PPP1R14A, also known as CPI-17 (17 kDa PKC-potentiated inhibitory protein of PP1), functions as a phosphorylation-dependent inhibitor of smooth muscle myosin phosphatase. When phosphorylated, it exhibits over 1,000 times greater inhibitory activity, effectively serving as a molecular switch that regulates the phosphorylation state of PPP1CA substrate and smooth muscle contraction .
The inhibition of myosin phosphatase by phosphorylated PPP1R14A leads to increased myosin phosphorylation, enhancing smooth muscle contraction without requiring elevated intracellular Ca²⁺ concentrations. This mechanism is central to calcium sensitization, where PPP1R14A enables increases in myosin phosphorylation and tension at constant calcium levels .
For maximum stability and retention of antibody activity, PPP1R14A antibodies should be stored at -20°C or lower temperatures . To prevent protein degradation from repeated freeze-thaw cycles, it is recommended to aliquot the antibody solution before freezing . When properly stored, most commercial PPP1R14A antibodies maintain their efficacy for approximately 12 months from the date of shipment .
Current research-grade PPP1R14A antibodies are available in multiple formats with distinct characteristics:
Host Species | Clonality | Clone Examples | Applications | Reference |
---|---|---|---|---|
Mouse | Monoclonal | F-4, 3A7, P4H10AT | WB, ELISA | |
Rabbit | Monoclonal | E305 | WB |
Mouse monoclonal antibodies offer high specificity and reproducibility between experimental batches. The 3A7 clone has been validated for western blotting and ELISA applications, with a detection limit for recombinant GST-tagged PPP1R14A of approximately 0.1ng/ml when used as a capture antibody . Rabbit monoclonal antibodies like E305 have been successfully used in western blot applications at dilutions of 1:2500 in mouse samples .
PPP1R14A exhibits a complex expression pattern across cancer types that challenges conventional tumor marker paradigms. Comprehensive pan-cancer analysis reveals that PPP1R14A is predominantly downregulated in major malignancies compared to matched normal tissues . This downregulation pattern is particularly notable in bladder urothelial carcinoma (BLCA), colon adenocarcinoma (COAD), and kidney renal papillary cell carcinoma (KIRP) .
For antibody-based detection methods, these findings necessitate:
Careful selection of control tissues that match the cancer stage being studied
Interpretation of immunohistochemistry results with consideration of this dual role
Potential use of PPP1R14A antibodies for diagnostic purposes, as ROC analysis indicates excellent diagnostic accuracy (AUC > 0.9) for multiple malignancies including BLCA, COAD, and KIRP
Resolving contradictory PPP1R14A expression patterns requires a multi-dimensional analytical framework:
Temporal dimension analysis: Separate studies of cancer initiation versus progression stages to delineate the transitional patterns of PPP1R14A expression. Evidence shows that while PPP1R14A is generally downregulated in tumor versus normal tissue comparison, its expression increases as cancer progresses in many cancer types .
Integrated multi-omics approach: Combine transcriptomic data with:
Methylation analysis: PPP1R14A promoter hypermethylation occurs in most common cancers, consistent with its initial downregulation
Phosphorylation studies: Examine phosphorylation levels at sites including S26 and T38, which show downregulation in breast cancer and colon cancer
Genetic alteration analysis: Assess copy number alterations and mutations, which reach 16.07% frequency in uterine carcinosarcoma and affect patient prognosis
Context-specific validation: For instance, in cholangiocarcinoma (CHOL) and head and neck squamous cell carcinoma (HNSC), high promoter methylation correlates with high transcription levels rather than suppression, suggesting non-canonical regulatory mechanisms that merit targeted investigation .
When designing antibody-based detection experiments, researchers should incorporate appropriate controls for each cancer type and stage, and validate findings using multiple methodological approaches.
Successful western blot analysis using PPP1R14A antibodies requires attention to several technical parameters:
Molecular weight interpretation: PPP1R14A detection can show varying molecular weights dependent on experimental conditions:
Antibody selection and dilution optimization:
Mouse monoclonal antibodies such as F-4 (Santa Cruz Biotechnologies, sc-48406) have been validated for mouse samples
Rabbit monoclonal antibody E305 (Abcam, ab32213) has been validated at 1:2500 dilution for mouse samples
Detection sensitivity varies by clone; the 3A7 clone can detect GST-tagged PPP1R14A at concentrations as low as 0.1ng/ml
Control selection: Include both positive and negative controls:
Studying PPP1R14A phosphorylation states is crucial given that phosphorylation increases its inhibitory activity over 1,000-fold and functions as a molecular switch in signaling pathways . Methodological approaches include:
Phospho-specific antibodies: While not explicitly mentioned in the search results, phospho-specific antibodies targeting key sites such as S26 and T38 would be essential for direct detection of phosphorylation states. These sites show altered phosphorylation levels in multiple cancer types .
Phosphatase treatment controls: Samples can be split and treated with/without phosphatases prior to western blotting with total PPP1R14A antibodies to determine the proportion of phosphorylated protein.
Phos-tag™ SDS-PAGE: This technique allows separation of phosphorylated and non-phosphorylated forms of proteins in acrylamide gels containing Phos-tag™ reagent, followed by detection with total PPP1R14A antibodies.
Mass spectrometry: For comprehensive phosphorylation site mapping, immunoprecipitation with PPP1R14A antibodies followed by mass spectrometry analysis provides detailed insight into multiple phosphorylation sites simultaneously.
Kinase/phosphatase assays: In vitro assays using purified PPP1R14A and relevant kinases (particularly PKC) or phosphatases, detected with total PPP1R14A antibodies, can establish phosphorylation/dephosphorylation kinetics.
The dual role of PPP1R14A in cancer biology necessitates sophisticated experimental designs:
The relationship between PPP1R14A promoter methylation and expression presents a complex regulatory landscape:
The data from these integrated analyses can help establish predictive models for when methylation will lead to silencing versus potential activation of PPP1R14A, informing both diagnostic applications of PPP1R14A antibodies and therapeutic strategies targeting epigenetic mechanisms.
PPP1R14A has been found to correlate significantly with levels of immune infiltrating cells and immune checkpoint genes , suggesting potential roles in tumor immunology. Methodological approaches include:
Multiplex immunofluorescence:
Co-stain tissue sections with PPP1R14A antibodies and markers for specific immune cell populations (CD8+ T cells, macrophages, etc.)
Quantify spatial relationships between PPP1R14A-expressing tumor cells and immune infiltrates
Correlate patterns with response to immunotherapy in retrospective patient cohorts
Flow cytometry:
Dissociate tumors into single-cell suspensions
Use PPP1R14A antibodies in combination with immune cell markers to quantify relationships between PPP1R14A expression and immune cell proportions
Sort cells based on PPP1R14A expression for downstream functional assays
Co-immunoprecipitation:
Use PPP1R14A antibodies to pull down protein complexes
Analyze interacting partners using mass spectrometry to identify potential interactions with immune signaling components
Confirm interactions using reciprocal co-IP and western blotting
In vitro co-culture systems:
Establish tumor cell lines with modulated PPP1R14A expression using CRISPR/Cas9 or siRNA
Co-culture with immune cells (T cells, macrophages)
Assess immune cell activation, cytokine production, and tumor cell killing
Use PPP1R14A antibodies to monitor expression in fixed and permeabilized cells via flow cytometry
Correlation with immunotherapy response biomarkers:
Analyze relationship between PPP1R14A expression (detected by antibodies) and established biomarkers like PD-L1, tumor mutational burden, and T cell infiltration
Stratify patient cohorts by PPP1R14A expression to retrospectively evaluate immunotherapy response rates
Researchers should be aware of several technical challenges when working with PPP1R14A antibodies:
Size discrepancies in western blots:
Phosphorylation state influence:
PPP1R14A function is heavily dependent on phosphorylation status
Solution: Consider using phospho-specific antibodies or Phos-tag™ gels to distinguish phosphorylated forms
Cancer context complexity:
Cross-reactivity concerns:
PPP1R14A belongs to a family of related inhibitor proteins
Solution: Validate antibody specificity using knockout/knockdown controls or peptide competition assays
Fixation sensitivity in immunohistochemistry:
Different fixation methods may affect epitope accessibility
Solution: Optimize antigen retrieval methods and validate with multiple antibody clones when possible
Selection of the optimal PPP1R14A antibody clone should be guided by the following considerations:
Application compatibility:
Species cross-reactivity:
Epitope accessibility:
Consider the structural context of your experiment (native vs. denatured protein)
Review the immunogen sequence used to generate the antibody to ensure epitope relevance
Published validation:
Clone-specific characteristics:
Where possible, preliminary testing of multiple antibody clones is recommended to identify the optimal reagent for your specific experimental system.
PPP1R14A antibodies could facilitate the development of targeted cancer therapies through several research pathways:
Biomarker development for precision medicine:
Target validation for drug development:
Antibody-based screening can identify cancer types with aberrant PPP1R14A function
Immunoprecipitation followed by mass spectrometry can reveal cancer-specific interacting partners
These interactions may represent novel therapeutic targets
Monitoring therapy response:
Serial biopsies analyzed with PPP1R14A antibodies can track expression changes during treatment
Changes in phosphorylation status may serve as pharmacodynamic markers
Potential antibody-drug conjugate (ADC) development:
For cancers where PPP1R14A is significantly upregulated during progression, modified therapeutic antibodies targeting surface-exposed epitopes could deliver cytotoxic payloads
Cancer type-specific expression patterns would need to be carefully validated
Combination therapy rational design:
Emerging technologies offer opportunities to expand PPP1R14A antibody applications:
Single-cell analysis:
Single-cell western blotting or CyTOF with PPP1R14A antibodies can reveal heterogeneity within tumors
This approach may help resolve seemingly contradictory bulk tissue findings regarding expression patterns
Spatial transcriptomics integration:
Combining immunohistochemistry using PPP1R14A antibodies with spatial transcriptomics can map expression patterns within the tumor microenvironment
This integrative approach could reveal functional niches where PPP1R14A plays critical roles
Organoid-based functional studies:
Patient-derived organoids representing different cancer stages can be analyzed with PPP1R14A antibodies
This system allows for controlled manipulation of PPP1R14A expression or phosphorylation to assess functional consequences
Intravital imaging:
Development of fluorescently labeled PPP1R14A antibody fragments for in vivo imaging
This approach could enable real-time tracking of expression changes during tumor evolution
Proximity labeling proteomics:
Fusion of PPP1R14A with proximity labeling enzymes (BioID, APEX) followed by detection with antibodies
This technique can map the PPP1R14A interactome in living cells under physiologically relevant conditions
The PPP1R14A gene is located on chromosome 19 in humans and chromosome 7 in mice . The protein consists of 147 amino acids and shares about 90% sequence identity with its porcine counterpart . The gene is expressed in various tissues, including the coronary arteries, aorta, and smooth muscle tissues .
PPP1R14A acts as a phosphorylation-dependent inhibitor of smooth muscle myosin phosphatase . When phosphorylated at the Thr-38 residue, it undergoes a conformational change that significantly increases its inhibitory activity . This inhibition leads to increased myosin phosphorylation and enhanced smooth muscle contraction, a phenomenon known as Ca²⁺ sensitization .
Multiple kinases, such as PKC, ROCK, PKN, ZIPK, ILK, and PAK, can phosphorylate PPP1R14A . Agonist stimulation of smooth muscle primarily enhances PPP1R14A phosphorylation through PKC and ROCK . In addition to its role in smooth muscle, PPP1R14A is also involved in long-term synaptic depression in Purkinje neurons .
PPP1R14A has been implicated in various diseases and conditions. It is up-regulated in some cancer cells, leading to hyperphosphorylation of tumor suppressor merlin/NF2 . In prostate cancer, PPP1R14A expression is associated with increased cell proliferation and certain genetic risk factors . Additionally, PPP1R14A has been linked to conditions such as Israeli Tick Typhus and Compensatory Emphysema .
Research on PPP1R14A continues to uncover its roles in cellular processes and disease mechanisms. The protein’s ability to regulate smooth muscle contraction and its involvement in cancer make it a potential target for therapeutic interventions. Antibodies against PPP1R14A, such as mouse anti-human PPP1R14A, are used in research to study its expression and function in various tissues .