
GCHFR knockoutComplete GCHFR KONoAltered baselineGCHFR necessity
GCHFR knockoutComplete GCHFR KOYesReduced/absent L-phe responseL-phe mechanism
GCHFR rescueKO + WT GCHFR mRNAYesRestored L-phe responseSpecificity confirmation
GCHFR mutantKO + mutant GCHFR mRNAYesAltered responseStructure-function
GCH1 knockoutGCH1 KO, WT GCHFRYesNo BH4 increasePathway validation

This comprehensive methodological approach will provide robust insights into GCHFR function in vivo, establishing its role in regulating BH4 biosynthesis and downstream physiological processes throughout Xenopus development.

What techniques can researchers use to study post-translational modifications of GCHFR in Xenopus laevis?

Post-translational modifications (PTMs) can significantly impact GCHFR function, altering its binding properties, regulatory capacity, and cellular localization. Investigating these modifications in Xenopus laevis requires specialized techniques and careful experimental design:

Identification of PTMs

  • Mass Spectrometry-Based Proteomics:

    • Employ LC-MS/MS with higher-energy collisional dissociation (HCD) or electron transfer dissociation (ETD) fragmentation

    • Compare PTM profiles across different developmental stages and tissues

    • Use enrichment strategies for specific modifications (phosphopeptide enrichment, immunoprecipitation with PTM-specific antibodies)

  • Targeted PTM Mapping:

    • Focus on evolutionarily conserved sites identified in mammalian systems

    • Develop multiple reaction monitoring (MRM) assays for quantitative analysis of specific modifications

    • Compare modification patterns between recombinant and endogenous GCHFR

  • Western Blotting with PTM-Specific Antibodies:

    • Use phospho-specific, acetyl-specific, or other PTM-specific antibodies

    • Validate antibody specificity with appropriate controls (dephosphorylated samples, acetylase/deacetylase treatments)

    • Compare modification levels across developmental stages and in response to stimuli

Functional Impact Assessment

  • Site-Directed Mutagenesis:

    • Generate phosphomimetic mutations (S/T → D/E) to simulate constitutive phosphorylation

    • Create phospho-null mutations (S/T → A) to prevent phosphorylation

    • Test the effects of these mutations on GCH1 binding and regulatory function

  • In Vitro Enzymatic Modification:

    • Treat purified recombinant GCHFR with specific kinases, phosphatases, acetylases, or deacetylases

    • Compare binding properties and regulatory function before and after enzymatic treatment

    • Identify which enzymes can modify GCHFR in vitro

  • Expression System Selection:

    • Compare GCHFR produced in different expression systems:

      • E. coli (no eukaryotic PTMs)

      • Insect cells (basic eukaryotic PTMs)

      • Xenopus egg extracts (native PTM machinery)

      • Mammalian cells (complex PTM patterns)

    • Determine how expression system affects functional properties

Cellular Signaling Integration

  • Stimulation Studies:

    • Treat Xenopus embryos or cell cultures with signaling pathway activators/inhibitors

    • Monitor changes in GCHFR PTMs using mass spectrometry or Western blotting

    • Correlate PTM changes with alterations in BH4 levels

  • Kinase/Phosphatase Manipulation:

    • Use specific inhibitors to block candidate kinases/phosphatases

    • Express constitutively active or dominant-negative versions of regulatory enzymes

    • Identify signaling pathways that regulate GCHFR function

Visualizing PTMs in vivo

  • PTM-Specific Fluorescent Biosensors:

    • Develop FRET-based sensors that respond to GCHFR phosphorylation/dephosphorylation

    • Express these sensors in developing Xenopus embryos

    • Monitor PTM dynamics in real-time during development

  • Immunohistochemistry with PTM-Specific Antibodies:

    • Map the spatial distribution of modified GCHFR in tissues

    • Correlate with developmental events or physiological states

    • Compare patterns between wild-type and manipulated embryos

Evolutionary Conservation Analysis

  • Cross-Species Comparison:

    • Compare PTM sites between Xenopus and mammalian GCHFR

    • Identify conserved modification sites that likely serve critical functions

    • Determine if regulatory enzymes targeting these sites are also conserved

This multi-faceted approach will provide comprehensive insights into how post-translational modifications regulate GCHFR function in Xenopus laevis, potentially revealing developmental stage-specific or tissue-specific regulatory mechanisms that control BH4 biosynthesis throughout the amphibian life cycle.

