PDE1A antibodies (e.g., Proteintech #12442-2-AP, Abcam #ab232801) are polyclonal rabbit IgG antibodies targeting human, mouse, and rat PDE1A. Key specifications include:
| Property | Details |
|---|---|
| Immunogen | Recombinant PDE1A fusion protein (aa 1–350 in human) |
| Molecular Weight | ~61 kDa (observed) / 545 amino acids (calculated) |
| Reactivity | Human, mouse, rat (validated); predicted cross-reactivity with pig |
| Applications | WB, IHC, IP, ELISA |
| Storage | -20°C in PBS with 0.02% sodium azide and 50% glycerol |
These antibodies recognize multiple PDE1A isoforms produced by alternative splicing, including the major 61–67 kDa forms expressed in brain, heart, and smooth muscle .
Cancer Metastasis: PDE1A overexpression in non-small cell lung cancer (NSCLC) correlates with poor prognosis and promotes metastasis by enhancing epithelial-mesenchymal transition (EMT). Antibodies confirmed PDE1A's role in STAT3/YTHDF2 signaling pathways .
Fibrosis: In aortic adventitial fibroblasts, PDE1A inhibition blocked TGF-β1-induced α-smooth muscle actin expression, a hallmark of myofibroblast formation .
Polycystic Kidney Disease: PDE1A knockout mice showed mild renal cysts and urine-concentrating defects, linked to dysregulated cAMP signaling .
Western Blot: Detects PDE1A at 61 kDa in human brain, heart, and lung tissues .
Immunohistochemistry: Highlights PDE1A localization in NSCLC tissues and normal Purkinje neurons .
Knockout Validation: Antibodies confirmed reduced PDE1A protein levels in Pde1a Del15/InsA mice, supporting gene-editing efficacy .
PDE1A hydrolyzes cAMP and cGMP, modulating downstream effectors:
Vinpocetine: A PDE1 inhibitor reduced NSCLC cell migration and EMT progression .
8-MMX/Trifluoperazine: PDE1A inhibitors tested in pulmonary hypertension and polycystic kidney disease models .
NSCLC: Targeting PDE1A-YTHDF2 interactions suppressed metastasis in mouse xenografts .
Cardiovascular: PDE1A knockout mice exhibited lower aortic blood pressure and improved cardiac ejection fraction .
PDE1A is a calcium/calmodulin-activated phosphodiesterase enzyme that regulates cyclic nucleotide degradation, specifically cAMP and cGMP. As a member of the PDE1 family, PDE1A serves as a critical intersection point between calcium signaling and cyclic nucleotide pathways. The enzyme is differentially expressed across various cell types including vascular smooth muscle cells, renal tubular epithelial cells, podocytes, and mesangial cells . PDE1A functions as a key regulator in multiple cellular processes including migration, adhesion, and cellular motility, which are critical parameters in cancer cell dissemination . Additionally, recent studies have shown PDE1A's involvement in tumor angiogenesis and metastatic potential, particularly in non-small cell lung cancer (NSCLC) .
When selecting a PDE1A antibody for research applications, consider the following critical parameters:
Antibody specificity: Select antibodies validated to distinguish between PDE1A and other PDE1 isoforms (PDE1B and PDE1C) as these have distinct expression patterns and functions.
Application compatibility: Ensure the antibody has been validated for your intended application (immunohistochemistry, Western blotting, immunofluorescence, etc.).
Epitope location: Consider whether the antibody recognizes an epitope that might be masked by protein-protein interactions, such as the YTHDF2 interaction demonstrated in recent studies .
Cross-reactivity profile: Verify the antibody has been tested against potential cross-reactive proteins, particularly other phosphodiesterases.
Species reactivity: Confirm compatibility with your experimental model organism (human, mouse, rat, etc.).
For immunofluorescence or proximity ligation assay applications, dilutions around 1:500 have been effective in published research protocols .
Validating PDE1A antibody specificity requires a multi-faceted approach:
Positive and negative control tissues/cells: Use tissues or cell lines with known high PDE1A expression (such as NSCLC cell lines) as positive controls and compare with low-expressing counterparts (such as normal human lung fibroblasts) .
siRNA knockdown validation: Transfect cells with PDE1A-specific siRNA and confirm reduction in antibody signal compared to control siRNA.
Overexpression validation: Compare antibody signal in cells transfected with PDE1A expression vector versus empty vector controls .
Western blot analysis: Confirm a single band at the expected molecular weight (~61 kDa for human PDE1A).
Peptide competition assay: Pre-incubate the antibody with a blocking peptide containing the target epitope and confirm signal elimination.
Multi-antibody validation: Compare staining patterns of different antibodies targeting distinct PDE1A epitopes.
Measuring PDE1A enzyme activity requires techniques that go beyond detecting protein presence:
Cyclic nucleotide hydrolysis assay: Quantify cAMP/cGMP degradation rates in tissue lysates with and without calcium/calmodulin stimulation. The calcium/calmodulin-dependent component represents PDE1 activity.
Selective inhibitor approach: Measure total PDE activity, then repeat with selective PDE1 inhibitors (such as vinpocetine or ITI-214) . The difference represents PDE1 contribution.
Immunoprecipitation-coupled activity assay: Immunoprecipitate PDE1A using validated antibodies, then measure PDE activity in the precipitate.
Real-time cGMP monitoring: Use highly sensitive fluorescent indicators to measure dynamic changes in cGMP levels in living cells, as demonstrated in cardiomyocyte studies .
Differential isoform inhibition: Compare activity profiles after selective knockdown of PDE1A versus PDE1B or PDE1C to distinguish isoform-specific contributions.
For accurate quantification, always include appropriate controls including heat-inactivated samples and samples with EGTA to chelate calcium and inhibit PDE1A activation.
When encountering contradictory data regarding PDE1A's signaling role, implement these resolution strategies:
Cell-type specific analysis: PDE1A functions may vary significantly between cell types. For example, in cardiomyocytes, PDE1 inhibition affects cGMP signaling , while in NSCLC cells, it influences the STAT3 pathway .
Isoform-specific manipulation: Use not only pharmacological inhibitors but also genetic approaches (siRNA, CRISPR) targeting specific PDE1 isoforms to distinguish their individual contributions.
Pathway intersection mapping: Since PDE1A sits at the intersection of calcium and cyclic nucleotide signaling, use calcium chelators and pathway-specific inhibitors to delineate which pathway predominates in your system.
Subcellular localization studies: Determine if PDE1A has different functions in distinct subcellular compartments using fractionation techniques combined with immunofluorescence.
Temporal dynamics analysis: Implement time-course experiments to resolve seemingly contradictory effects that may occur at different time points after stimulation.
Protein complex identification: Use proximity ligation assays or co-immunoprecipitation to identify specific protein interactions that may explain context-dependent functions .
Distinguishing between PDE1 isoforms requires a combinatorial approach:
Isoform-specific knockdown: Use targeted siRNA for each isoform and compare phenotypic consequences. Research has shown that siPDE1A exerts stronger suppression effects on migration of NCI-H1299 cells than siPDE1B or siPDE1C .
Expression profiling: Quantify relative expression levels of each isoform in your system using qRT-PCR and Western blotting with isoform-specific antibodies.
Rescue experiments: After knockdown of all PDE1 activity, selectively re-express individual isoforms to identify their specific contributions.
Subcellular distribution analysis: Map the distinct subcellular localization patterns of each isoform using isoform-specific antibodies in immunofluorescence studies.
Calcium sensitivity profiling: The three isoforms have different sensitivities to calcium/calmodulin activation, which can be exploited experimentally.
Substrate preference analysis: PDE1 isoforms show differential preferences for cAMP versus cGMP, which can be measured in enzymatic assays.
| PDE1 Isoform | Migration Effect in NSCLC | EMT Progression Impact | Relative Expression in Invasive NSCLC |
|---|---|---|---|
| PDE1A | Strong suppression when knocked down | Strong suppression | Significantly higher in highly invasive cells |
| PDE1B | Moderate suppression | Moderate suppression | Lower differential expression |
| PDE1C | Mild suppression | Mild suppression | Lower differential expression |
Table adapted from functional studies comparing PDE1 isoforms in NSCLC cells
The choice of fixation and antigen retrieval methods significantly impacts PDE1A antibody performance:
Fixation protocol:
For most tissues including lung and kidney, 4% paraformaldehyde (PFA) for 24 hours provides optimal preservation of PDE1A epitopes.
For vascular tissues, a shorter fixation (12-16 hours) may yield better results.
Fresh-frozen sections may be preferable for certain epitopes sensitive to fixation.
Antigen retrieval methods:
Heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) at 95-98°C for 20 minutes generally works well for PDE1A.
For formalin-fixed tissues, consider Tris-EDTA buffer (pH 9.0) if citrate buffer yields weak signals.
Enzymatic retrieval (using proteinase K) is typically not recommended as it may destroy PDE1A epitopes.
Tissue-specific considerations:
Lung tissue: Extended rinsing steps help reduce background from endogenous peroxidases.
Brain tissue: Lighter fixation (2% PFA) may improve antibody penetration.
Kidney tissue: Requires thorough blocking of endogenous biotin to prevent non-specific staining.
The Human Protein Atlas protocols have successfully visualized PDE1A in normal lung and lung cancer tissues .
Optimizing PLA for PDE1A interaction studies requires careful consideration of several parameters:
Antibody selection: Use antibodies from different species for PDE1A and its potential interaction partners. For example, rabbit anti-PDE1A combined with mouse antibodies against potential partners like YTHDF2 .
Antibody validation: Validate both antibodies independently by immunofluorescence before attempting PLA.
Dilution optimization: Start with 1:500 dilutions for both antibodies as used in previous PLA studies for cGMP-interacting proteins , then optimize as needed.
Controls:
Negative controls: Omit one primary antibody
Biological controls: Use cells with PDE1A knockdown
Positive controls: Include known protein interaction pairs
Signal quantification: Use appropriate software to quantify PLA puncta, applying statistical methods such as Kruskal-Wallis for comparing PLA signals between different experimental conditions .
Protocol refinements:
Extend blocking time to 1-2 hours to reduce non-specific interactions
Optimize incubation temperature (4°C overnight often yields better results than room temperature)
Consider using amplification steps for low-abundance interactions
Detecting phosphorylated PDE1A requires specialized approaches:
Phospho-specific antibodies: Utilize antibodies specifically targeting known phosphorylation sites of PDE1A. If commercial phospho-specific antibodies are unavailable, consider custom antibody development.
Phosphatase inhibitors: Include comprehensive phosphatase inhibitor cocktails in all extraction buffers (containing sodium fluoride, sodium orthovanadate, β-glycerophosphate, and phosphatase inhibitor cocktails).
Enrichment strategies:
Phosphoprotein enrichment using TiO₂ or IMAC columns prior to immunoblotting
Immunoprecipitation of total PDE1A followed by phospho-specific detection
Use of Phos-tag™ SDS-PAGE for mobility shift detection of phosphorylated forms
Mass spectrometry approaches:
Targeted LC-MS/MS after immunoprecipitation
Parallel reaction monitoring (PRM) for specific phosphopeptides
SILAC or TMT labeling for quantitative phosphoproteomics
Validation methods:
Use phosphatase treatment controls to confirm phospho-specific signals
Apply kinase inhibitors relevant to known PDE1A kinases
Implement site-directed mutagenesis of phosphorylation sites
Based on recent findings in NSCLC , a comprehensive experimental design should include:
Expression correlation analysis:
Compare PDE1A expression between primary tumors and matched metastatic lesions
Analyze correlation between PDE1A expression and metastasis-free survival in patient cohorts
Examine PDE1A expression in cell lines with differential metastatic potential
In vitro functional assays:
Wound healing assays comparing control vs. PDE1A-knockdown/overexpressing cells
Transwell migration and invasion assays
3D spheroid invasion assays in appropriate matrix
EMT marker assessment (E-cadherin, N-cadherin) by immunoblotting and immunofluorescence
Mechanistic investigations:
Assess PDE1A regulation of specific pathways implicated in metastasis (STAT3, IL-6/JAK)
Investigate protein-protein interactions (e.g., PDE1A-YTHDF2) using co-immunoprecipitation and PLA
Examine effects on mRNA stability of metastasis-related genes
In vivo metastasis models:
Tail vein injection metastasis model with PDE1A-modulated cells
Orthotopic implantation with spontaneous metastasis assessment
Analyze metastatic nodules in lungs or other organs
Pharmacological validation:
Test effects of selective PDE1 inhibitors like vinpocetine on metastatic potential
Compare with genetic manipulation to confirm target specificity
When studying PDE1A inhibition effects on cyclic nucleotide signaling, incorporate these essential controls:
Inhibitor specificity controls:
Pathway validation controls:
Include direct activators of adenylyl cyclase (forskolin) or guanylyl cyclase (NO donors)
Use cell-permeable cAMP/cGMP analogs to confirm downstream effects
Apply specific inhibitors of PKA (e.g., H-89) or PKG (e.g., KT5823) to distinguish pathway contributions
Temporal controls:
Perform detailed time-course experiments to capture both rapid and delayed responses
Implement washout experiments to confirm reversibility
Readout validation:
Cell type controls:
Compare effects in cells with high versus low endogenous PDE1A expression
Test in multiple cell types relevant to the biological question
These controls will help distinguish direct effects of PDE1A inhibition from off-target effects and determine pathway specificity.
Recent research has identified a novel PDE1A interaction with YTHDF2 that affects STAT3 signaling in NSCLC . To investigate this interaction effectively:
Interaction validation approaches:
Co-immunoprecipitation with antibodies against both PDE1A and YTHDF2
Proximity ligation assay to visualize interactions in situ
FRET or BiFC assays for dynamic interaction studies in living cells
Domain mapping using truncated constructs to identify interaction regions
Functional relationship studies:
Knockdown YTHDF2 and assess effects on PDE1A function and localization
Knockdown PDE1A and examine YTHDF2's RNA-binding activity
Perform rescue experiments with wild-type and mutant constructs
m6A-mediated regulation analysis:
Perform MeRIP-seq to identify m6A-modified transcripts affected by PDE1A-YTHDF2 interaction
RNA stability assays for candidate targets like SOCS2
Use m6A writer (METTL3/14) or eraser (FTO, ALKBH5) manipulations to modify the pathway
STAT3 pathway investigation:
Monitor STAT3 phosphorylation status with PDE1A/YTHDF2 manipulation
Assess nuclear translocation of STAT3 using fractionation and imaging
Measure STAT3 target gene expression
Test IL-6 stimulation with and without PDE1A/YTHDF2 modulation
Integrated multi-omics approach:
PDE1A antibodies can be valuable tools for patient stratification in precision oncology:
Tissue microarray analysis:
Develop immunohistochemistry scoring systems for PDE1A expression
Correlate expression with patient outcomes and treatment responses
Create cutoff values for high versus low expression groups
Multimarker panels:
Combine PDE1A staining with other markers in the pathway (YTHDF2, STAT3, SOCS2)
Develop integrated scoring systems that predict response to PDE1 inhibition
Validate in retrospective patient cohorts before prospective studies
Liquid biopsy applications:
Explore detection of PDE1A in circulating tumor cells
Investigate PDE1A in exosomes as potential biomarkers
Correlate with tissue expression and clinical outcomes
Functional testing:
Develop ex vivo drug sensitivity assays using patient-derived tumor samples
Correlate PDE1A expression with sensitivity to PDE1 inhibitors
PDE1A's role in cardiovascular physiology requires careful experimental design to separate direct from indirect effects:
Cell-type specific analyses:
Use cardiomyocyte-specific conditional PDE1A knockout models
Compare with vascular smooth muscle-specific knockouts
Analyze endothelial-specific effects separately
Ex vivo preparations:
Isolated heart preparations to evaluate direct cardiac effects
Isolated vessel myography to assess direct vascular effects
Compare responses in tissue from knockout versus wild-type animals
Pharmacological dissection:
Use selective PDE1 inhibitors with different pharmacokinetic profiles
Apply inhibitors before and after autonomic blockade
Compare acute versus chronic inhibition effects
Signaling pathway resolution:
Hemodynamic separations:
PDE1 inhibition has been shown to reduce blood pressure in both normotensive and spontaneously hypertensive rats, suggesting direct vascular effects .
Antibody-based techniques can provide valuable insights into PDE1A's role in kidney disease:
Cell-type mapping:
Perform high-resolution co-localization studies to map PDE1A expression in specific nephron segments
Use dual-immunostaining with nephron segment markers (podocin, AQP1, NKCC2, NCC)
Compare expression patterns in healthy versus diseased kidneys
Pathological correlations:
Quantify PDE1A expression in kidney biopsies from various nephropathies
Correlate with markers of kidney injury (KIM-1, NGAL)
Assess relationship with fibrosis markers and inflammatory infiltrates
Mechanistic studies:
Use phospho-specific antibodies to track PDE1A activation status
Perform PLA to identify disease-specific protein interaction networks
Combine with functional readouts of cAMP/cGMP signaling
Intervention monitoring:
Research has shown that PDE1 inhibition with selective inhibitors like LY1 can significantly improve renal function in diabetic kidney disease models, reducing albuminuria , suggesting antibody-based monitoring of PDE1A could help track disease progression and treatment response.