PSPH antibodies are immunoglobulin-based reagents designed to recognize phosphoserine phosphatase, an enzyme encoded by the PSPH gene (NCBI Gene ID: 5723). PSPH converts O-phospho-L-serine to L-serine, a precursor for neurotransmitters, nucleotides, and antioxidants like glutathione . Commercial antibodies are available in monoclonal and polyclonal formats, with applications spanning Western blot (WB), immunohistochemistry (IHC), immunoprecipitation (IP), and immunofluorescence (IF/ICC) .
Epitopes: Most antibodies target recombinant fragments (e.g., amino acids 1–200 or 1–225) .
Molecular Weight: Detects ~25 kDa bands in WB, consistent with PSPH’s calculated mass .
Validation: Knockout cell lines (e.g., A549 PSPH-KO) confirm specificity .
Non-Canonical Pathways: PSPH interacts with IRS-1 to promote lung cancer progression independently of L-serine synthesis .
Immune Evasion: In hepatocellular carcinoma, PSPH upregulates CCL2 (attracting macrophages) while downregulating CXCL10 (reducing CD8+ T cells) .
PSPH (Phosphoserine phosphatase) is an enzyme that catalyzes the final step in the biosynthesis of serine, specifically the conversion of L-phosphoserine to L-serine. Antibodies targeting PSPH serve as critical tools for several fundamental research purposes:
Detection and quantification of PSPH protein expression across various cell and tissue types
Visualization of subcellular localization patterns within cellular compartments
Investigation of PSPH's role in serine metabolism and interconnected metabolic pathways
Examination of PSPH's potential involvement in pathological conditions where serine metabolism is altered
The centrality of PSPH in cellular metabolism makes antibodies against this protein valuable for understanding basic biochemical processes, disease mechanisms, and potential therapeutic targets. Commercial antibodies like the rabbit polyclonal Anti-PSPH Antibody (A39888) have been validated for Western blot and immunofluorescence applications with human samples .
The abbreviation PSP can create confusion in scientific literature as it may refer to different proteins:
PSPH: Phosphoserine phosphatase, an enzyme involved in serine biosynthesis
PSP: Parotid Secretory Protein, associated with salivary function and implicated in Sjögren's syndrome diagnosis
When reviewing literature or selecting antibodies, researchers should carefully verify:
The full protein name and context of the study
The specific immunogen used to raise the antibody
The molecular weight of the target (approximately 25 kDa for human PSPH)
The expected tissue expression pattern
In clinical contexts, PSP antibodies (anti-parotid secretory protein) are studied alongside other early biomarkers (SP1, CA6) in Sjögren's syndrome diagnosis, though research indicates these early autoantibodies underperform compared to classical markers . Meanwhile, PSPH antibodies are primarily used in basic research to study serine metabolism .
Comprehensive validation of PSPH antibodies should include the following methodological approaches:
Specificity Assessment:
Western blot analysis confirming detection of the expected molecular weight protein
Testing across multiple cell lines with varying PSPH expression levels
Genetic validation using PSPH knockdown/knockout models to demonstrate signal specificity
Peptide competition assays where pre-incubation with immunizing antigen blocks detection
Application-Specific Optimization:
For Western blotting: Determine optimal antibody dilution (e.g., 1:1000 dilution as used for the A39888 antibody), blocking conditions, and detection methods
For immunofluorescence: Confirm expected subcellular localization and optimize fixation methods (e.g., paraformaldehyde fixation for HeLa cells at 1:200 dilution)
For immunohistochemistry: Test different antigen retrieval methods and compare with known expression patterns
Cross-Reactivity Evaluation:
Assessment in multiple species if cross-reactivity is expected
Testing against closely related phosphatases to confirm specificity
In silico analysis of epitope conservation across related proteins
This rigorous validation approach helps ensure that observed signals genuinely represent PSPH protein and increases confidence in experimental findings.
