SPR Mouse

Sepiapterin Reductase Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Biochemical Properties and Structure

SPR Mouse is a non-glycosylated polypeptide chain containing 285 amino acids (1–262 residues) with a molecular mass of 30.3 kDa . It is fused to a 23-amino acid His-tag at the N-terminus for purification and immobilization purposes. The protein’s structure is stabilized through proprietary chromatographic techniques, ensuring high purity (>95% as determined by SDS-PAGE) .

PropertyValue/Description
Molecular Mass30.3 kDa
Amino Acids285 (1–262 residues)
Post-Translational ModificationsNon-glycosylated
Purification MethodProprietary chromatography
Purity>95% (SDS-PAGE)
Formulation20 mM Tris-HCl (pH 8.5), 1 mM DTT, 10% glycerol

Production and Stability

SPR Mouse is synthesized in E. coli using a pET plasmid system. The recombinant protein is purified via affinity chromatography, leveraging its His-tag for efficient isolation . Storage recommendations include freezing at -20°C with 0.1% HSA or BSA as a carrier protein to prevent aggregation during thawing cycles. Avoiding repeated freeze-thaw cycles is critical to maintaining structural integrity .

Potential Applications

While direct research on SPR Mouse is limited in the provided sources, its functional role in BH4 biosynthesis suggests applications in:

  • Therapeutic Development: BH4 deficiency is linked to cardiovascular and neurological disorders. SPR inhibitors or modulators may target BH4 pathways.

  • Enzyme Kinetics Studies: SPR’s catalytic activity (e.g., reducing sepiapterin to tetrahydrobiopterin) can be analyzed using techniques like surface plasmon resonance (SPR) or fluorescence polarization (FP) .

  • Structural Biology: The His-tag facilitates immobilization on sensor chips for real-time binding assays, enabling interaction studies with inhibitors or cofactors .

Research Gaps and Limitations

No peer-reviewed studies explicitly analyzing SPR Mouse were identified in the provided sources. Most references focus on SPR technology or unrelated mouse proteins (e.g., prion proteins in ). Further validation is needed to:

  • Characterize its catalytic efficiency (kcatk_{cat}, KMK_M).

  • Explore cross-species binding interactions (e.g., human vs. mouse SPR).

  • Investigate therapeutic relevance in disease models.

Product Specs

Introduction

Sepiapterin Reductase is a crucial enzyme involved in the biosynthesis of tetrahydrobiopterin (BH4), a vital cofactor for aromatic amino acid hydroxylases, including tyrosine hydroxylase, which plays a critical role in dopamine synthesis. This enzyme, belonging to the aldo-keto reductase family, catalyzes the NADPH-dependent reduction of pteridine derivatives. Mutations in the Sepiapterin Reductase gene can lead to DOPA-responsive dystonia due to sepiapterin reductase deficiency, characterized by sustained involuntary muscle contractions, often resulting in abnormal postures. Sepiapterin reductase is also involved in the reduction of exogenous carbonyl compounds and phenylpropanedione.

Description

Recombinant SPR Mouse, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 285 amino acids (with amino acids 1-262 being the SPR protein) and possessing a molecular mass of 30.3 kDa. This protein is engineered with a 23 amino acid His-tag at its N-terminus and is purified using proprietary chromatographic techniques.

Physical Appearance
A clear, sterile-filtered solution.
Formulation

The SPR protein solution is provided at a concentration of 1 mg/ml and is formulated in a buffer containing 20 mM Tris-HCl (pH 8.5), 1 mM DTT, and 10% glycerol.

Stability

For short-term storage (2-4 weeks), the protein should be stored at 4°C. For extended storage, it is recommended to store the protein at -20°C. To further enhance long-term stability, the addition of a carrier protein (0.1% HSA or BSA) is advisable. It is crucial to avoid repeated freeze-thaw cycles to maintain protein integrity.

Purity

The purity of the SPR protein is determined to be greater than 95% using SDS-PAGE analysis.

Synonyms

SDR38C1, SPR, Dystonia, Sepiapterin reductase, mCG_128676.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSMEAGGLG CAVCVLTGAS RGFGRALAPQ LARLLSPGSV MLVSARSESM LRQLKEELGA QQPDLKVVLA AADLGTEAGV QRLLSAVREL PRPEGLQRLL LINNAATLGD VSKGFLNVND LAEVNNYWAL NLTSMLCLTS GTLNAFQDSP GLSKTVVNIS SLCALQPYKG WGLYCAGKAA RDMLYQVLAA EEPSVRVLSY APGPLDNDMQ QLARETSKDP ELRSKLQKLK SDGALVDCGT SAQKLLGLLQ KDTFQSGAHV DFYDC.

Q&A

What is Sepiapterin Reductase (SPR) and why is it important in mouse models?

Sepiapterin Reductase is an aldo-keto reductase that catalyzes the NADPH-dependent reduction of pteridine derivatives and plays an essential role in the biosynthesis of tetrahydrobiopterin (BH4) . BH4 functions as a necessary cofactor for aromatic amino acid hydrolases, including tyrosine hydroxylase, which is the rate-limiting enzyme in dopamine synthesis . The importance of SPR in mouse models stems from its critical role in neurotransmitter synthesis and metabolism. SPR-deficient mice display significantly disturbed pterin profiles and greatly diminished levels of dopamine, norepinephrine, and serotonin, indicating that SPR is essential for homeostasis of BH4 and for the normal functions of BH4-dependent enzymes . These mouse models allow researchers to study the biochemical pathways involved in neurotransmitter synthesis and the pathophysiology of conditions resulting from disruptions in these pathways.