How can researchers optimize recombinant GCHFR for structural studies?

Structural characterization of Xenopus laevis GCHFR provides critical insights into its function and regulatory mechanisms. The following methodological framework addresses the specialized requirements for producing and purifying GCHFR suitable for high-resolution structural studies:

Construct Design Optimization

  • Truncation Strategies:

    • Generate both full-length and N-terminally truncated versions (similar to mammalian studies that used GCH1 lacking the first 42 amino acids)

    • Create multiple constructs with varying N- and C-terminal boundaries based on predicted disorder and domain organization

    • Include flexible linkers when using fusion partners to prevent structural interference

  • Surface Engineering:

    • Identify and mutate surface-exposed residues with high conformational entropy (e.g., K/E→A)

    • Reduce surface charge patches that might impede crystallization

    • Consider the SER (Surface Entropy Reduction) approach to create crystal contacts

  • Fusion Partners for Crystallization:

    • Employ crystallization chaperones like T4 lysozyme or BRIL for challenging proteins

    • Use MBP or GST N-terminal fusions with precision protease sites for purification and optional removal

    • Consider nanobody co-crystallization to stabilize specific conformations

Expression System Selection

  • E. coli-Based Systems:

    • Test specialized strains designed for structural biology (SHuffle, Origami) that enhance disulfide bond formation

    • Optimize induction temperature (typically 16-18°C) and duration for proper folding

    • Co-express with chaperones if aggregation or inclusion body formation occurs

  • Eukaryotic Systems for Complex Structures:

    • Baculovirus-infected insect cells for higher-order assemblies

    • HEK293 GnTI- cells for structural studies requiring glycosylation but with limited heterogeneity

    • Consider cell-free expression systems for rapid screening of construct variants

Purification Strategy for Crystallography-Grade Protein

  • Multi-Step Purification Approach:

    • Affinity chromatography (typically IMAC for His-tagged proteins)

    • Ion exchange chromatography to remove charged contaminants

    • Size exclusion chromatography as a final polishing step

    • Consider on-column refolding for difficult-to-express constructs

  • Complex Formation:

    • Co-purify GCHFR with GCH1 to stabilize the complex

    • Add L-phenylalanine during purification to enhance complex stability (known to improve binding affinity eightfold)

    • Use GraFix (gradient fixation) method for stabilizing large assemblies

  • Quality Control Measures:

    • Dynamic light scattering to assess monodispersity

    • Thermal shift assays to optimize buffer conditions

    • Mass spectrometry to confirm exact mass and detect modifications

    • Negative stain EM to verify homogeneity of larger complexes

Crystallization Optimization

  • Initial Screening:

    • Employ sparse matrix commercial screens (typically 500-1000 conditions)

    • Test both vapor diffusion (sitting/hanging drop) and batch crystallization methods

    • Include both apo-GCHFR and the GCH1-GFRP complex with/without L-phenylalanine

  • Optimization Strategies:

    • Fine grid screens around promising conditions

    • Additive screens to improve crystal quality

    • Seeding techniques to control nucleation

    • Try in situ proteolysis to remove flexible regions during crystallization

  • Alternative Approaches:

    • Consider lipidic cubic phase (LCP) crystallization for membrane-associated forms

    • Use microfluidic approaches for efficient screening with minimal protein consumption

    • Try crystallization at different temperatures (4°C, 16°C, 20°C)

Complementary Structural Techniques

  • Cryo-Electron Microscopy:

    • Particularly suitable for the larger GCH1-GFRP complex (approximately 380 kDa)

    • Provides structure without crystallization

    • Enables visualization of different conformational states

  • Small-Angle X-ray Scattering (SAXS):

    • Provides low-resolution envelope of protein shape in solution

    • Useful for analyzing conformational changes upon complex formation or ligand binding

    • Complementary to crystallography and EM approaches

  • NMR Spectroscopy:

    • For studying dynamics of specific regions or domains

    • Mapping binding interfaces with chemical shift perturbation experiments

    • Investigation of smaller GCHFR fragments when complete structure determination is challenging

By implementing this comprehensive approach, researchers can optimize their chances of obtaining high-quality structural data for Xenopus laevis GCHFR, providing crucial insights into the molecular mechanisms underlying BH4 biosynthesis regulation across species.

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.