The choice between polyclonal and monoclonal PSPH antibodies significantly impacts experimental design and interpretation:
Polyclonal PSPH Antibodies (e.g., A39888 rabbit polyclonal):
Recognize multiple epitopes on the PSPH protein
Generally provide stronger signals due to binding at multiple sites
Better for detecting low-abundance PSPH protein
More robust against minor protein denaturation or modifications
Ideal for initial characterization and Western blotting
Require careful batch-to-batch validation
Monoclonal PSPH Antibodies:
Recognize a single epitope on the PSPH protein
Provide higher specificity for a particular region of PSPH
Yield more consistent results across experiments with less background
Better for distinguishing between closely related proteins or isoforms
Ideal for applications requiring standardization
May have reduced sensitivity compared to polyclonal antibodies
Methodological Adaptations:
When using polyclonal antibodies:
More stringent blocking conditions to reduce background
Lower antibody concentrations often sufficient
Greater attention to lot-to-lot variation
When using monoclonal antibodies:
May require more sensitive detection systems
Critical epitope accessibility considerations
More consistent performance in quantitative applications
The experimental goal should guide antibody selection: use polyclonal antibodies when maximum sensitivity is needed, and monoclonal antibodies when specificity and reproducibility are paramount.
For successful co-immunoprecipitation (co-IP) studies using PSPH antibodies, researchers should implement these methodological strategies:
Antibody Selection and Preparation:
Choose antibodies that recognize native (non-denatured) PSPH
Polyclonal antibodies often work well for co-IP as they bind multiple epitopes
Consider directly conjugating the PSPH antibody to beads to avoid co-elution of antibody chains
Optimized Lysis Conditions:
Use gentle, non-denaturing lysis buffers (RIPA or NP-40 based) to preserve protein-protein interactions
Include phosphatase inhibitors to maintain physiologically relevant phosphorylation states
Test different salt concentrations to balance between preserving interactions and reducing background
Critical Control Experiments:
Pre-clear lysates with protein A/G beads to reduce non-specific binding
Include isotype control antibodies to identify non-specific interactions
Perform reciprocal co-IPs with antibodies against identified interaction partners
Use PSPH-depleted samples as negative controls
Detection and Validation Strategies:
Optimize elution conditions for efficient release of PSPH complexes
Consider native elution for downstream functional assays or denaturing elution for mass spectrometry
Validate interactions through orthogonal methods like proximity ligation assays
Confirm biological relevance through functional studies
These methodological considerations maximize the specificity and efficiency of co-IP experiments, increasing confidence in identified PSPH interaction partners.
Polyspecificity (non-specific interactions) is a critical concern when using antibodies for research. Based on recent developments in antibody assessment, researchers should consider:
Evaluating Antibody Polyspecificity:
The PolySpecificity Particle (PSP) assay provides a sensitive flow cytometry-based method for evaluating antibody polyspecificity that overcomes limitations of traditional methods
This assay uses micron-sized magnetic beads coated with Protein A to capture and evaluate the polyspecificity of various antibodies
Methodological Approach to Reduce Non-Specific Binding:
Use well-defined protein reagents as blocking agents (ovalbumin shows highest classification accuracy at 94%)
Implement appropriate negative controls in each experiment
Test antibodies at very low concentrations (0.46–15 μg/mL) where non-specific interactions are minimized
Consider pre-absorbing antibodies with potential cross-reactive proteins
Data Interpretation:
Evaluate signal-to-noise ratios critically
Use multiple antibodies targeting different epitopes to confirm findings
Be cautious when interpreting signals from complex biological samples
Consider potential cross-reactivity when analyzing closely related phosphatases
Understanding and controlling for polyspecificity is essential when using PSPH antibodies, particularly in complex biological systems where multiple related phosphatases may be present.
PSPH antibodies can contribute to biomarker research and diagnostics through several methodological approaches:
Tissue Expression Profiling:
Use immunohistochemistry with validated PSPH antibodies to examine expression across normal and diseased tissues
Implement digital pathology for quantitative assessment of expression levels
Correlate expression with clinical parameters and disease progression
Multi-marker Panels:
Integrate PSPH antibody detection with other relevant biomarkers
In autoimmune conditions like Sjögren's syndrome, consider the context of other biomarkers (though PSP in this context refers to parotid secretory protein, not PSPH)
Develop standardized detection protocols for clinical implementation
Methodological Considerations for Diagnostic Applications:
Establish rigorous cutoff values through ROC curve analysis
Ensure analytical validation including intra- and inter-assay variability assessment
Implement proper normalization strategies for quantitative measurements
Consider automation for reproducibility in clinical settings
Novel Detection Approaches:
Explore highly sensitive detection methods like the PSP assay, which offers greater sensitivity than standard ELISAs
Consider multiplex approaches to simultaneously detect PSPH alongside other biomarkers
Evaluate microfluidic platforms for point-of-care applications
While PSPH itself has not been extensively validated as a clinical biomarker, the methodological principles apply to developing any antibody-based diagnostic approach. Additionally, researchers should note that in Sjögren's syndrome, PSP (parotid secretory protein) antibodies may serve as early disease markers, though they currently underperform compared to classical autoantibodies .
Cross-reactivity is a significant challenge when using PSPH antibodies in complex biological samples. Researchers can implement these methodological approaches to address this issue:
Comprehensive Cross-Reactivity Profiling:
Test antibodies against recombinant proteins with structural similarity to PSPH
Create a cross-reactivity matrix documenting specificity profiles across potential targets
Perform comparative analysis with multiple antibodies targeting different PSPH epitopes
Genetic Validation Strategies:
Use CRISPR/Cas9 knockout of PSPH to create definitive negative controls
Implement siRNA knockdown to validate signals in systems where knockout isn't feasible
Compare antibody performance in wild-type versus PSPH-depleted samples
Advanced Signal Discrimination Methods:
Employ dual-labeling approaches with antibodies targeting different PSPH epitopes
Implement proximity ligation assays requiring two antibodies in close proximity for signal
Consider super-resolution microscopy to improve specificity determination
Computational Approaches:
Apply spectral unmixing algorithms to separate PSPH signal from cross-reactive signals
Develop machine learning models trained on validated positive and negative samples
Implement deconvolution algorithms to resolve mixed signals in complex tissues
Technical Controls and Validation:
Include pre-absorption controls with immunizing peptide or recombinant PSPH
Compare results from antibody-based detection with orthogonal methods
Validate all critical findings with multiple antibodies and methodologies
These strategies can significantly reduce the impact of cross-reactivity on experimental outcomes and increase confidence in the specificity of detected signals.
The combination of PSPH antibody-based techniques with mass spectrometry (MS) creates powerful research workflows:
Antibody-Based Enrichment for MS:
Use immunoprecipitation with PSPH antibodies followed by MS (IP-MS) to identify the PSPH interactome
Implement on-bead digestion protocols to minimize sample loss during preparation
For quantitative comparisons, consider using stable isotope labeling strategies like SILAC or TMT
Cross-Linking Mass Spectrometry (XL-MS):
Stabilize transient PSPH interactions with chemical crosslinkers
Verify crosslinking efficiency using PSPH antibodies before MS analysis
Identify crosslinked peptides to map PSPH interaction interfaces at amino acid resolution
Post-Translational Modification Mapping:
Immunoprecipitate PSPH using specific antibodies
Use MS analysis to identify and quantify phosphorylation, acetylation, or other modifications
Correlate modifications with PSPH enzymatic activity and cellular function
Absolute Quantification Strategies:
Develop targeted MS assays with reference peptides for absolute PSPH quantification
Compare antibody-based quantification with MS-based methods for cross-validation
Establish calibration curves using recombinant PSPH standards
Spatial Analysis Integration:
Perform laser microdissection of PSPH-positive regions (identified by antibody staining) followed by MS
Correlate mass spectrometry imaging with PSPH immunohistochemistry on adjacent sections
Implement multiplex imaging mass cytometry with PSPH antibodies labeled with metal isotopes
These integrated approaches leverage the enrichment capabilities of PSPH antibodies while harnessing the analytical power of mass spectrometry, providing deeper insights into PSPH biology.
When studying PSPH within protein complexes, several methodological considerations are critical:
Sample Preparation for Complex Preservation:
Optimize gentle lysis conditions (digitonin, CHAPS, or low concentrations of NP-40)
Include stabilizers like glycerol to maintain complex integrity
Consider chemical crosslinking for stabilizing transient interactions
Validate complex preservation using known PSPH-interacting proteins
Antibody Selection for Complex Analysis:
Test multiple antibodies targeting different PSPH epitopes to identify those accessible in complexes
Consider the impact of post-translational modifications on epitope recognition
Validate that antibody binding doesn't disrupt complex integrity
Analytical Separation Strategies:
Implement Blue Native PAGE to separate intact complexes containing PSPH
Use size-exclusion chromatography to fractionate native complexes by size
Consider density gradient centrifugation for additional separation by complex density
In Situ Complex Detection:
Employ proximity ligation assays (PLA) to verify PSPH interactions in intact cells
Implement super-resolution microscopy with appropriate co-localization statistics
Use FRET-based approaches to confirm direct protein-protein interactions
Functional Validation:
Correlate the presence of PSPH in complexes with enzymatic activity
Design enzyme activity assays compatible with immunoprecipitated complexes
Validate functional significance through targeted disruption of specific interactions
These approaches help overcome the challenges of studying multi-protein complexes while maintaining their native properties, providing insights into PSPH's regulatory interactions and functional roles within larger protein assemblies.
