PPP5C monoclonal antibodies are developed using hybridoma technology, ensuring specificity for the PPP5C antigen. Key characteristics include:
PPP5C monoclonal antibodies bind to epitopes on the PPP5C protein, enabling:
Detection: Western blot (WB) validation at 1:500–1:1000 dilutions .
Functional Studies: Inhibition or activation studies via immunoprecipitation (IP) .
Dephosphorylation Assays: Monitoring PPP5C’s role in pathways like Wnt/β-catenin (via Dvl2 dephosphorylation) .
PPP5C overexpression is linked to tumor progression. Monoclonal antibodies have been used to:
Identify elevated PPP5C levels in breast and prostate cancers .
Study PPP5C’s interaction with oncogenic kinases (e.g., RAF1) .
PPP5C (Protein Phosphatase 5 Catalytic subunit) is a serine/threonine phosphatase belonging to the protein phosphatase catalytic subunit family. It functions primarily in pathways regulated by reversible phosphorylation at serine and threonine residues, many of which control cell growth and differentiation processes . The protein participates in signaling pathways responding to hormones and cellular stress conditions .
PPP5C is known to regulate several important biological processes:
Stress response signaling
Cell growth and differentiation regulation
Calcium channel activity modulation through interaction with ISOC (store-operated calcium channels)
Additionally, dysregulation of PPP5C has been implicated in disease states, with elevated levels potentially associated with breast cancer development .
PPP5C monoclonal antibodies are available in different clones with varying specifications for research applications. The following table summarizes key characteristics of two prominent clones:
| Specification | Clone OTI6C2 | Clone OTI2G2 |
|---|---|---|
| Applications | WB, IHC, FC | WB, IHC, IF |
| Recommended Dilutions | WB 1:250-500, IHC 1:50, FLOW 1:100 | WB 1:500-1000, IHC 1:50, IF 1:100 |
| Species Reactivity | Dog, Human, Monkey, Mouse, Rat | Dog, Human, Mouse, Rat |
| Host | Mouse | Mouse |
| Isotype | IgG2b | IgG1 |
| Immunogen | Full-length human recombinant PPP5C protein produced in HEK293T cells | Full-length human recombinant PPP5C protein produced in HEK293T cells |
| Concentration | 1 mg/ml | 0.57 mg/ml |
| Predicted Target Size | 56.7 kDa | 56.7 kDa |
Both antibodies recognize the full-length PPP5C protein and are purified using affinity chromatography (protein A/G) from either mouse ascites fluids or tissue culture supernatant .
To maintain optimal activity of PPP5C monoclonal antibodies, researchers should follow these evidence-based storage and handling protocols:
Temperature: Store antibodies at -20°C as received in the manufacturer's packaging .
Buffer composition: PPP5C antibodies are typically supplied in PBS (pH 7.3) containing 1% BSA, 50% glycerol, and 0.02% sodium azide . This formulation helps maintain stability during freeze-thaw cycles.
Stability period: When stored properly, the antibodies are stable for 12 months from the date of receipt .
Shipping conditions: The antibodies are shipped on blue ice to maintain integrity during transport .
Aliquoting: For frequent users, it's recommended to prepare small aliquots upon receipt to minimize freeze-thaw cycles.
Working dilutions: Prepare fresh working dilutions on the day of the experiment according to the recommended dilution ratios for specific applications (WB, IHC, FC, or IF) .
Safety considerations: Note that the buffer contains sodium azide, which is toxic. Handle with appropriate precautions and dispose of according to safety regulations.
Different experimental applications require specific protocols for optimal results with PPP5C monoclonal antibodies:
Western Blot (WB) Protocol:
Sample preparation: Prepare cell or tissue lysates using standard RIPA buffer with protease inhibitors.
Protein separation: Load 20-50 μg of protein per lane on 10-12% SDS-PAGE gels.
Transfer: Transfer proteins to PVDF or nitrocellulose membranes.
Blocking: Block membranes with 5% non-fat milk or BSA in TBST for 1 hour at room temperature.
Primary antibody: Dilute PPP5C antibody at 1:250-1000 (depending on clone) in blocking buffer and incubate overnight at 4°C.
Washing: Wash 3× with TBST, 5 minutes each.
Secondary antibody: Incubate with HRP-conjugated anti-mouse IgG at 1:5000 for 1 hour at room temperature.
Detection: Use enhanced chemiluminescence substrate.
