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) .
Property | Value/Description |
---|---|
Molecular Mass | 30.3 kDa |
Amino Acids | 285 (1–262 residues) |
Post-Translational Modifications | Non-glycosylated |
Purification Method | Proprietary chromatography |
Purity | >95% (SDS-PAGE) |
Formulation | 20 mM Tris-HCl (pH 8.5), 1 mM DTT, 10% glycerol |
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 .
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 .
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 (, ).
Explore cross-species binding interactions (e.g., human vs. mouse SPR).
Investigate therapeutic relevance in disease models.
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.
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.
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.
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.
The purity of the SPR protein is determined to be greater than 95% using SDS-PAGE analysis.
SDR38C1, SPR, Dystonia, Sepiapterin reductase, mCG_128676.
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.
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.
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.
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 .
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.
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:
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:
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.
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.
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.
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 Type | Source | Integration Method | Outcome Measure |
---|---|---|---|
SPR Binding Kinetics | Biacore T200 measurements | Statistical correlation | Binding parameters (KD, kon, koff) |
SNP Profiles | Mouse Phenome Database | Genomic mapping | Target protein variation |
Strain Phenotypes | Mouse Phenome Database | Multivariate analysis | Phenotypic correlates of binding |
Gene Expression | Tissue-specific transcriptomics | Network analysis | Context-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 .
Common artifacts in SPR measurements with mouse antibodies and their remediation strategies include:
Mass transport limitation:
Baseline drift:
Heterogeneous binding profiles:
Regeneration damage:
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:
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.
Validating the phenotypic characteristics of SPR-knockout mice requires a comprehensive multi-level approach:
Genetic validation:
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:
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:
Response to intervention:
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.
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:
Pre-injection processing:
Reference sample preparation:
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:
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.
Interpreting kinetic binding data from SPR experiments with mouse proteins requires systematic analysis and consideration of several factors:
Kinetic parameter extraction:
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:
Concentration series validation:
Reference-relative interpretation:
Biological context integration:
This structured approach to data interpretation enables researchers to extract maximum biological significance from SPR kinetic data while avoiding common pitfalls in analysis.
Integrating SPR mouse model data with human disease databases requires sophisticated bioinformatic approaches that bridge species differences while maintaining biological relevance:
Ortholog mapping frameworks:
Phenotype ontology alignment:
Pathway-centric integration methods:
Multi-omics data integration:
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 Type | Mouse Source | Human Source | Integration Method |
---|---|---|---|
Genomic | MPD SNP data | ClinVar/OMIM | Synteny mapping |
Transcriptomic | SPR−/− tissues | Patient biopsies | Co-expression networks |
Metabolomic | BH4/neurotransmitter profiles | CSF biomarkers | Metabolic pathway mapping |
Phenotypic | Standardized phenotyping | HPO annotations | Semantic 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 .
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:
Competitive binding validation:
Multiple capture formats:
Advanced data processing approaches:
Sample pre-treatment optimization:
Controls for tissue extract validation:
Quantitative assessment metrics:
Assessment Approach | Specific Binding Indicator | Non-Specific Binding Indicator |
---|---|---|
Concentration dependence | Saturable response | Linear, non-saturable response |
Competition assays | >80% signal reduction | <20% signal reduction |
Kinetic profiles | Defined association/dissociation | Rapid association with minimal dissociation |
Buffer sensitivity | Minimal impact of mild detergents | Significant 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 .
Several emerging technologies are significantly enhancing the application of SPR mouse models in personalized medicine research:
Microfluidic SPR array platforms:
CRISPR-engineered precision mouse models:
Single-cell SPR technologies:
Integrated multi-omics approaches:
Organ-on-chip SPR integration:
AI-augmented SPR data analysis:
Humanized SPR mouse models:
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.
Longitudinal studies with SPR-deficient mice can significantly inform therapeutic development for human disorders through several methodological approaches:
Developmental trajectory mapping:
Biomarker evolution profiling:
Treatment timing optimization:
Combination therapy assessment:
Compensatory mechanism identification:
Long-term safety evaluation:
Treatment discontinuation studies:
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.
Combining SPR technology with mouse genomic editing creates powerful approaches for target validation with numerous applications:
Precision binding site verification:
Mechanism-based safety assessment:
Resistance mechanism characterization:
Allosteric modulator development:
Isoform-specific targeting:
Biomarker qualification:
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.
Translating SPR mouse model findings to human applications requires careful consideration of several critical factors to ensure relevance and accuracy:
Species-specific metabolic differences:
Genetic background effects:
Developmental timing disparities:
Physiological parameter scaling:
Endpoint translation:
Refinement of preclinical modeling:
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.
Researchers can build effective collaborative networks to advance SPR mouse research through strategic approaches that leverage complementary expertise and resources:
Cross-disciplinary partnership structures:
Resource sharing platforms:
Standardized methodological frameworks:
Translational research infrastructure:
Technology integration initiatives:
Funding mechanism utilization:
Knowledge dissemination strategies:
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.
Ethical considerations guiding the development and use of SPR mouse models should address several key dimensions:
Refinement of experimental design:
Reduction strategies:
Replacement considerations:
Severity classification and mitigation:
Genetic modification considerations:
Translational value assessment:
Transparency and reporting standards:
End-user engagement:
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 .
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 .
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.
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 .