What are the basic applications of Surface Plasmon Resonance with mouse antibodies?

Surface Plasmon Resonance (SPR) with mouse antibodies is primarily used to characterize antibody-antigen interactions in real-time without requiring sample labeling . The basic applications include:

  • Determination of binding kinetics (association and dissociation rates) between mouse antibodies and their target antigens

  • Measurement of binding affinities (KD values) for mouse antibody-antigen complexes

  • Epitope mapping to identify specific binding regions on target antigens

  • Screening of hybridoma supernatants for antibody production

  • Cross-reactivity testing of mouse monoclonal antibodies

The technique works by immobilizing either the antibody or antigen on a sensor chip and flowing the binding partner over the surface. Changes in the refractive index caused by binding events are detected as resonance units (RU), which are proportional to the mass on the surface . This allows researchers to observe binding events in real-time and calculate precise kinetic parameters that help characterize molecular interactions involving mouse-derived antibodies or proteins.

How should researchers select the appropriate SPR sensor chip for studying mouse antibody interactions?

  • Immobilization strategy: For mouse antibodies, consider:

    • Direct amine coupling when antibody orientation is not critical

    • Capture-based approaches (using protein A/G or anti-mouse antibodies) when preserving antibody orientation is essential for antigen binding

  • Target size: For smaller analytes interacting with mouse antibodies, chips with higher surface densities may be necessary to generate sufficient signal

  • Sample complexity: When working with complex mouse tissue extracts or serum samples, chips with lower non-specific binding properties (like CM4 or C1) may be preferable

  • Regeneration requirements: Consider chip stability when multiple regeneration cycles are needed for screening experiments

The choice between standard amine coupling and capture-mediated immobilization strategies is particularly important, as the latter often generates more homogeneous surfaces with better-oriented antibodies, which can significantly improve antigen detection sensitivity . For mouse monoclonal antibodies like 1H6, specialized mouse antibody capture kits can provide optimal orientation and binding capacity while preserving antibody functionality .

What are the optimal protein expression and purification methods for mouse SPR recombinant proteins?

The optimal expression and purification methods for mouse SPR recombinant proteins typically involve bacterial expression systems, particularly Escherichia coli, followed by affinity chromatography. Based on established protocols:

  • Expression system: E. coli represents the preferred expression host for mouse SPR recombinant proteins, as demonstrated by the successful production of high-purity preparations . The bacterial system allows for cost-effective scaling and typically yields non-glycosylated polypeptide chains with preserved enzymatic activity.

  • Affinity tags: Incorporation of histidine tags (His-tags) facilitates purification via metal affinity chromatography. The standard construct includes an N-terminal His-tag followed by a thrombin cleavage site (e.g., MGSSHHHHHH SSGLVPRGSH) .

  • Buffer optimization: For maximum stability, mouse SPR proteins should be formulated in Tris-HCl buffer (20mM, pH 8.5) containing reducing agents (1mM DTT) and stabilizers (10% glycerol) .

  • Purification verification: SDS-PAGE analysis should confirm purity greater than 95%, with expected molecular weight corresponding to the 262 amino acid sequence plus any tags .

  • Storage conditions: For short-term use (2-4 weeks), store at 4°C; for long-term storage, maintain at -20°C with added carrier proteins (0.1% HSA or BSA) to prevent degradation during freeze-thaw cycles .

These protocols yield functional mouse SPR proteins suitable for enzymatic assays, structural studies, and as standards in biochemical analyses of BH4 metabolism and related pathways.

What controls are essential when designing experiments with SPR-knockout mice?

When designing experiments with SPR-knockout mice, several essential controls must be incorporated to ensure valid and interpretable results:

  • Genotyping controls: Accurate PCR-based genotyping is critical, using validated primer sets such as:

    • For mutant allele detection: SprF1 (5′-AAGTGGTGCTGGCAGCCGCCGAT-3′) and NeoP3 (5′-CGGTGCTGTCCATCTGCACGAGAC-3′)

    • For wild-type allele detection: srex2F (5′-CCTCCATGCTCTGTTTGACT-3′) and srex2R (5′-GTTCCCCTCCTTGCCTAGC-3′)

  • Genetic background controls: Include littermate wild-type (+/+) and heterozygous (+/-) mice to control for background strain effects, particularly when using hybrid backgrounds (e.g., 129/B6) .

  • Biochemical validation controls:

    • Tissue-specific measurements of BH4 levels in brain and liver

    • Neurotransmitter quantification in relevant brain regions (dopamine, serotonin, norepinephrine)

    • Serum phenylalanine measurements to confirm the phenylketonuria phenotype

  • Behavioral controls: Include standardized assessments of locomotor activity with appropriate age and sex-matched controls, as SPR knockout affects movement capabilities .

  • Treatment controls: When testing therapeutic interventions (such as BH4 supplementation), include vehicle-treated groups and dose-response cohorts to establish efficacy thresholds .

  • Environmental controls: Maintain consistent housing conditions as environmental factors may influence phenotype severity, particularly for neurobehavioral outcomes.

Proper experimental design should account for sex differences, as they may influence the severity of the phenotype, and should include sufficient sample sizes to accommodate the generally higher variability observed in knockout mouse studies compared to wild-type controls.

How can Surface Plasmon Resonance be optimized for detecting small-molecule interactions with mouse SPR protein?

Optimizing Surface Plasmon Resonance for small-molecule interactions with mouse SPR protein requires several specialized approaches:

  • High-density immobilization: For small-molecule detection, immobilize high densities of mouse SPR protein (>10,000 RU) on the sensor chip to generate sufficient signal-to-noise ratio, as small molecules produce lower response values due to their lower mass .

  • Specialized sensor chips: Consider using sensor chips designed for small molecule detection, such as CM7 (higher density carboxymethylated dextran) or planar surfaces that minimize mass transport limitations .

  • Reversed assay format: When possible, immobilize the small molecule (or a derivative) and flow the mouse SPR protein as the analyte to amplify the response signal due to the larger mass of the protein .

  • Solvent correction: Implement proper solvent correction procedures when working with compounds requiring organic solvents like DMSO, which can cause bulk refractive index changes .

  • Temperature control: Maintain tight temperature control (±0.1°C) throughout experiments, as small molecule binding is often enthalpy-driven and highly temperature-sensitive .

  • Reference channel design: Design reference surfaces that closely mimic the active surface minus the specific binding capability to account for non-specific binding and bulk effects .

  • Single-cycle kinetics: Employ single-cycle kinetics approaches to minimize consumption of valuable compounds and reduce regeneration steps that might damage the immobilized mouse SPR protein .

Using these specialized approaches, researchers can achieve detection of compounds with molecular weights as low as 100-200 Da binding to mouse SPR protein, with affinity determinations possible in the nanomolar to millimolar range.

What are the current challenges in correlating SPR mouse knockout phenotypes with human SPR deficiency disorders?

The correlation between SPR mouse knockout phenotypes and human SPR deficiency disorders faces several significant challenges:

  • Phenotypic divergence: SPR-deficient mice display phenylketonuria (elevated phenylalanine levels) which is not typically observed in human patients, suggesting species-specific differences in compensatory metabolic pathways or enzyme redundancy . This metabolic difference complicates direct translation of therapeutic approaches.

  • Severity gradient: The mouse model presents with more severe phenotypes, including pronounced dwarfism and dramatically reduced brain BH4 levels, whereas human presentations are more variable and often milder . This disparity makes dosing predictions for therapeutic interventions challenging.

  • Developmental timing: Neurodevelopmental consequences of SPR deficiency appear to follow different trajectories in mice versus humans, potentially due to differences in brain development timelines and compensatory mechanisms during critical periods.

  • Limited behavioral correlates: While impaired locomotion is observed in both species, the complex cognitive and psychiatric manifestations seen in human patients are difficult to model and assess in mice.

  • Treatment response variations: Though both mice and humans respond to BH4 supplementation, the dosing requirements, pharmacokinetics, and long-term treatment outcomes differ considerably between species .

  • Genetic background effects: The mouse phenotype can be influenced by strain background effects (often using 129/B6 hybrid backgrounds), which adds complexity when translating to the diverse genetic backgrounds in human patients .

Addressing these challenges requires integrated approaches combining detailed biochemical profiling, cross-species pharmacokinetic modeling, and the potential development of humanized mouse models that better recapitulate the pathophysiology of human SPR deficiency.

How can researchers integrate SPR binding data with mouse genomic information for drug discovery?

Integrating SPR binding data with mouse genomic information can significantly enhance drug discovery processes through several methodological approaches:

  • Pharmacogenomic correlation: Cross-reference SPR-derived binding affinity data (KD, kon, koff) with strain-specific SNP variations from the Mouse Phenome Database to identify genetic determinants of binding variation . This approach can reveal:

    • Target protein variants affecting drug binding profiles

    • Genetic modifiers influencing drug response pathways

    • Potential off-target interactions

  • Target validation via knockouts: Combine SPR interaction data with phenotypic data from corresponding gene knockout models (available through MPD) to establish target engagement requirements . This correlation helps:

    • Validate on-target effects versus potential off-target activities

    • Establish minimum binding requirements for therapeutic effect

    • Identify compensatory mechanisms affecting drug efficacy

  • Multiplexed SPR screening with genomic stratification: Design SPR screens using proteins derived from different mouse strains with known genomic differences to:

    • Identify compounds with robust binding across genetic variations

    • Discover strain-specific interactions that may predict personalized medicine applications

    • Establish structure-activity relationships informed by genetic variation

  • Data integration framework:

Data TypeSourceIntegration MethodOutcome Measure
SPR Binding KineticsBiacore T200 measurementsStatistical correlationBinding parameters (KD, kon, koff)
SNP ProfilesMouse Phenome DatabaseGenomic mappingTarget protein variation
Strain PhenotypesMouse Phenome DatabaseMultivariate analysisPhenotypic correlates of binding
Gene ExpressionTissue-specific transcriptomicsNetwork analysisContext-dependent binding effects

This integrated approach leveraging the Mouse Phenome Database's extensive SNP data (8+ million unique genomic locations across multiple strains) with quantitative SPR binding measurements enables researchers to develop pharmaceuticals with optimized target engagement profiles across diverse genetic backgrounds .

What are common artifacts in SPR measurements with mouse antibodies and how can they be addressed?

Common artifacts in SPR measurements with mouse antibodies and their remediation strategies include:

  • Mass transport limitation:

    • Issue: When mouse antibody-antigen binding is faster than analyte delivery to the surface, creating artificially slow association rates

    • Solution: Reduce immobilization levels, increase flow rates (>30 μL/min), and use lower ligand densities when working with mouse antibodies

  • Baseline drift:

    • Issue: Progressive signal decrease or increase during the experiment

    • Solution: Ensure thorough system equilibration with running buffer (>1 hour), maintain consistent temperature (±0.1°C), and degas all buffers to prevent air bubble formation

  • Heterogeneous binding profiles:

    • Issue: Non-uniform mouse antibody orientation creating multiple binding populations with different kinetics

    • Solution: Implement capture-based immobilization using anti-mouse antibody capture kits rather than direct amine coupling to ensure uniform orientation

  • Regeneration damage:

    • Issue: Progressive loss of activity after multiple regeneration cycles

    • Solution: Optimize regeneration conditions (test mild options first: 10mM glycine pH 2.5, then progress to more stringent conditions if needed), and implement single-cycle kinetics approaches for sensitive mouse antibodies

  • Non-specific binding:

    • Issue: Background signal from sample components binding to the reference or active surface

    • Solution: Add surfactants (0.005-0.05% P20), increase NaCl concentration (150-300mM), and include bovine serum albumin (0.1-1mg/mL) in running buffer; implement reference surface correction with appropriate negative controls

  • Analyte precipitation/aggregation:

    • Issue: Spikes or irregular sensorgrams due to protein aggregates

    • Solution: Centrifuge samples (10,000×g for 10 minutes), filter through 0.22μm filters immediately before injection, and maintain mouse protein samples at appropriate temperatures to prevent denaturation

By systematically addressing these common artifacts, researchers can significantly improve the quality and reliability of SPR data when working with mouse antibodies and their targets.

How can researchers validate the phenotypic characteristics of SPR-knockout mice?

Validating the phenotypic characteristics of SPR-knockout mice requires a comprehensive multi-level approach:

  • Genetic validation:

    • Confirm genotype using multiple PCR primer sets targeting both wild-type and mutant alleles

    • Verify absence of SPR mRNA expression through RT-PCR or RNA sequencing

    • Confirm deletion of targeted genomic region through sequencing

  • Biochemical validation:

    • Enzyme activity assays: Measure SPR enzymatic activity in tissue homogenates (liver, brain) to confirm complete functional knockout

    • Metabolite profiling: Quantify BH4 levels using HPLC with electrochemical detection or LC-MS/MS in:

      • Brain tissues (striatum, cortex, cerebellum)

      • Liver

      • Blood

    • Neurotransmitter quantification: Measure dopamine, serotonin, and their metabolites in relevant brain regions

  • Phenotypic characterization:

    • Growth parameters: Track body weight, length, and growth curves from birth through adulthood

    • Metabolic parameters: Monitor serum phenylalanine levels, amino acid profiles, and glucose metabolism

    • Behavioral assessment:

      • Locomotor activity in open field tests

      • Motor coordination using rotarod

      • Cognitive function using maze tests

  • Response to intervention:

    • BH4 supplementation: Verify reversal of biochemical abnormalities

    • Neurotransmitter precursor therapy: Document improvements in motor function

    • Dose-response relationships: Establish therapeutic thresholds for interventions

  • Cross-validation with published phenotypes:

    • Compare observed phenotypes with published data on SPR-knockout models

    • Establish concordance with human SPR deficiency cases where possible

This multi-level validation approach ensures that observed phenotypes are directly attributable to SPR deficiency rather than potential confounding factors such as genetic background effects or compensatory mechanisms during development.

What quality control measures should be implemented when preparing mouse samples for SPR analysis?

Implementing stringent quality control measures when preparing mouse samples for SPR analysis is essential for generating reliable and reproducible results:

  • Sample purity assessment:

    • Proteins/antibodies: Verify purity >95% via SDS-PAGE and/or size exclusion chromatography

    • Measure aggregation: Use dynamic light scattering or analytical ultracentrifugation to detect aggregates that may cause artifacts

    • Activity testing: Confirm biological activity/binding capacity prior to SPR analysis

  • Buffer optimization protocols:

    • pH and ionic strength verification: Measure and adjust to match running buffer (±0.05 pH units, ±5mM salt)

    • Surfactant consistency: Ensure identical surfactant concentrations in sample and running buffers

    • Degassing: Remove dissolved air to prevent bubble formation during injections

  • Pre-injection processing:

    • Centrifugation: 10,000-15,000×g for 10 minutes immediately before analysis

    • Filtration: Pass through 0.22μm filters to remove particulates

    • Temperature equilibration: Allow samples to reach system temperature (20-25°C typically)

  • Reference sample preparation:

    • Concentration determination: Measure protein concentration using multiple methods (UV280, BCA, Bradford)

    • Standard curve validation: Prepare fresh concentration standards with verified reference material

    • Aliquoting: Prepare single-use aliquots to avoid freeze-thaw cycles

  • Storage and stability monitoring:

    • Short-term stability: Verify signal stability at experimental temperature over expected experiment duration

    • Long-term storage: Add stabilizers (10% glycerol, 0.1% BSA) for proteins stored below -20°C

    • Freeze-thaw testing: Validate sample performance after multiple freeze-thaw cycles if reuse is planned

  • Documentation requirements:

    • Maintain detailed records of sample source, preparation date, buffer composition

    • Document all processing steps and quality control results

    • Include batch/lot numbers of antibodies or recombinant proteins

Implementation of these quality control measures significantly reduces experimental variability and ensures that observed binding phenomena represent true molecular interactions rather than artifacts of sample preparation.

How should researchers interpret kinetic binding data from SPR experiments with mouse proteins?

Interpreting kinetic binding data from SPR experiments with mouse proteins requires systematic analysis and consideration of several factors:

  • Kinetic parameter extraction:

    • Association rate constant (ka): Typically ranges from 10³ to 10⁷ M⁻¹s⁻¹ for protein-protein interactions

    • Dissociation rate constant (kd): Typical range 10⁻¹ to 10⁻⁶ s⁻¹

    • Equilibrium dissociation constant (KD = kd/ka): Ranges from nanomolar to micromolar for most biological interactions

  • Fitting model selection:

    • 1:1 Langmuir binding: Simplest model, appropriate when residual plots show random distribution around zero

    • Heterogeneous ligand: When mouse antibody orientation creates two distinct binding populations

    • Two-state conformational change: When binding induces conformational changes in mouse proteins

    • Mass transport limited: When diffusion to the surface limits observed binding rates

  • Data quality assessment metrics:

    • Chi-square values: Should be <10% of Rmax

    • Residual distribution: Should be random and <5% of maximum response

    • U-value: Statistical parameter indicating uniqueness of the fit, should be <25

  • Concentration series validation:

    • Dose-dependency: Verify response proportionality to concentration

    • Replicate consistency: CV <20% between technical replicates

    • Range appropriateness: Test concentrations spanning 0.1-10× KD when possible

  • Reference-relative interpretation:

    • Compare with published values for similar interactions

    • Establish relative affinity rankings when screening multiple compounds

    • Consider structure-activity relationships when interpreting binding differences

  • Biological context integration:

    • Correlate binding parameters with functional activity in cell-based assays

    • Compare SPR-derived KD values with IC50/EC50 values from functional assays

    • Consider how kinetic parameters (particularly residence time, 1/kd) may influence in vivo activity

This structured approach to data interpretation enables researchers to extract maximum biological significance from SPR kinetic data while avoiding common pitfalls in analysis.

What bioinformatic approaches can integrate SPR mouse model data with human disease databases?

Integrating SPR mouse model data with human disease databases requires sophisticated bioinformatic approaches that bridge species differences while maintaining biological relevance:

  • Ortholog mapping frameworks:

    • Utilize resources like HomoloGene, OrthoMCL, or ENSEMBL Compara to establish high-confidence mouse-human gene relationships

    • Implement sequence-based similarity metrics with functional domain conservation analysis

    • Validate ortholog predictions through phylogenetic tree construction

  • Phenotype ontology alignment:

    • Map mouse phenotypes to Human Phenotype Ontology (HPO) terms

    • Utilize Mammalian Phenotype Ontology to standardize mouse phenotypic descriptions

    • Calculate semantic similarity scores between mouse model phenotypes and human disease manifestations

  • Pathway-centric integration methods:

    • Map biochemical alterations in SPR mouse models to human pathway databases (KEGG, Reactome)

    • Identify conserved pathway perturbations across species

    • Analyze network topology to identify species-specific differences in pathway regulation

  • Multi-omics data integration:

    • Correlate transcriptomic signatures between mouse models and human patient samples

    • Map metabolomic profiles across species with emphasis on BH4 and neurotransmitter pathways

    • Perform cross-species proteome analysis focusing on post-translational modifications

  • Machine learning approaches:

    • Train algorithms on parallel mouse-human datasets to identify predictive features

    • Implement disease-specific classifiers using mouse model data as training sets

    • Develop translational scoring systems to prioritize mouse phenotypes by human relevance

  • Integration architecture: An effective bioinformatic pipeline might include:

Data TypeMouse SourceHuman SourceIntegration Method
GenomicMPD SNP dataClinVar/OMIMSynteny mapping
TranscriptomicSPR−/− tissuesPatient biopsiesCo-expression networks
MetabolomicBH4/neurotransmitter profilesCSF biomarkersMetabolic pathway mapping
PhenotypicStandardized phenotypingHPO annotationsSemantic similarity

These approaches enable researchers to maximize the translational value of SPR mouse models while acknowledging species-specific differences that might affect therapeutic development strategies .

How can researchers distinguish between specific and non-specific binding in SPR experiments with mouse tissue extracts?

Distinguishing between specific and non-specific binding in SPR experiments with mouse tissue extracts presents unique challenges due to sample complexity. Researchers should implement a comprehensive strategy:

  • Reference surface design strategies:

    • Negative control ligands: Immobilize irrelevant proteins of similar size/properties

    • Denatured ligands: Use heat-inactivated or chemically denatured versions of the target protein

    • Density matching: Ensure equivalent levels of immobilization on reference and active surfaces

  • Competitive binding validation:

    • Pre-incubate tissue extracts with soluble target to block specific binding sites

    • Perform concentration-dependent competition assays to verify binding specificity

    • Calculate IC50 values that should correlate with direct binding KD values

  • Multiple capture formats:

    • Compare different immobilization chemistries (amine, thiol, biotin)

    • Test both direct coupling and oriented capture approaches

    • Verify binding with inverted assay formats (if possible)

  • Advanced data processing approaches:

    • Double-referencing: Subtract both reference surface and buffer-only injections

    • Modified fitting models: Incorporate non-specific binding parameters in kinetic models

    • Multivariate analysis: Apply principal component analysis to distinguish binding patterns

  • Sample pre-treatment optimization:

    • Implement size exclusion or affinity pre-purification steps

    • Add blocking agents (BSA, dextran, non-ionic detergents) to reduce non-specific interactions

    • Modify ionic strength and pH to minimize non-specific electrostatic interactions

  • Controls for tissue extract validation:

    • Test extracts from wild-type vs. knockout mice to confirm specificity

    • Use fractionated tissue extracts to identify interfering components

    • Include positive control analytes with known binding profiles

  • Quantitative assessment metrics:

Assessment ApproachSpecific Binding IndicatorNon-Specific Binding Indicator
Concentration dependenceSaturable responseLinear, non-saturable response
Competition assays>80% signal reduction<20% signal reduction
Kinetic profilesDefined association/dissociationRapid association with minimal dissociation
Buffer sensitivityMinimal impact of mild detergentsSignificant reduction with detergents

By implementing this multifaceted approach, researchers can confidently differentiate between specific interactions of interest and non-specific binding artifacts when working with complex mouse tissue extracts in SPR experiments .

What emerging technologies are enhancing SPR mouse model applications in personalized medicine?

Several emerging technologies are significantly enhancing the application of SPR mouse models in personalized medicine research:

  • Microfluidic SPR array platforms:

    • Enable simultaneous testing of multiple mouse strain-derived proteins

    • Allow for high-throughput screening of genetic variants affecting drug binding

    • Facilitate personalized drug response prediction based on genetic background differences

  • CRISPR-engineered precision mouse models:

    • Generate mice with human-specific mutations in SPR gene

    • Create isogenic lines differing only in specific disease-associated variants

    • Enable direct comparison of variant-specific drug responses in genetically defined backgrounds

  • Single-cell SPR technologies:

    • Analyze binding interactions at the individual cell level

    • Map tissue-specific variation in drug target expression and binding

    • Correlate binding affinity with cellular phenotypes in heterogeneous tissues

  • Integrated multi-omics approaches:

    • Combine SPR binding data with transcriptomics, proteomics, and metabolomics

    • Map drug binding parameters to downstream pathway modulation

    • Identify biomarkers predictive of therapeutic response based on molecular signatures

  • Organ-on-chip SPR integration:

    • Incorporate SPR sensors into microfluidic organ models

    • Measure real-time binding in physiologically relevant microenvironments

    • Assess how tissue-specific factors modulate drug-target interactions

  • AI-augmented SPR data analysis:

    • Implement machine learning algorithms to predict in vivo efficacy from SPR parameters

    • Develop models translating mouse strain differences to human population variation

    • Identify complex multivariate patterns correlating binding profiles with treatment outcomes

  • Humanized SPR mouse models:

    • Replace mouse SPR gene with human variants to better mimic human disease

    • Express human target proteins in mouse tissues for more translational drug binding studies

    • Model population-specific genetic variations affecting SPR function and drug responses

These technologies collectively enable a systems pharmacology approach to personalized medicine, where genetic variation, molecular binding parameters, and physiological responses can be integrated to predict individual treatment outcomes with unprecedented precision.

How might longitudinal studies with SPR-deficient mice inform therapeutic development for human disorders?

Longitudinal studies with SPR-deficient mice can significantly inform therapeutic development for human disorders through several methodological approaches:

  • Developmental trajectory mapping:

    • Track biochemical, physiological, and behavioral changes from birth through adulthood

    • Identify critical developmental windows for therapeutic intervention

    • Correlate neurodevelopmental milestones between mouse models and human patients

  • Biomarker evolution profiling:

    • Monitor dynamic changes in pterin metabolites, neurotransmitters, and related biomarkers

    • Establish predictive biomarkers for disease progression

    • Identify early diagnostic markers that precede symptom onset

  • Treatment timing optimization:

    • Implement intervention at different developmental stages to determine optimal therapeutic windows

    • Compare early versus late intervention outcomes

    • Assess the reversibility of specific phenotypes at different disease stages

  • Combination therapy assessment:

    • Test sequential and simultaneous administration of BH4 supplementation with neurotransmitter precursors

    • Evaluate synergistic effects of multiple therapeutic approaches

    • Determine optimal dosing regimens for combination treatments

  • Compensatory mechanism identification:

    • Characterize adaptive responses that emerge over time in SPR-deficient mice

    • Identify molecular pathways that could be therapeutically augmented

    • Map the temporal dynamics of compensatory gene expression

  • Long-term safety evaluation:

    • Monitor chronic treatment effects on growth, development, and organ systems

    • Assess potential toxicities associated with sustained therapeutic interventions

    • Identify biomarkers of treatment-related adverse effects

  • Treatment discontinuation studies:

    • Evaluate persistence of therapeutic benefits after treatment cessation

    • Identify rebound phenomena requiring maintenance therapy

    • Determine minimum effective treatment durations

These longitudinal approaches would generate comprehensive datasets relating therapeutic interventions to disease stage, enabling the development of more precisely targeted treatments for human SPR deficiency and related disorders. The temporal dimension of such studies provides crucial insights into disease evolution that cannot be obtained from cross-sectional analyses alone.

What are the potential applications of combining SPR technology with mouse genomic editing for target validation?

Combining SPR technology with mouse genomic editing creates powerful approaches for target validation with numerous applications:

  • Precision binding site verification:

    • Use CRISPR to generate mice with specific mutations in predicted binding sites

    • Compare SPR binding profiles between wild-type and mutant proteins

    • Correlate binding affinity changes with physiological phenotypes

    • Establish structure-function relationships with unprecedented precision

  • Mechanism-based safety assessment:

    • Create mouse models with humanized binding domains for specific drug targets

    • Use SPR to assess on-target binding to the humanized protein

    • Map off-target binding profiles across related proteins

    • Identify species-specific differences in binding that may affect safety or efficacy translation

  • Resistance mechanism characterization:

    • Engineer mouse models with clinically-observed resistance mutations

    • Measure altered binding kinetics using SPR

    • Correlate binding changes with functional outcomes

    • Develop next-generation compounds to overcome specific resistance mechanisms

  • Allosteric modulator development:

    • Create mice with mutations in predicted allosteric sites

    • Use SPR to detect conformational changes in protein-protein interactions

    • Identify compounds that modify protein function through allosteric mechanisms

    • Validate allosteric binding sites through correlation of in vitro and in vivo data

  • Isoform-specific targeting:

    • Generate mice expressing only specific protein isoforms

    • Use SPR to screen compounds for isoform selectivity

    • Correlate isoform-specific binding with tissue-specific effects

    • Develop therapeutics with improved tissue selectivity profiles

  • Biomarker qualification:

    • Engineer mice with reporter-tagged variants of drug targets

    • Use SPR to calibrate biomarker responses to target engagement

    • Establish quantitative relationships between binding occupancy and biomarker modulation

    • Validate pharmacodynamic markers for clinical development

This integration of technologies enables bidirectional validation, where genomic editing confirms mechanisms identified through SPR studies, while SPR provides quantitative binding data that explains phenotypes observed in genetically modified mice. The approach represents a significant advancement over traditional methods by establishing clear causal relationships between molecular interactions and physiological outcomes.

What are the key considerations for translating SPR mouse model findings to human applications?

Translating SPR mouse model findings to human applications requires careful consideration of several critical factors to ensure relevance and accuracy:

  • Species-specific metabolic differences:

    • Account for the notable differences in phenylalanine metabolism between SPR-deficient mice and human patients

    • Adjust dosing protocols based on species-specific pharmacokinetics and metabolic rates

    • Consider alternate routes of administration that may be more translatable between species

  • Genetic background effects:

    • Recognize that mouse strain backgrounds influence phenotype severity and treatment responses

    • Utilize data from multiple mouse strains to predict response variability in diverse human populations

    • Consider the impact of genetic modifiers present in human patients but absent in controlled mouse models

  • Developmental timing disparities:

    • Account for differences in neurodevelopmental timelines between mice and humans

    • Adjust the timing of interventions based on relative developmental stages rather than absolute age

    • Consider the impact of treatment at equivalent developmental windows rather than chronological age

  • Physiological parameter scaling:

    • Implement appropriate allometric scaling when translating dosing from mice to humans

    • Consider species differences in blood-brain barrier permeability affecting central nervous system drug delivery

    • Account for differences in receptor expression, distribution, and density between species

  • Endpoint translation:

    • Develop translational biomarkers that reflect disease mechanisms across species

    • Focus on conserved biochemical pathways rather than species-specific phenotypes

    • Utilize homologous physiological parameters that can be measured in both mice and humans

  • Refinement of preclinical modeling:

    • Consider using humanized mice expressing human SPR variants to better predict clinical responses

    • Implement translational pharmacokinetic/pharmacodynamic modeling to bridge mouse and human data

    • Develop in vitro systems using human cells to complement mouse in vivo findings

By addressing these considerations systematically, researchers can maximize the predictive value of SPR mouse models while acknowledging their limitations, ultimately improving the success rate of translating preclinical findings to effective human therapies.

How can researchers build collaborative networks to advance SPR mouse research?

Researchers can build effective collaborative networks to advance SPR mouse research through strategic approaches that leverage complementary expertise and resources:

  • Cross-disciplinary partnership structures:

    • Connect basic scientists studying SPR biochemistry with clinical researchers treating SPR deficiency

    • Integrate SPR mouse genetics experts with surface plasmon resonance technology specialists

    • Bridge neuroscience, metabolism, and developmental biology through focused research consortia

  • Resource sharing platforms:

    • Establish centralized repositories for SPR mouse strains with standardized phenotyping

    • Develop shared databases integrating molecular, cellular, and behavioral phenotypes

    • Implement material transfer agreements that facilitate broad access to specialized models

  • Standardized methodological frameworks:

    • Develop consensus protocols for SPR mouse characterization and experimental procedures

    • Create standardized reporting formats for SPR binding studies to enable meta-analysis

    • Establish common data elements for phenotypic characterization

  • Translational research infrastructure:

    • Form partnerships between academic institutions and clinical centers treating SPR deficiency

    • Create patient registries linked to preclinical research databases

    • Develop biobanks of patient samples paired with corresponding mouse model tissues

  • Technology integration initiatives:

    • Establish core facilities specializing in both SPR technology and mouse model phenotyping

    • Develop integrated data analysis pipelines combining SPR binding data with in vivo outcomes

    • Create training programs focusing on translational approaches in SPR research

  • Funding mechanism utilization:

    • Target consortium grants specifically designed for multi-investigator collaborations

    • Leverage industry partnerships for technology development and compound screening

    • Pursue patient advocacy organization funding to accelerate therapeutic development

  • Knowledge dissemination strategies:

    • Organize focused workshops bringing together diverse expertise in SPR research

    • Create open-access resources summarizing current knowledge and methodological approaches

    • Develop web-based platforms for real-time data sharing and collaborative analysis

By implementing these approaches, researchers can create synergistic networks that accelerate progress in understanding SPR biology, developing improved mouse models, and translating findings to human applications more efficiently than would be possible through isolated research efforts.

What ethical considerations should guide the development and use of SPR mouse models?

Ethical considerations guiding the development and use of SPR mouse models should address several key dimensions:

  • Refinement of experimental design:

    • Implement power analyses to determine minimum necessary sample sizes

    • Develop non-invasive monitoring techniques to reduce stress and discomfort

    • Refine behavioral testing protocols to minimize animal distress while maintaining scientific validity

  • Reduction strategies:

    • Utilize advanced statistical approaches to maximize information from each animal

    • Implement longitudinal study designs when appropriate to reduce total animal numbers

    • Share tissues and data across research groups to prevent duplicate experiments

  • Replacement considerations:

    • Develop in vitro models using mouse or human cells where appropriate

    • Utilize computational modeling to predict outcomes and reduce animal testing

    • Implement organoid technologies as complementary approaches

  • Severity classification and mitigation:

    • Accurately classify the severity of SPR mouse phenotypes (e.g., dwarfism, impaired locomotion)

    • Establish humane endpoints specific to the manifestations of SPR deficiency

    • Implement supportive care protocols to minimize suffering

  • Genetic modification considerations:

    • Evaluate whether less invasive approaches could answer the research question

    • Consider potential off-target effects of genetic engineering techniques

    • Monitor for unintended consequences of genetic modifications

  • Translational value assessment:

    • Regularly reassess the predictive value of mouse models for human conditions

    • Consider whether phenotypic differences from human disease limit translational potential

    • Balance scientific benefit against animal welfare concerns

  • Transparency and reporting standards:

    • Implement ARRIVE guidelines for comprehensive reporting of animal experiments

    • Document all procedures, including unsuccessful approaches, to prevent repetition

    • Make detailed methodologies publicly available to support reproducibility

  • End-user engagement:

    • Involve patient advocacy groups in research prioritization

    • Consider the perspectives of individuals with SPR deficiency when designing studies

    • Ensure research goals align with meaningful patient outcomes

Product Science Overview

Introduction

Sepiapterin reductase (SPR) is an enzyme that plays a crucial role in the biosynthesis of tetrahydrobiopterin (BH4), an essential cofactor for various enzymatic reactions. This enzyme is part of the short-chain dehydrogenase/reductase (SDR) family and is involved in the reduction of sepiapterin to dihydrobiopterin, which is subsequently converted to BH4 .

Structure and Function

SPR is a homodimer composed of two subunits. It catalyzes the NADPH-dependent reduction of various carbonyl substances, including derivatives of pteridines . The enzyme’s active site contains key amino acid residues such as Ser-158, Tyr-171, and Lys-175, which are crucial for proton transfer and stabilization of the carbonyl group of substrates .

Biological Significance

BH4 is a key cofactor for several enzymes, including nitric oxide synthases (NOSs), aromatic amino acid hydroxylases, and alkylglycerol monooxygenase. These enzymes are involved in various biological processes such as monoamine neurotransmitter formation, immune response, cardiovascular function, and endothelial dysfunction . Therefore, SPR is indirectly associated with these critical physiological functions.

Clinical Relevance

Mutations or deficiencies in SPR can lead to various diseases, including brain dysfunction, chronic pain, cardiovascular disease, and cancer . The enzyme’s wide distribution in different tissues further underscores its importance in maintaining normal physiological functions.

Recombinant SPR

Recombinant mouse SPR is produced using genetic engineering techniques to express the mouse SPR gene in a suitable host system. This recombinant protein is used in research to study the enzyme’s structure, function, and role in disease. It also serves as a tool for screening potential therapeutic compounds targeting SPR .

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 2024 Thebiotek. All Rights Reserved.