Immunohistochemistry (IHC) Protocol:
Sample preparation: Use formalin-fixed, paraffin-embedded tissue sections (4-6 μm thick).
Deparaffinization and rehydration: Standard xylene and ethanol series.
Antigen retrieval: Heat-induced epitope retrieval in citrate buffer (pH 6.0).
Blocking: 3% hydrogen peroxide followed by protein block.
Primary antibody: Dilute PPP5C antibody at 1:50 and incubate overnight at 4°C.
Detection: Use a polymer detection system appropriate for mouse primary antibodies.
Counterstain: Hematoxylin.
Expected result: Variable staining patterns depending on tissue type and PPP5C expression levels.
Immunofluorescence (IF) Protocol:
Sample preparation: Fix cells with 4% paraformaldehyde for 15 minutes.
Permeabilization: 0.1% Triton X-100 for 10 minutes.
Blocking: 5% normal serum in PBS for 30 minutes.
Primary antibody: Dilute PPP5C antibody at 1:100 and incubate overnight at 4°C.
Secondary antibody: Fluorophore-conjugated anti-mouse IgG at 1:500 for 1 hour at room temperature.
Counterstain: DAPI for nuclear visualization.
Mounting: Anti-fade mounting medium.
Validating antibody specificity is critical for ensuring reliable research results. For PPP5C monoclonal antibodies, implement these validation strategies:
Genetic manipulation controls:
Knockdown validation:
Peptide competition assay:
Pre-incubate the antibody with excess purified recombinant PPP5C protein.
A specific antibody will show reduced or eliminated signal in subsequent applications.
Multiple antibody validation:
Signal correlation with known expression patterns:
Verify that detected signal corresponds with tissues/cells known to express PPP5C.
Check subcellular localization against known distribution patterns.
Molecular weight verification:
Cross-reactivity testing:
Test antibody against closely related phosphatases to ensure specificity.
When designing experiments using PPP5C monoclonal antibodies, include these essential controls:
Positive controls:
Negative controls:
Loading/technical controls:
Housekeeping protein detection (e.g., β-actin, GAPDH) for Western blots
Tissue-specific markers for IHC/IF to verify tissue integrity
Functional controls:
Dose-dependent controls:
Serial dilution of primary antibody to determine optimal concentration
Titration experiments to verify signal specificity
Cross-species validation:
Subcellular fractionation controls:
In studies examining PP5C subcellular localization, include markers for different cellular compartments
The high-resolution crystal structure of PPP5C at 1.6 Å provides critical insights into its catalytic mechanism and exceptional enzymatic efficiency:
The PPP5C catalytic domain contains a distinctive Asp271-M1:M2-W1-His427-His304-Asp274 motif that is essential for its phosphatase activity . This arrangement creates a unique metal-binding pocket that orchestrates the hydrolysis of phosphoprotein substrates through:
Bimetal mechanism: Two metal ions (M1 and M2, typically Mn2+ or Fe2+) are precisely positioned within the catalytic pocket . These metals:
Activate a water molecule (W1) for nucleophilic attack
Stabilize the transition state during phosphate hydrolysis
Coordinate with conserved aspartate and histidine residues
Nucleophilic activation: The catalytic water molecule (W1) positioned between the two metal ions becomes highly nucleophilic, enabling attack on the phosphorus atom of the substrate .
Substrate binding pocket: The structure reveals a surface groove that accommodates phosphoprotein substrates, with specificity determined by:
The depth and width of the binding pocket
Charge distribution within the catalytic site
Interactions with conserved residues in the active site
C-terminal subdomain: The structural analysis revealed a previously unidentified subdomain in the C-terminus that likely contributes to:
Understanding this structural basis helps explain why PPP5C and related phosphatases are among the most catalytically efficient enzymes known, with implications for designing specific inhibitors for research and potential therapeutic applications .
PPP5C plays a crucial role in calcium channel regulation and endothelial barrier function through several molecular mechanisms:
ISOC channel regulation:
PPP5C is an essential component of the store-operated calcium channel (ISOC) heterocomplex .
It co-localizes with TRPC4, an essential subunit of ISOC channels, as demonstrated by co-precipitation studies .
PPP5C is found in the same membrane fractions as the ISOC heterocomplex in pulmonary artery endothelial cells (PAECs) .
FKBP51-mediated inhibition:
Catalytic activity requirement:
Endothelial barrier protection:
Potential mechanisms:
This research has significant implications for understanding vascular permeability regulation and potential therapeutic targets for conditions involving endothelial barrier dysfunction, such as acute respiratory distress syndrome or pulmonary edema.