PSPH antibodies enable several methodological approaches for investigating cancer metabolism:
Expression Profiling Across Cancer Types:
Use PSPH antibodies for tissue microarray analysis of diverse cancer types
Implement standardized immunohistochemistry scoring
Correlate expression with clinical outcomes and molecular subtypes
Metabolic Pathway Activation Assessment:
Apply multiplex immunohistochemistry to examine PSPH alongside other serine biosynthesis enzymes (PHGDH, PSAT1)
Quantify co-expression patterns to identify tumors with activated serine synthesis pathway
Integrate with metabolomic data to validate pathway activity
Tumor Heterogeneity Characterization:
Perform single-cell analysis using flow cytometry with PSPH antibodies
Identify and isolate PSPH-high subpopulations for subsequent metabolic profiling
Correlate PSPH expression with stemness markers and resistance phenotypes
Therapeutic Target Validation:
Monitor PSPH expression before and after treatment with metabolism-targeting therapies
Use PSPH antibodies to confirm target engagement of PSPH inhibitors
Develop companion diagnostic approaches for stratifying patients for serine metabolism-targeted therapies
Mechanistic Studies:
Use PSPH antibodies in ChIP experiments to identify transcription factors regulating PSPH
Implement PSPH proximity labeling approaches to map the local protein environment
Correlate PSPH expression with metabolic vulnerabilities through functional genomics
These approaches can advance understanding of serine metabolism's role in cancer and potentially identify new therapeutic strategies targeting this pathway.
When working with PSPH antibodies in autoimmune disease research, several interpretative considerations are important:
Distinguishing Between Different PSP Contexts:
Be aware that PSP in autoimmune literature often refers to Parotid Secretory Protein, not Phosphoserine phosphatase (PSPH)
In Sjögren's syndrome research, anti-PSP antibodies are studied as potential early biomarkers
Carefully verify which protein is being referred to in each research context
Biomarker Evaluation Methods:
In Sjögren's syndrome, early autoantibodies (including anti-PSP) showed lower sensitivity (55.6%) and specificity (26.9%) compared to classical markers
When evaluating biomarker performance, consider:
Sensitivity and specificity calculations against gold standard diagnostic criteria
Comparison with established biomarkers (e.g., anti-Ro/La antibodies for Sjögren's)
Context of clinical symptoms and other laboratory findings
Methodological Integration:
Consider multiplex approaches when studying autoantibody profiles
Integrate antibody studies with functional assessments of affected tissues
When possible, correlate antibody findings with histopathological features
Clinical Translation Considerations:
Standardize testing conditions and cutoff values
Implement appropriate controls for each assay
Consider the temporal dynamics of antibody development in disease progression
Understanding these contexts helps researchers appropriately design and interpret studies involving PSPH or PSP antibodies in autoimmune disease research.
Phosphoserine Phosphatase (PSPH) is an enzyme encoded by the PSPH gene in humans. This enzyme plays a crucial role in the biosynthesis of L-serine, an amino acid essential for various cellular functions. The mouse anti-human PSPH antibody is a monoclonal antibody derived from hybridization of mouse SP2/0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human PSPH .
The PSPH gene is located on chromosome 7 in humans and chromosome 5 in mice . The protein encoded by this gene belongs to a subfamily of phosphotransferases and is responsible for the third and final step in L-serine formation. This step involves the magnesium-dependent hydrolysis of L-phosphoserine to L-serine .
Deficiency in PSPH is associated with several disorders, including Phosphoserine Phosphatase Deficiency and Visual Epilepsy . Homozygous or compound heterozygous mutations in PSPH can cause Neu–Laxova syndrome . Moreover, downregulation of PSPH has been shown to potentiate tumor immune responses, making it a potential target for immune-based cancer therapies .
The mouse anti-human PSPH antibody is used in various research applications, including: