PDE6A Antibody

Shipped with Ice Packs
In Stock

Description

Characterization of PDE6A Antibodies

PDE6A antibodies vary in specificity, reactivity, and applications. Key products include:

Catalog NumberHost/IsotypeReactivityApplicationsObserved MWImmunogen
67832-1-Ig Rabbit/IgGHuman, Mouse, RatWB100–110 kDaPDE6A fusion protein
21200-1-AP Rabbit/IgGHuman, Mouse, RatWB, IHC, IF, ELISA100–110 kDaPDE6A fusion protein
A07800-1 Rabbit/IgGHuman, Mouse, RatWB, IHC, Flow Cytometry100 kDaPDE6A fusion protein
ABIN7168417 Rabbit/IgGHuman, MouseWB, IHC, ELISA100 kDaRecombinant PDE6A (AA 656–860)

Key Features:

  • Molecular Weight: Observed at ~100 kDa, aligning with PDE6A’s calculated size (860 aa) .

  • Cross-Reactivity: Most antibodies target human, mouse, and rat PDE6A, with some showing broader reactivity (e.g., zebrafish, dog) .

  • Conjugation: Typically unconjugated, enabling flexibility in downstream assays .

Western Blot (WB)

PDE6A antibodies are widely used to quantify protein expression in retinal lysates. For example:

  • Protocol:

    1. Resolve 50 μg retinal lysates on 5–20% SDS-PAGE.

    2. Transfer to nitrocellulose and block with 5% non-fat milk.

    3. Incubate with primary antibody (0.25–1 μg/mL) overnight.

    4. Detect via HRP-conjugated secondary antibodies and ECL .

Example: PDE6A antibodies detect a ~100 kDa band in rod-enriched samples, validating their specificity .

Immunohistochemistry (IHC)

Used to localize PDE6A in retinal sections:

  • Protocol:

    1. Fix tissues in 4% PFA, section, and block with 10% goat serum.

    2. Incubate with primary antibody (1 μg/mL) overnight.

    3. Visualize using SABC-DAB or fluorescent secondary antibodies .

Findings:

  • Retinal Localization: Strong staining in rod outer segments (ROS) of wild-type retina, absent in Pde6a-mutant models .

  • Disease Models: Detects PDE6A mislocalization in Pde6a Q70X zebrafish, correlating with ROS shrinkage .

Immunofluorescence (IF)

Identifies subcellular PDE6A distribution in cultured cells:

  • Protocol:

    1. Fix cells with 4% PFA, block with 10% goat serum.

    2. Incubate with primary antibody (4 μg/mL) overnight.

    3. Stain with DyLight®488-conjugated secondary antibodies and counterstain with DAPI .

Example: PDE6A antibodies reveal cytoplasmic localization in CACO-2 cells, useful for studying protein trafficking .

Role in Retinitis Pigmentosa (RP)

PDE6A mutations cause autosomal recessive RP, characterized by photoreceptor degeneration. Key studies:

ModelFindingsAntibody Use
Pde6a Zebrafish ROS shrinkage and visual impairment correlate with PDE6A loss.IHC to detect PDE6A absence in mutants.
Pde6a-mutant Mice PARP inhibition delays photoreceptor death in early-stage RP models.WB to assess PDE6A expression.
Dog Models AAV-mediated Pde6a gene therapy restores PDE6A in rod outer segments.IHC to confirm viral vector efficacy.

Mechanistic Insights:

  • Photoreceptor Death: PDE6A deficiency disrupts cGMP hydrolysis, leading to calcium overload and apoptosis .

  • Therapeutic Potential: Gene therapy in dogs reduces rhodopsin mislocalization and preserves photoreceptors .

Gene Therapy Trials

AAV-delivered Pde6a restores PDE6A expression in rod outer segments, as shown in canine models :

ParameterOutcome
PDE6A ExpressionSustained for ≥3 months post-injection.
Functional RescuePartial recovery of ERG responses.
SafetyDose-dependent toxicity observed; optimized vectors needed for humans.

Diagnostic and Prognostic Use

PDE6A antibodies may aid in diagnosing RP subtypes and monitoring disease progression:

  • Biomarker Potential: Detects PDE6A loss in retinal biopsies or fluid samples.

  • Limitations: Cross-reactivity with non-rod tissues (e.g., ovarian follicles) requires careful validation .

Product Specs

Buffer
Phosphate buffered saline (PBS) with 0.02% sodium azide, 50% glycerol, pH 7.3.
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery time may vary depending on the purchase method and location. Please consult your local distributor for specific delivery details.
Synonyms
5''-cyclic phosphodiesterase subunit alpha antibody; CGPR A antibody; GMP PDE alpha antibody; GMP-PDE alpha antibody; PDE 6 alpha antibody; PDE 6A antibody; PDE V B1 antibody; PDE V-B1 antibody; PDE6A antibody; PDE6A_HUMAN antibody; PDEA antibody; Phosphodiesterase 6 alpha antibody; Phosphodiesterase 6A alpha subunit antibody; Phosphodiesterase 6A antibody; Phosphodiesterase 6A cGMP specific rod alpha antibody; Retinal Rod Photoreceptor cGMP Phosphodiesterase alpha antibody; Rod cGMP specific 3' 5' cyclic phosphodiesterase alpha subunit antibody; Rod cGMP-specific 3'' antibody
Target Names
PDE6A
Uniprot No.

Target Background

Function
This protein is involved in the transmission and amplification of visual signals.
Gene References Into Functions
  1. This study provides the first phenotypic description of autosomal recessive retinitis pigmentosa (arRP) caused by homozygous IVS6+1G>A mutations in PDE6A. These mutations appear to be associated with severe RP, leading to early extinction of rod responses and reduced macular function. PMID: 27820873
  2. This report describes two splice acceptor site variations in PDE6A identified in consanguineous Pakistani families exhibiting characteristic symptoms of autosomal recessive retinitis pigmentosa. PMID: 26321862
  3. This research investigates the amino acid residues responsible for the selectivity of tadalafil binding to two closely related phosphodiesterases, PDE5 and PDE6. PMID: 23033484
  4. The p.Val685Met mutation in PDE6A is identified as a causative factor for retinal degeneration in humans. PMID: 21039428
  5. Rod phosphodiesterase-6 PDE6A and PDE6B subunits demonstrate enzymatic equivalence. PMID: 20940301
  6. Full transcriptional activity of the PDE6A gene requires the combined action of both Nrl and Crx transcription factors. PMID: 15001570
  7. This study identified homozygous single base pair changes (c.889C->T), single base pair insertions (c.2218-2219insT), and a single base pair substitution in the splice acceptor site (IVS10-2A->G) in three distinct families. PMID: 17110911

Show More

Hide All

Database Links

HGNC: 8785

OMIM: 180071

KEGG: hsa:5145

STRING: 9606.ENSP00000255266

UniGene: Hs.567314

Involvement In Disease
Retinitis pigmentosa 43 (RP43)
Protein Families
Cyclic nucleotide phosphodiesterase family
Subcellular Location
Cell membrane; Lipid-anchor; Cytoplasmic side.

Q&A

What is PDE6A and what biological role does it play?

PDE6A (phosphodiesterase 6A, cGMP-specific, rod, alpha) is a crucial protein component of the phototransduction cascade in retinal rod cells. It belongs to the cyclic nucleotide phosphodiesterase family and is also known by alternative names including GMP-PDE alpha, PDE V-B1, and PDEA. PDE6A functions primarily in rod photoreceptors, where it hydrolyzes cGMP to 5'-GMP, an essential step in the visual signal transduction pathway. The protein has a calculated molecular weight of approximately 100 kDa (860 amino acids) and typically appears as a 100-110 kDa band on Western blots . Loss of function or mutations in PDE6A are associated with retinal degeneration and retinitis pigmentosa, making it an important target for both basic vision research and therapeutic development .

What applications can PDE6A antibodies be used for in research?

PDE6A antibodies have been validated for multiple experimental applications essential to vision research and neuroscience. The primary applications include Western Blot (WB), Immunohistochemistry (IHC), Immunofluorescence (IF), and ELISA. According to published literature, these antibodies demonstrate reactivity with human, mouse, and rat samples, making them versatile tools for comparative studies across species . Specifically, PDE6A antibodies have been used to monitor protein expression in retinal tissue, assess photoreceptor preservation in degenerative models, and evaluate the efficacy of gene editing approaches targeting PDE6A mutations . The antibody applications are supported by at least 6 publications for WB and 1 publication each for IHC and IF techniques, confirming their reliability for these methodologies .

What tissue samples are most appropriate for PDE6A antibody testing?

The most appropriate tissue samples for PDE6A antibody testing are primarily retinal tissues, where PDE6A is naturally expressed at high levels. According to validation data, positive Western Blot detection has been confirmed in mouse and rat eye tissue as well as human brain tissue . For immunohistochemistry applications, positive detection has been observed in mouse eye tissue, human colon cancer tissue, and human ovary cancer tissue . Immunofluorescence applications have been specifically validated in retinal tissue, which represents the gold standard for PDE6A detection . When examining genetically modified models or disease states affecting PDE6A expression, appropriate controls should include wild-type retinal tissue to establish baseline expression levels .

What are the recommended dilutions for different experimental techniques?

Optimal dilution ratios vary by application technique and should be empirically determined for each experimental system. The following table provides recommended dilution ranges based on validated protocols:

ApplicationRecommended Dilution Range
Western Blot (WB)1:1000-1:6000
Immunohistochemistry (IHC)1:250-1:1000
Immunofluorescence (IF)1:50-1:200

It is strongly recommended that researchers titrate the antibody in their specific testing system to achieve optimal results, as sample preparation methods and detection systems can significantly influence optimal concentration . Additionally, antigen retrieval conditions can affect antibody performance in IHC applications, with suggested protocols including TE buffer (pH 9.0) or alternatively citrate buffer (pH 6.0) .

How should researchers optimize antigen retrieval for PDE6A immunohistochemistry?

Optimizing antigen retrieval is critical for successful PDE6A immunodetection in fixed tissues. The recommended primary approach utilizes TE buffer at pH 9.0, which has been validated to effectively unmask PDE6A epitopes in paraffin-embedded sections . If suboptimal results are obtained, an alternative approach using citrate buffer at pH 6.0 may prove effective for certain tissue preparations . The antigen retrieval protocol should be optimized based on fixation method, tissue type, and section thickness. For retinal tissues, which represent the primary site of PDE6A expression, special care should be taken during processing as these delicate structures can be easily damaged. When working with archived tissues or those with extended fixation times, increasing the antigen retrieval duration may improve signal intensity. Researchers should validate their antigen retrieval protocol by including positive control tissues (such as wild-type retina) and negative controls (either tissues known not to express PDE6A or primary antibody omission) .

What approaches can be used to validate PDE6A antibody specificity?

Validating antibody specificity is essential for ensuring reliable experimental results. For PDE6A antibodies, multiple complementary approaches should be employed:

  • Genetic validation: Compare staining patterns between wild-type tissues and those from PDE6A knockout or mutant models (such as Pde6a-D670G mice). Complete absence or significant reduction of signal in knockout/mutant samples provides strong evidence of specificity .

  • Peptide competition assays: Pre-incubate the antibody with excess PDE6A immunizing peptide before application to tissues. Specific antibodies will show dramatically reduced or eliminated staining.

  • Multiple antibody validation: Compare staining patterns using different antibodies targeting distinct epitopes of PDE6A. Concordant results strengthen confidence in specificity.

  • Molecular weight confirmation: In Western blot applications, confirm that the detected band appears at the expected molecular weight of 100-110 kDa .

  • Cross-reactivity assessment: Test the antibody against related phosphodiesterase family members (especially PDE6B and PDE6G) to ensure signal specificity.

  • Correlation with mRNA expression: Compare protein detection patterns with PDE6A mRNA expression data from in situ hybridization or RNA-seq studies.

How can researchers effectively monitor PDE6A expression in gene editing experiments?

Monitoring PDE6A expression in gene editing experiments requires a multifaceted approach combining genomic, protein, and functional analyses:

  • Genomic verification: Confirm successful editing at the DNA level using deep sequencing to quantify correction efficiency of the target mutation. For example, in Pde6a-D670G mutation correction studies, researchers achieved 9.4 ± 5.5% target mutation correction using prime editing approaches .

  • Protein expression analysis: Employ Western blotting with PDE6A antibodies (recommended dilution 1:1000-1:6000) to quantify protein restoration. Compare band intensity to both untreated mutant samples and wild-type controls to determine the degree of protein expression rescue .

  • Immunofluorescence localization: Use IF (recommended dilution 1:50-1:200) to assess proper subcellular localization of the restored protein, particularly focusing on rod outer segments where PDE6A naturally functions .

  • Co-localization studies: Perform dual immunolabeling with PDE6A antibodies and markers of rod photoreceptors (e.g., Rhodopsin, Recoverin) to confirm cell-specific restoration .

  • Morphological assessment: Measure structural parameters such as outer nuclear layer thickness and rod outer segment length to correlate protein restoration with photoreceptor preservation .

  • Temporal analysis: Monitor expression at multiple time points to assess the stability and durability of the gene editing effect.

What are common causes of weak or absent PDE6A signal in Western blots?

When encountering weak or absent PDE6A signal in Western blot applications, researchers should systematically troubleshoot:

  • Sample preparation issues: PDE6A is predominantly expressed in photoreceptors, which can be easily damaged during tissue extraction. Ensure tissues are rapidly processed and maintained at appropriate temperatures to prevent protein degradation.

  • Protein extraction efficiency: PDE6A is associated with membranes in rod outer segments, requiring efficient extraction methods. Consider using specialized extraction buffers containing appropriate detergents (e.g., Triton X-100 or CHAPS) to solubilize membrane-associated proteins.

  • Antibody dilution: If using dilutions at the higher end of the recommended range (closer to 1:6000), consider using a more concentrated antibody preparation (e.g., 1:1000) .

  • Detection system sensitivity: Ensure your secondary antibody and detection reagents are appropriate for potentially low-abundance targets. Enhanced chemiluminescence (ECL) systems with increased sensitivity may be required.

  • Transfer efficiency: PDE6A is a relatively large protein (100-110 kDa), which may require optimized transfer conditions. Extended transfer times or lower methanol concentrations in transfer buffers may improve results for larger proteins.

  • Blocking optimization: Excessive blocking can mask epitopes. Test different blocking reagents (BSA vs. non-fat dry milk) and concentrations.

  • Genetic factors: In studies using PDE6A mutant models, protein levels may be intrinsically low due to nonsense-mediated decay or protein instability. Include wild-type samples as positive controls .

How can researchers minimize background in PDE6A immunofluorescence experiments?

Reducing background in PDE6A immunofluorescence applications is critical for accurate interpretation of results:

  • Optimize antibody concentration: Begin with the middle of the recommended dilution range (approximately 1:100) and adjust based on signal-to-noise ratio .

  • Blocking optimization: Extend blocking time (1-2 hours at room temperature or overnight at 4°C) using a blocking solution containing both serum (5-10%) matched to the secondary antibody host species and BSA (1-3%).

  • Washing stringency: Implement additional and extended washing steps with buffers containing 0.1-0.3% Triton X-100 or Tween-20 to remove unbound antibody.

  • Secondary antibody controls: Always include controls omitting primary antibody to identify non-specific binding of secondary antibodies.

  • Autofluorescence reduction: Treat retinal sections with sodium borohydride (1mg/ml in PBS for 2 minutes) before antibody incubation to reduce tissue autofluorescence, particularly important for aged or fixed retinal tissues.

  • Cross-adsorbed secondary antibodies: Use highly cross-adsorbed secondary antibodies to minimize species cross-reactivity.

  • Antigen retrieval optimization: Fine-tune antigen retrieval conditions, as excessive retrieval can increase background while insufficient retrieval may reduce specific signal .

  • Photobleaching prevention: Minimize exposure to light during all steps and mount slides with anti-fade mounting media containing appropriate photo-protectants.

What controls are essential for validating PDE6A antibody experiments?

Rigorous experimental controls are essential for validating PDE6A antibody results:

  • Positive tissue controls: Include wild-type retinal tissue known to express PDE6A as a positive control in all experiments .

  • Negative tissue controls: Include tissues known not to express PDE6A (e.g., liver) to confirm antibody specificity.

  • Genetic controls: When available, include samples from PDE6A knockout animals or cellular models to confirm signal specificity.

  • Primary antibody omission: Process sections without primary antibody but with all other reagents to identify non-specific secondary antibody binding.

  • Isotype controls: Use matched concentration of irrelevant antibodies of the same isotype (Rabbit IgG) to identify potential Fc-receptor mediated binding .

  • Absorption controls: Pre-incubate primary antibody with immunizing peptide to demonstrate binding specificity.

  • Cross-validation controls: Compare results using alternative detection methods (e.g., in situ hybridization for mRNA) to confirm expression patterns.

  • Dilution series: Test a range of antibody dilutions to identify optimal signal-to-noise ratios and demonstrate signal specificity.

How should researchers quantify and report PDE6A expression changes in retinal degeneration models?

Quantification and reporting of PDE6A expression changes requires standardized approaches:

How do PDE6A expression levels correlate with photoreceptor survival in gene therapy experiments?

Research has established critical correlations between PDE6A expression and photoreceptor survival:

In gene therapy experiments targeting PDE6A mutations, such as the Pde6a-D670G mouse model, successful correction of the mutation and restoration of PDE6A expression strongly correlates with photoreceptor preservation. Key correlative metrics include:

  • Protein restoration threshold: Even partial restoration of PDE6A expression (significantly below wild-type levels) can lead to measurable photoreceptor preservation. In AAV-PE treated mice, PDE6A protein was restored to detectable but lower-than-wild-type levels while still providing significant therapeutic benefit .

  • Structural preservation metrics: PDE6A restoration correlates with:

    • Outer nuclear layer thickness (15.9 ± 3.8 μm in treated versus minimal in untreated retinas)

    • Rod outer segment length (4.9 ± 1.1 μm in treated, approximately 40% of wild-type length, versus nearly absent in untreated retinas)

  • Molecular marker correlation: PDE6A expression restoration correlates with preserved expression of other photoreceptor-specific proteins including Recoverin (rod marker), Cone arrestin and M-opsin (cone markers), and Rhodopsin .

  • Editing efficiency correlation: In gene editing approaches, a target editing efficiency of 9.4 ± 5.5% was sufficient to achieve significant photoreceptor preservation, suggesting a relatively low therapeutic threshold .

  • Bystander effects: The precision of gene editing approaches affects outcomes. Prime editing approaches that eliminate bystander editing showed superior preservation of photoreceptor morphology compared to base editing approaches with bystander effects .

What are the considerations for interpreting off-target effects when using PDE6A antibodies in gene editing studies?

Accurate interpretation of off-target effects requires careful experimental design:

  • Antibody cross-reactivity: Confirm PDE6A antibody specificity to ensure observed signals represent genuine PDE6A expression rather than cross-reactivity with related phosphodiesterases or other proteins .

  • Genomic off-target analysis: When evaluating gene editing approaches, sequence the most likely predicted off-target sites based on computational algorithms. In PDE6A gene editing studies, eight predicted potential off-target sites showed no obvious off-target editing above background levels in treated mice .

  • Tissue-specific off-target assessment: In addition to analyzing the target tissue (retina), examine non-targeted organs such as brain and liver for unintended editing effects. Studies have shown no significant off-target editing in these tissues following AAV-delivered prime editing treatment targeting PDE6A .

  • Control for detection sensitivity: Establish the detection threshold of your sequencing approach and clearly report this as the lower limit for potential off-target detection.

  • Distinguish technical artifacts: Separate true off-target effects from technical artifacts in sequencing or antibody binding by including appropriate controls (untreated samples processed identically).

  • Functional relevance assessment: For any detected off-target effects, assess whether they occur in coding regions and whether they could result in functional consequences.

  • Temporal analysis: Monitor potential off-target effects over extended time periods to detect any progressive accumulation of unintended modifications.

How do different gene editing approaches compare in correcting PDE6A mutations?

Recent research has evaluated multiple gene editing strategies for treating PDE6A-associated retinal degeneration:

What methodological innovations are emerging for studying PDE6A function in retinal models?

Innovative methodological approaches are advancing PDE6A research:

  • Cell Line Models: The development of engineered cell lines carrying specific PDE6A mutations (such as the N2a cell model with the Pde6a-D670G mutation) provides valuable platforms for initial screening of gene editing approaches before moving to more complex animal models .

  • In Vivo Electroporation: Techniques for electroporation of plasmids expressing genome editing systems into neonatal retinas allow for rapid preliminary validation of therapeutic approaches without the complexity of viral vector production .

  • Dual AAV Delivery Systems: The development of dual AAV systems capable of delivering larger gene editing constructs has expanded the toolkit for in vivo PDE6A research, enabling delivery of complex prime editing machinery that exceeds the packaging capacity of single AAV vectors .

  • Engineered Deaminase Variants: The development of engineered deaminases (such as AID-N51A) with improved precision and reduced bystander editing has enhanced the potential for precise correction of PDE6A mutations .

  • Multimodal Assessment: Integration of genomic, protein expression, and structural analyses provides comprehensive evaluation of therapeutic interventions:

    • Deep sequencing for precise quantification of on-target and off-target editing

    • Immunofluorescence for spatial analysis of protein restoration

    • Histological assessment for structural preservation

    • Functional readouts of vision restoration

  • Comparative Analysis Frameworks: Standardized approaches for side-by-side comparison of different therapeutic strategies (base editing vs. prime editing vs. gene supplementation) enable objective evaluation of relative efficacy and safety profiles .

What are the key considerations for translating PDE6A research to clinical applications?

Translating PDE6A research to clinical applications requires addressing several critical considerations:

  • Mutation Specificity: PDE6A-associated retinitis pigmentosa can result from various mutations, each potentially requiring customized gene editing approaches with specific guide RNAs, adding complexity to clinical translation .

  • Delivery Optimization:

    • Dual AAV-PE systems face challenges including delivery complexity, potential instability, higher dose requirements, and increased immunogenicity compared to single-vector gene therapy approaches .

    • Optimization of viral serotypes, injection protocols, and vector design is essential for maximizing transduction efficiency in human retinas.

  • Therapeutic Threshold Determination:

    • Identifying the minimum level of PDE6A restoration required for clinical benefit is crucial for setting efficacy standards.

    • Animal models suggest even partial restoration (less than wild-type levels) may provide significant therapeutic benefit .

  • Timing Considerations:

    • Intervention timing is critical, as retinal degeneration progresses with age.

    • Establishing the therapeutic window for intervention requires careful assessment of disease progression in human patients versus animal models.

  • Safety Assessment:

    • Comprehensive evaluation of off-target effects using unbiased genome-wide approaches rather than just predicted sites.

    • Long-term monitoring for delayed adverse effects, particularly for permanent genome modifications.

  • Comparative Effectiveness:

    • Direct comparison with alternative approaches (gene supplementation, cell therapy) is needed to determine the optimal therapeutic strategy.

    • Cost-effectiveness and scalability considerations will impact clinical adoption.

  • Regulatory Pathways:

    • Gene editing approaches face distinct regulatory considerations compared to conventional gene therapy.

    • Developing standardized analytical methods for characterizing editing precision and efficiency to meet regulatory requirements.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.