PPP5C antibodies can be employed in sophisticated experimental approaches to elucidate its role in cellular stress response pathways:
Stress-induced phosphorylation changes:
Use PPP5C antibodies in conjunction with phospho-specific antibodies to analyze how various stressors (oxidative stress, heat shock, DNA damage) affect PPP5C substrates.
Implement quantitative Western blot analysis to track temporal changes in PPP5C expression and localization following stress induction.
Compare these changes between wild-type cells and those expressing catalytically inactive PPP5C mutants .
Protein-protein interaction networks:
Perform co-immunoprecipitation with PPP5C antibodies followed by mass spectrometry to identify stress-specific interaction partners.
Use proximity ligation assays (PLA) to visualize and quantify in situ interactions between PPP5C and stress response proteins like heat shock proteins.
Investigate the dynamic assembly of PPP5C-containing complexes through size-exclusion chromatography followed by Western blotting.
Subcellular trafficking:
Track PPP5C redistribution during stress responses using immunofluorescence with PPP5C antibodies .
Implement live-cell imaging with fluorescently-tagged PPP5C in combination with immunostaining of fixed time points.
Use subcellular fractionation followed by Western blotting to quantify PPP5C redistribution between cellular compartments during stress.
MAPK pathway regulation:
Since PPP5C participates in MAPK signaling , use PPP5C antibodies to investigate its association with pathway components during stress.
Perform phosphatase activity assays on immunoprecipitated PPP5C to measure stress-induced changes in enzymatic activity.
Compare phosphorylation states of MAPK pathway components between normal and PPP5C-depleted cells under stress conditions.
Hormonal stress responses:
Investigate PPP5C's role in glucocorticoid receptor (GR) signaling using hormone stimulation experiments.
Monitor PPP5C-GR interactions using co-immunoprecipitation with PPP5C antibodies.
Analyze how stress-induced hormonal changes affect PPP5C expression, localization, and activity.
ChIP-seq applications:
Use chromatin immunoprecipitation (ChIP) with PPP5C antibodies to identify potential chromatin-associated functions during stress responses.
Investigate whether PPP5C directly or indirectly regulates stress-responsive gene expression.
Researchers may encounter several technical challenges when working with PPP5C monoclonal antibodies. Here are common issues and evidence-based solutions:
High background in immunostaining:
Cause: Insufficient blocking, excessive antibody concentration, or non-specific binding
Solution:
Multiple bands in Western blot:
Cause: Cross-reactivity, degradation products, splice variants, or post-translational modifications
Solution:
Weak or no signal:
Cause: Low expression levels, epitope masking, over-fixation, or antibody degradation
Solution:
Optimize antigen retrieval for IHC (try citrate buffer pH 6.0 vs. EDTA buffer pH 9.0)
Increase antibody concentration or incubation time
Try different lysis buffers for Western blot
Test antibody on positive control samples with known PPP5C expression
Verify antibody stability and storage conditions
Inconsistent results between experiments:
Cause: Antibody batch variation, protocol inconsistency, or sample handling
Solution:
Poor immunoprecipitation efficiency:
Cause: Insufficient antibody-antigen binding, buffer incompatibility
Solution:
Pre-clear lysates to reduce non-specific binding
Optimize antibody-to-bead and antibody-to-lysate ratios
Extend binding time (overnight at 4°C)
Try different detergent concentrations in IP buffer
Distinguishing PPP5C from other similar phosphatases requires careful experimental design and multiple validation approaches:
Structural and sequence-based discrimination:
PPP5C has distinctive tetratricopeptide repeat (TPR) domains in its N-terminus that differentiate it from other phosphatases .
Its molecular weight (56.7 kDa) differs from other phosphatases, aiding identification in Western blots.
PPP5C's unique C-terminal subdomain revealed by crystallography can be targeted with domain-specific antibodies.
Antibody-based discrimination:
Use antibodies generated against unique epitopes not present in other phosphatases.
Validate antibody specificity through immunoprecipitation followed by mass spectrometry.
Perform cross-reactivity tests with recombinant proteins of related phosphatases.
Functional discrimination:
Localization-based discrimination:
Molecular genetic approaches:
Biochemical separation:
Use ion exchange chromatography to separate phosphatases based on charge differences.
Implement size exclusion chromatography to distinguish PPP5C-containing complexes.
Design immunodepletion experiments to selectively remove PPP5C from samples.
Association with specific binding partners:
Several quantitative methods can be employed to analyze PPP5C expression and activity with high precision:
Expression quantification methods:
Western blot densitometry:
Suitable for semi-quantitative analysis of PPP5C protein levels.
Normalize to appropriate loading controls (β-actin, GAPDH).
Use digital imaging systems with linear dynamic range.
Implement standard curves with recombinant PPP5C protein for absolute quantification.
qRT-PCR:
Quantify PPP5C mRNA expression levels.
Design primers spanning exon-exon junctions to avoid genomic DNA amplification.
Use the 2^(-ΔΔCT) method with appropriate reference genes.
ELISA:
Develop sandwich ELISA using two different epitope-targeting antibodies.
Establish standard curves with recombinant PPP5C protein.
Offers higher throughput than Western blotting.
Mass spectrometry:
For absolute quantification, implement multiple reaction monitoring (MRM) or parallel reaction monitoring (PRM).
Use isotopically labeled peptide standards (AQUA peptides) specific to PPP5C.
Enables simultaneous quantification of post-translational modifications.
Activity quantification methods:
Phosphatase activity assays:
Measure activity using artificial substrates like p-nitrophenyl phosphate (pNPP).
For more specificity, use phosphopeptides based on known PPP5C substrates.
Include controls with catalytically inactive PPP5C mutants .
Cellular reporter systems:
Develop FRET-based biosensors for real-time monitoring of PPP5C activity.
Use phosphorylation-responsive luciferase reporters linked to PPP5C substrates.
Implement these systems in wild-type versus PP5C-/- backgrounds .
Phospho-specific Western blotting:
Monitor phosphorylation states of known PPP5C substrates.
Quantify changes in phosphorylation upon PPP5C manipulation.
Include time-course analyses to capture dynamics.
Phosphoproteomics:
Compare global phosphorylation changes between normal and PPP5C-deficient cells.
Implement stable isotope labeling (SILAC or TMT) for quantitative comparison.
Focus on serine/threonine phosphorylation sites affected by PPP5C activity.
Localization and interaction quantification:
Quantitative immunofluorescence:
Measure PPP5C intensity in different subcellular compartments.
Use automated image analysis software for unbiased quantification.
Implement co-localization coefficients to quantify association with other proteins.
Proximity ligation assay (PLA):
Quantify in situ PPP5C interactions with proteins like TRPC4 or HSP90.
Provides spatial information about protein interactions.
Allows quantification of interaction events per cell.
FRAP (Fluorescence Recovery After Photobleaching):
Measure PPP5C mobility and binding dynamics in living cells.
Quantify changes in mobility upon cellular stress or stimulation.
PPP5C's involvement in multiple signaling pathways positions it as a potentially significant factor in various disease mechanisms and therapeutic strategies:
Cutting-edge methodologies are expanding our ability to study PPP5C phosphatase activity with unprecedented precision:
CRISPR-based approaches:
CRISPR-Cas9 gene editing has been used to generate PP5C-/- cell lines for functional studies .
Advanced applications include:
CRISPR activation (CRISPRa) and interference (CRISPRi) for temporal control of PPP5C expression
Base editing for introducing specific catalytic mutations without disrupting expression
Prime editing for precise modification of regulatory sequences
Optogenetic control systems:
Light-inducible PPP5C activation/inactivation systems allow:
Spatiotemporal control of phosphatase activity
Reversible modulation of activity in specific subcellular compartments
Real-time correlation between activity changes and cellular responses
Single-molecule approaches:
Super-resolution microscopy techniques (STORM, PALM) to visualize individual PPP5C molecules
Single-molecule FRET to monitor conformational changes during substrate binding
Single-cell phosphoproteomics to analyze cell-to-cell variability in PPP5C activity
Microfluidic platforms:
Droplet-based single-cell analysis of PPP5C activity
Gradient generators to study PPP5C response to varying concentrations of stimuli
Organ-on-chip models for tissue-specific PPP5C function analysis
Biosensor development:
Genetically encoded FRET-based sensors specific for PPP5C activity
Split-luciferase complementation systems for monitoring PPP5C-substrate interactions
Chemiluminescent probes for non-invasive imaging of PPP5C activity in vivo
Structural biology advances:
Systems biology approaches:
Network analysis of PPP5C interactome under different cellular conditions
Integration of phosphoproteomics, transcriptomics, and metabolomics data
Computational modeling of PPP5C-regulated signaling networks
Protein engineering methods: