Pyridoxine 5′-phosphate oxidase (PNPO) is an enzyme critical for synthesizing pyridoxal 5′-phosphate (PLP), the bioactive form of vitamin B6. PNPO antibodies are immunochemical tools designed to detect and quantify PNPO in research and diagnostic settings. These antibodies enable studies on PNPO’s role in diseases such as cancer, epilepsy, and metabolic disorders .
PNPO antibodies have been pivotal in identifying PNPO overexpression in cancers and its role in tumor progression:
Mechanistic Insights:
Epilepsy: PNPO mutations (e.g., R225H) reduce PLP synthesis, causing seizures. Antibodies validated PNPO expression in Drosophila models .
Prognostic Biomarker: High PNPO levels correlate with advanced tumor stages in breast and ovarian cancers .
Therapeutic Target:
Proteintech 15552-1-AP:
Abcam ab203745:
Lysis: Use RIPA buffer for protein extraction.
Electrophoresis: 10% SDS-PAGE.
Transfer: PVDF membrane, 100 V for 1.5 hr.
Blocking: 5% non-fat milk, 1 hr.
Pyridoxamine 5'-phosphate oxidase (PNPO) is a crucial enzyme in vitamin B6 metabolism that catalyzes the oxidation of pyridoxine 5'-phosphate and pyridoxamine 5'-phosphate to pyridoxal 5'-phosphate (PLP), which serves as a cofactor for numerous enzymatic reactions. PNPO has a calculated and observed molecular weight of approximately 30 kDa and is encoded by the PNPO gene (Gene ID: 55163) . This enzyme is particularly significant in research due to its involvement in essential metabolic pathways and its association with neurological disorders. PNPO deficiency leads to abnormal results for amino acid and neurotransmitter analyses in urine, plasma, or cerebrospinal fluid (CSF), reflecting secondary dysfunction of different metabolic pathways dependent on PLP as a cofactor . Researchers study PNPO to better understand vitamin B6 metabolism and related neurological conditions, making PNPO antibodies valuable tools for investigating this enzyme's expression, localization, and function in various tissues and cell types.
Most commercially available PNPO antibodies demonstrate reactivity with human, mouse, and rat samples, making them suitable for comparative studies across these species . For instance, PNPO antibody 15552-1-AP has been positively tested in human tissues (brain, heart, liver), mouse brain tissue, and rat brain tissue for Western blotting applications . Similarly, PNPO Antibody OTI1G9 shows reactivity with human, mouse, and rat samples for applications including flow cytometry and immunohistochemistry . When planning cross-species experiments, it is advisable to verify the specific reactivity of your chosen antibody through the manufacturer's validation data. Some antibodies may have limited species reactivity; for example, certain monoclonal antibodies like ABIN782454 might recognize only human PNPO without confirmed cross-reactivity in other species . Therefore, researchers should carefully select antibodies based on their experimental model organisms and verify reactivity through preliminary validation experiments.
PNPO antibodies are versatile tools that can be used in multiple experimental applications:
When selecting a PNPO antibody for a specific application, researchers should consider the validation data provided by manufacturers and optimize the antibody concentration for their specific experimental conditions. It is recommended to titrate the antibody in each testing system to obtain optimal results, as the optimal concentration may be sample-dependent .
Validating antibody specificity is crucial for ensuring reliable research results. For PNPO antibodies, a comprehensive validation strategy should include:
Positive and negative control samples: Use tissues known to express PNPO (such as liver, brain, heart) as positive controls . For negative controls, consider PNPO knockout cell lines or PNPO-depleted samples using siRNA.
Multiple detection methods: Validate findings using at least two independent methods (e.g., Western blot and immunohistochemistry) to confirm consistency of results.
Molecular weight verification: Confirm that the detected band in Western blots corresponds to the expected molecular weight of PNPO (30 kDa) .
Pre-absorption controls: Incubate the antibody with excess purified PNPO protein before immunostaining to demonstrate binding specificity.
Cross-reactivity assessment: If working with mouse samples using mouse monoclonal antibodies, researchers should be aware of potential high background signal in IHC and ICC experiments. Mouse-On-Mouse blocking reagents may be necessary to reduce this background . This is particularly important when using antibodies like OTI1G9, which is a mouse monoclonal antibody that reacts with mouse PNPO.
The experimental validation should be systematically documented, as antibody performance can vary between lots and applications. Researchers should maintain detailed records of validation experiments to ensure reproducibility and reliability of their results.
Sample preparation protocols vary depending on the application and sample type:
For Western Blotting (WB):
Tissue homogenization or cell lysis should be performed in appropriate lysis buffers containing protease inhibitors.
For PNPO detection, successful results have been obtained from MCF-7 cells, human brain tissue, human heart tissue, human liver tissue, mouse brain tissue, and rat brain tissue .
Standard protein quantification methods should be used to ensure equal loading.
Samples should be denatured at 95-100°C for 5 minutes in sample buffer containing SDS and a reducing agent.
Recommended antibody dilutions range from 1:500 to 1:3000, but optimal concentration should be determined experimentally .
For Immunohistochemistry (IHC):
For human liver tissue, antigen retrieval with TE buffer pH 9.0 is recommended; alternatively, citrate buffer pH 6.0 may be used .
Standard deparaffinization and rehydration protocols should be followed for paraffin-embedded tissues.
Blocking with appropriate serum (typically 5-10% normal serum from the same species as the secondary antibody) is recommended.
Primary antibody dilutions typically range from 1:20 to 1:200 .
For Immunofluorescence (IF)/ICC:
Cells should be fixed with 4% paraformaldehyde and permeabilized with 0.1-0.5% Triton X-100.
Blocking with appropriate serum is recommended to reduce non-specific binding.
Primary antibody dilutions typically range from 1:10 to 1:100 .
For Flow Cytometry:
Cells should be fixed and permeabilized using standard protocols.
Optimal antibody dilution should be determined experimentally .
For each protocol, researchers should optimize conditions based on their specific sample types and experimental goals. The inclusion of appropriate positive and negative controls is essential for accurate interpretation of results.
When studying PNPO deficiency, researchers should employ a multi-faceted approach combining antibody-based techniques with metabolic and genetic analyses:
Immunoblotting for protein expression: Western blotting with PNPO antibodies can assess protein expression levels in patient-derived samples compared to controls. This technique can help determine if mutations affect protein stability or expression levels.
Immunohistochemistry for tissue localization: IHC can reveal altered expression patterns or subcellular localization of PNPO in affected tissues. For human liver tissue samples, antigen retrieval with TE buffer pH 9.0 is recommended, with antibody dilutions ranging from 1:20 to 1:200 .
Complementary metabolic analyses: Since PNPO deficiency affects multiple metabolic pathways, researchers should correlate antibody-based findings with metabolic markers. These may include:
Genetic correlation: Antibody studies should be correlated with genetic findings. For instance, mutations such as the homozygous missense G>A transition at nucleotide 284 in exon 3 (c.284G>A) or C>T transition at nucleotide 283 in exon 3 causing an R95C substitution have been identified in PNPO deficiency cases .
Functional assays: Combine antibody detection with functional assays measuring PNPO enzymatic activity to establish the relationship between protein expression and function.
Researchers should be aware that some patients with molecular confirmation of PNPO deficiency may show normal or atypical amino acid or neurotransmitter profiles , highlighting the importance of a comprehensive approach combining antibody-based protein detection with metabolic and genetic analyses.
Cross-reactivity can significantly impact experimental results when using PNPO antibodies. To address these issues:
Select appropriate antibody formats: When using mouse monoclonal antibodies (such as OTI1G9) on mouse tissues, researchers should be aware of potential high background signal in IHC and ICC experiments. Mouse-On-Mouse blocking reagents may be necessary to reduce this background . Specific blocking reagents like those under catalog numbers PK-2200-NB and MP-2400-NB can be utilized.
Validation with multiple antibodies: Use antibodies raised against different epitopes of PNPO to confirm findings. Compare results from polyclonal antibodies (like 15552-1-AP, which is a rabbit polyclonal ) with those from monoclonal antibodies (like AT2C7, which is a mouse monoclonal ) to ensure consistency.
Pre-absorption controls: Pre-incubate the antibody with excess purified antigen before immunostaining to confirm specificity. Any remaining signal after pre-absorption might indicate cross-reactivity.
Knockout/knockdown controls: Use PNPO knockout or knockdown samples to verify antibody specificity. Any signal in these samples may represent cross-reactivity.
Optimize blocking conditions: Increase the concentration of blocking agents (BSA, serum, or commercial blocking solutions) and extend blocking time to reduce non-specific binding.
Adjust antibody concentration: Titrate antibody dilutions to find the optimal concentration that maximizes specific signal while minimizing background. For WB applications with PNPO antibody 15552-1-AP, dilutions ranging from 1:500 to 1:3000 are recommended .
Secondary antibody controls: Include secondary-only controls to ensure that non-specific binding is not due to the secondary antibody.
By implementing these strategies, researchers can minimize cross-reactivity issues and obtain more reliable results when using PNPO antibodies.
The choice between polyclonal and monoclonal PNPO antibodies depends on several factors:
Recommendations for specific applications:
For Western Blotting: Both polyclonal and monoclonal antibodies can be effective. Polyclonal antibodies like 15552-1-AP (1:500-1:3000 dilution) may provide higher sensitivity , while monoclonals may offer cleaner results with less background.
For Immunohistochemistry: Consider the nature of the sample and fixation method. Polyclonal antibodies may be more tolerant of fixation-induced epitope modifications. For human liver tissue using 15552-1-AP, antigen retrieval with TE buffer pH 9.0 is recommended, with dilutions of 1:20-1:200 .
For Flow Cytometry: Monoclonal antibodies like OTI1G9 are often preferred due to their consistency and lower background.
For Co-localization Studies: Monoclonal antibodies from different host species allow easier multi-color staining with minimal cross-reactivity.
For Detecting Low-Abundance Proteins: Polyclonal antibodies may provide higher sensitivity due to recognition of multiple epitopes.
The optimal choice depends on the specific research question, sample type, and experimental conditions. When possible, validating findings with both monoclonal and polyclonal antibodies can provide more robust results.
Optimizing Western blot protocols for PNPO detection requires careful consideration of several parameters:
Sample Preparation:
Gel Percentage and Transfer Conditions:
Antibody Selection and Dilution:
Blocking Conditions:
Use 5% non-fat dry milk or BSA in TBST for blocking
Block for 1-2 hours at room temperature or overnight at 4°C
The choice between milk and BSA may affect background; test both if necessary
Primary Antibody Incubation:
Washing Steps:
Perform at least 3-5 washes with TBST, 5-10 minutes each
Thorough washing is critical for reducing background
Secondary Antibody Selection:
Use species-appropriate HRP-conjugated secondary antibodies
Dilute according to manufacturer's recommendations (typically 1:2000-1:10000)
Incubate for 1-2 hours at room temperature
Detection Method:
ECL substrates with different sensitivities are available; choose based on expected expression level
For low expression, consider enhanced chemiluminescence substrates
Optimize exposure time based on signal strength
Stripping and Reprobing:
If reprobing is necessary, use mild stripping buffers to preserve the membrane
Verify complete stripping before reprobing
Quantification:
Use appropriate software for densitometric analysis
Normalize PNPO expression to loading controls (e.g., GAPDH, β-actin)
By systematically optimizing these parameters, researchers can achieve specific and sensitive detection of PNPO in Western blot experiments. It is recommended to maintain detailed records of optimization steps to ensure reproducibility.
PNPO deficiency is associated with various neurological disorders, particularly neonatal epileptic encephalopathy. Researchers can use PNPO antibodies to investigate these relationships through several methodological approaches:
Comparative Tissue Expression Analysis:
Use immunohistochemistry with PNPO antibodies (e.g., 15552-1-AP at 1:20-1:200 dilution) to compare PNPO expression in brain tissues from patients with neurological disorders versus controls
Map the expression pattern of PNPO in different brain regions to identify areas most affected by PNPO deficiency
Correlate expression patterns with clinical symptoms and disease severity
Cellular Localization Studies:
Correlation with Metabolic Biomarkers:
Combine immunoblotting data with analysis of metabolic markers known to be affected in PNPO deficiency
Correlate PNPO protein levels with abnormalities in amino acids (elevated glycine, reduced arginine) and neurotransmitter metabolites (elevated homovanillic acid, vanillactic acid)
Assess the relationship between protein expression and severity of metabolic derangements
Genetic-Protein Expression Correlation:
Use PNPO antibodies to assess how specific mutations (such as c.284G>A or c.283C>T resulting in R95C substitution) affect protein expression, stability, and localization
Compare protein expression levels in cells expressing wild-type versus mutant PNPO
Develop cellular models expressing patient-specific mutations to assess functional consequences
Therapeutic Response Assessment:
Use PNPO antibodies to monitor changes in protein expression following treatment interventions (e.g., pyridoxal 5'-phosphate supplementation)
Correlate changes in protein expression with clinical improvement and normalization of metabolic parameters
Interaction Studies:
By integrating these approaches, researchers can gain comprehensive insights into the molecular mechanisms linking PNPO deficiency to neurological disorders, potentially identifying new therapeutic targets and biomarkers for disease progression and treatment response.
Successful immunohistochemistry (IHC) with PNPO antibodies requires tailored approaches for different tissue types:
Liver Tissue (Highest PNPO Expression):
Antigen retrieval with TE buffer pH 9.0 is specifically recommended for human liver tissue; alternatively, citrate buffer pH 6.0 may be used
For polyclonal PNPO antibody 15552-1-AP, use dilutions of 1:20-1:200
Include positive controls from known PNPO-expressing liver samples
Consider shorter primary antibody incubation times due to high expression levels
Brain Tissue:
PNPO has been positively detected in human and rodent brain tissue via Western blotting , suggesting IHC should be feasible
Optimize fixation conditions: overfixation may mask epitopes
For formalin-fixed paraffin-embedded (FFPE) samples, extended antigen retrieval may be necessary
Consider multiple antigen retrieval methods (heat-induced vs. enzymatic) to identify optimal conditions
Use fluorescent detection methods for co-localization with neuronal or glial markers
Heart Tissue:
PNPO has been detected in human heart tissue via Western blotting
Consider lower antibody concentrations than used for liver due to potentially lower expression
Background autofluorescence can be problematic; use appropriate blocking of endogenous peroxidase and biotin
Sudan Black B treatment can reduce autofluorescence if using fluorescent detection
Cell Lines (e.g., MCF-7, HeLa):
PNPO has been detected in MCF-7 cells (WB, IP) and HeLa cells (IF/ICC)
For cultured cells, shorter fixation times (10-15 minutes with 4% paraformaldehyde) are typically sufficient
Permeabilization optimization is critical: try different concentrations of Triton X-100 (0.1-0.5%) or alternative permeabilization agents
Universal Considerations:
Fixation: Optimize fixation time; overfixation can mask epitopes while underfixation can compromise tissue morphology
Blocking: Use 5-10% normal serum from the secondary antibody host species
Controls: Include tissue-specific positive and negative controls
Mouse tissues with mouse antibodies: When using mouse monoclonal antibodies on mouse tissues, employ Mouse-On-Mouse blocking reagents to reduce background
Multi-labeling: For co-localization studies, select antibodies raised in different host species
Optimization Strategy:
Test multiple antibody dilutions (start with manufacturer's recommendations)
Compare different antigen retrieval methods
Vary incubation times and temperatures
Document all optimization steps for reproducibility
Interpretation Considerations:
PNPO is an intracellular enzyme; expected staining pattern is cytoplasmic
Be aware of potential nuclear or membrane staining which may indicate non-specific binding
Quantify staining intensity using appropriate image analysis software for comparative studies
By adapting these methodological considerations to specific tissue types, researchers can optimize IHC protocols for PNPO detection across different experimental contexts.
Integrating PNPO antibody-based techniques with metabolomic analyses provides a powerful approach to understanding vitamin B6 metabolism disorders:
Correlation of Protein Expression with Metabolite Profiles:
Tissue-Specific Integrative Analysis:
Use immunohistochemistry (1:20-1:200 dilution) to map PNPO expression across different tissues
Perform tissue-specific metabolomics on the same samples
Create tissue-specific maps of PNPO expression and vitamin B6-dependent metabolite patterns
This approach can identify tissues most vulnerable to PNPO deficiency
Cellular Compartmentalization Studies:
Temporal Analysis During Disease Progression:
Monitor PNPO expression and metabolite profiles at different disease stages
Track changes in both protein expression and metabolite concentrations during disease progression or treatment
This temporal integration can identify early biomarkers and treatment response indicators
Multi-Enzyme Analysis:
Use antibodies against multiple enzymes in the vitamin B6 pathway alongside PNPO
Correlate expression patterns of multiple enzymes with comprehensive metabolomics
This approach can identify compensatory mechanisms or cascade effects in the pathway
Functional Validation Through Intervention Studies:
Manipulate PNPO expression (overexpression, knockdown) in cellular or animal models
Monitor resulting changes in metabolite profiles
Correlate degree of expression change with magnitude of metabolite alterations
Patient-Derived Sample Analysis:
Analyze PNPO protein levels in patient samples using antibody-based techniques
Perform metabolomics on the same samples
Correlate specific mutations (e.g., c.284G>A, c.283C>T) with protein expression patterns and metabolite profiles
This can help establish genotype-phenotype correlations at the molecular level
Data Integration and Modeling:
Develop mathematical models incorporating both protein expression data and metabolite concentrations
Use these models to predict metabolic consequences of altered PNPO expression
Validate model predictions with experimental interventions
This integrated approach provides a comprehensive understanding of how alterations in PNPO affect vitamin B6 metabolism at multiple levels, from molecular to systemic, and can guide the development of more targeted diagnostic and therapeutic strategies for PNPO deficiency and related disorders.
When validating a newly purchased PNPO antibody, researchers should perform a systematic series of experiments to ensure specificity, sensitivity, and reproducibility:
Western Blot Validation:
Test the antibody on positive control samples known to express PNPO (e.g., MCF-7 cells, human brain, heart, or liver tissue)
Verify that the detected band appears at the expected molecular weight (30 kDa for PNPO)
Test multiple antibody dilutions (e.g., 1:500, 1:1000, 1:3000 for PNPO antibody 15552-1-AP) to determine optimal concentration
Include negative controls (tissues or cells with low/no PNPO expression) if available
If possible, include PNPO knockout or knockdown samples as specificity controls
Peptide Competition Assay:
Pre-incubate the antibody with excess immunizing peptide or recombinant PNPO protein
Compare results of Western blot or IHC with and without peptide competition
Specific signals should be significantly reduced or eliminated after peptide competition
Cross-Platform Validation:
Confirm PNPO detection using multiple applications (WB, IHC, IF/ICC) as appropriate for the antibody's intended use
For PNPO antibody 15552-1-AP, test in WB (1:500-1:3000), IP (0.5-4.0 μg), IHC (1:20-1:200), and IF/ICC (1:10-1:100)
Compare staining/detection patterns across platforms to ensure consistency
Reproducibility Assessment:
Repeat key validation experiments at least three times
Test reproducibility across different lots of the antibody if possible
Document batch variation for future reference
Immunohistochemistry Optimization:
Test multiple antigen retrieval methods (e.g., TE buffer pH 9.0 and citrate buffer pH 6.0 for PNPO in human liver tissue)
Optimize blocking conditions to minimize background
Compare staining patterns with published literature on PNPO localization
If using mouse monoclonal antibodies on mouse tissues, test with Mouse-On-Mouse blocking reagents to reduce background
Immunoprecipitation Validation:
Cross-Reactivity Assessment:
Documentation and Reporting:
Create a detailed validation report including all experimental conditions
Document lot number, dilutions, exposure times, and all relevant experimental parameters
Include representative images from all validation experiments
Note any limitations or specific conditions required for optimal performance
Thorough validation ensures that subsequent experimental results will be reliable and reproducible. The validation process may also identify optimal conditions for each application, saving time and resources in future experiments.
When researchers encounter discrepancies between different PNPO antibodies used in the same experimental system, a systematic approach can help resolve these inconsistencies:
Epitope Mapping and Comparison:
Identify the epitopes recognized by each antibody
Different antibodies may target different regions of PNPO; for example, compare polyclonal antibody 15552-1-AP (targeting a fusion protein immunogen) with monoclonal antibody AT2C7 (targeting AA 57-261)
Epitope accessibility may vary depending on protein conformation, fixation methods, or post-translational modifications
Discrepancies may be explained by differential epitope recognition under specific experimental conditions
Cross-Validation with Orthogonal Methods:
Implement non-antibody-based detection methods (e.g., mass spectrometry, mRNA quantification)
Compare antibody-based results with these orthogonal approaches
This can help determine which antibody provides more accurate representation of PNPO expression
Isotype and Host Species Considerations:
Compare results from antibodies of different isotypes and host species (e.g., rabbit polyclonal IgG vs. mouse monoclonal IgG1 )
Test if secondary antibody cross-reactivity could explain discrepancies
For mouse tissues, mouse monoclonal antibodies may require Mouse-On-Mouse blocking reagents to reduce background
Sensitivity and Specificity Analysis:
Determine detection thresholds for each antibody through dilution series
Assess signal-to-noise ratios under identical conditions
Test specificity using PNPO knockout/knockdown samples or peptide competition assays
Higher affinity antibodies may detect lower levels of PNPO that others miss
Application-Specific Optimization:
Optimize conditions specifically for each antibody and application
For Western blotting, test different blocking agents, incubation times, and detection methods
For IHC/IF, compare different fixation and antigen retrieval methods (e.g., TE buffer pH 9.0 vs. citrate buffer pH 6.0)
Discrepancies may resolve when each antibody is used under its optimal conditions
Batch and Storage Effects:
Reproducibility Assessment:
Have multiple researchers perform the same experiments independently
Test in different laboratories if possible
Persistent discrepancies across multiple experimenters suggest genuine antibody differences rather than technical variation
Biological Variability Analysis:
Determine if discrepancies are consistent across different samples/tissues
Pattern-based discrepancies (e.g., differences only in certain cell types) may reflect biological reality rather than technical issues
Consider if antibodies might be detecting different isoforms or post-translationally modified versions of PNPO
Reporting and Documentation:
Document all conditions where discrepancies occur
Report findings to antibody manufacturers for their records
Consider publishing observations as technical notes to assist other researchers
By systematically investigating these factors, researchers can better understand the source of discrepancies and determine which antibody provides the most reliable results for their specific research question and experimental system.
Advances in PNPO antibody technology hold significant potential for deepening our understanding of vitamin B6-dependent enzymes in neurological disorders:
Development of Phospho-Specific PNPO Antibodies:
Future antibodies targeting specific phosphorylation sites on PNPO could reveal regulatory mechanisms
Phosphorylation state may affect PNPO activity in different neurological conditions
This approach could identify previously unknown regulatory pathways affecting vitamin B6 metabolism in the brain
Conformation-Specific Antibodies:
Antibodies recognizing specific conformational states of PNPO could distinguish between active and inactive forms
This would allow researchers to map the distribution of catalytically active PNPO in different brain regions
Changes in the ratio of active/inactive PNPO might serve as biomarkers for neurological disorders
Super-Resolution Microscopy Compatible Antibodies:
New antibody formats optimized for super-resolution microscopy techniques could reveal subcellular localization with unprecedented detail
This could identify specific neuronal compartments (synapses, dendrites, axons) where PNPO is most active
Alterations in nanoscale localization patterns might be early indicators of pathology
Mutation-Specific Antibodies:
Multiplexed Antibody Approaches:
Advanced multiplexing technologies allowing simultaneous detection of PNPO alongside multiple vitamin B6-dependent enzymes
This could create comprehensive maps of the "B6-dependent enzyme network" in health and disease
Patterns of coordinated changes across multiple enzymes might identify new disease subtypes
In vivo Imaging Antibody Derivatives:
Development of antibody fragments or derivatives that can cross the blood-brain barrier
These could enable in vivo imaging of PNPO in animal models or potentially humans
Real-time monitoring of PNPO distribution during disease progression or treatment
Integration with Single-Cell Technologies:
Antibodies optimized for single-cell analysis techniques like mass cytometry or CODEX
This would allow cell type-specific analysis of PNPO expression in complex neural tissues
Identification of particularly vulnerable neuronal populations in vitamin B6-related disorders
Therapeutic Antibody Applications:
Development of antibodies that can modulate PNPO activity
These could serve as experimental tools to manipulate vitamin B6 metabolism in specific tissues
Potential therapeutic applications in conditions where targeted modulation of PNPO activity might be beneficial
Proximity Labeling Applications:
Antibody-based proximity labeling techniques to identify proteins interacting with PNPO in neural tissues
This could reveal novel interaction partners specific to neuronal cells
Changes in the PNPO interactome in disease states might identify new therapeutic targets
Pyridoxamine 5’-phosphate oxidase (PNPO) is an enzyme that plays a crucial role in the metabolism of vitamin B6. It is responsible for the conversion of pyridoxamine 5’-phosphate (PMP) and pyridoxine 5’-phosphate (PNP) to pyridoxal 5’-phosphate (PLP), the active form of vitamin B6. This enzyme is essential for various biological processes, including amino acid metabolism, neurotransmitter synthesis, and gene expression regulation.
PNPO is a flavoprotein that requires flavin mononucleotide (FMN) as a cofactor for its enzymatic activity. The enzyme is encoded by the PNPO gene, which is located on chromosome 17 in humans. The PNPO protein consists of 261 amino acids and has a molecular weight of approximately 30 kDa .
The enzyme catalyzes the oxidation of PMP and PNP to PLP through a two-step reaction. In the first step, PMP or PNP is oxidized to pyridoxal 5’-phosphate (PLP) and hydrogen peroxide (H2O2). In the second step, the hydrogen peroxide is reduced to water, completing the reaction cycle .
PLP, the product of the PNPO-catalyzed reaction, is a vital coenzyme involved in numerous enzymatic reactions. It participates in the metabolism of amino acids, the synthesis of neurotransmitters such as serotonin, dopamine, and gamma-aminobutyric acid (GABA), and the regulation of gene expression. PLP deficiency can lead to various health issues, including neurological disorders, anemia, and impaired immune function .
Mouse anti-human PNPO antibodies are valuable tools in various research applications, including: