PDE8A Antibody

Shipped with Ice Packs
In Stock

Description

Key Applications and Dilutions

ApplicationDilution RangeValidated Tissues/Cells
Western Blot (WB)1:500 - 1:2,400Mouse testis/spleen, SH-SY5Y cells
Immunoprecipitation0.5-4.0 μg/1-3 mg lysateMouse testis
IHC1:20 - 1:200Human colon cancer (TE buffer pH 9.0 recommended)

Cardiac Function Regulation

PDE8A modulates cAMP pools in ventricular myocytes, influencing excitation-contraction coupling:

  • Calcium Dynamics: PDE8A knockout (PDE8A⁻/⁻) mice exhibit amplified Ca²⁺ transients and L-type Ca²⁺ currents during β-adrenergic stimulation, indicating PDE8A's role in buffering cAMP near Ca²⁺ handling proteins .

  • Mechanism: PDE8A deficiency increases cAMP levels post-βAR activation but does not alter global PKA activity, suggesting compartmentalized cAMP regulation .

Steroidogenesis in Leydig Cells

PDE8A and PDE8B jointly regulate basal testosterone production:

  • Subcellular Localization: PDE8A associates with mitochondria, while PDE8B is cytosolic .

  • Functional Synergy: Combined PDE8A/PDE8B knockout abolishes PF-04957325 (PDE8 inhibitor)-induced steroidogenesis, confirming their non-redundant roles .

ERK Signaling Modulation

PDE8A binds Raf-1 kinase to enhance ERK pathway activity:

  • Interaction: PDE8A associates with Raf-1 via residues 454–465, protecting it from inhibitory PKA phosphorylation .

  • Cellular Impact: Disrupting PDE8A-Raf-1 binding reduces EGF-induced ERK activation and cell migration, highlighting its role in growth signaling .

Target Protein Characteristics

Gene Symbol: PDE8A
UniProt ID: O60658
Isoforms: 5 splice variants (93 kDa, 88 kDa, 51 kDa, 66 kDa x2)
Functional Domains: Catalytic PDE domain, phosphorylation/glycosylation sites

Validation and Citations

  • Peer-Reviewed Studies: Cited in 5+ publications investigating mitochondrial cAMP dynamics, ovarian follicular cells, and Alzheimer’s disease .

  • Selectivity: Recognizes PDE8A without cross-reactivity to PDE8B or other PDE families in knockout validation .

Technical Considerations

  • Buffer: PBS with 0.02% sodium azide, 50% glycerol (pH 7.3)

  • Storage: Stable at -20°C for one year; avoids freeze-thaw cycles

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery times may vary depending on the purchase method and location. For precise delivery times, please consult your local distributors.
Synonyms
5''-cyclic phosphodiesterase 8A antibody; cAMP specific cyclic nucleotide phosphodiesterase 8A antibody; FLJ16150 antibody; High affinity cAMP specific and IBMX insensitive 3'5' cyclic phosphodiesterase 8A antibody; High affinity cAMP-specific and IBMX-insensitive 3'' antibody; HsT19550 antibody; PDE 8A antibody; PDE8A antibody; PDE8A_HUMAN antibody; Phosphodiesterase 8A antibody; Phosphodiesterase8A antibody; Weakly similar to 3'5' cyclic nucleotide phosphodiesterase antibody
Target Names
PDE8A
Uniprot No.

Target Background

Function
PDE8A is an enzyme that hydrolyzes the second messenger cAMP, a critical regulator of many physiological processes. It may play a role in maintaining basal cAMP levels and/or in cAMP regulation of germ cell development. Binding to RAF1 can reduce RAF1 'Ser-259' inhibitory phosphorylation and stimulate RAF1-dependent EGF-activated ERK-signaling. Moreover, PDE8A provides protection against cell death induced by hydrogen peroxide and staurosporine.
Gene References Into Functions
  1. Single nucleotide polymorphisms (SNPs) within the SCHIP1 and PDE8A genes were associated with facial size measurements in both genome-wide association studies (GWAS) and replication cohorts, surpassing a stringent genome-wide significance threshold adjusted for multiple testing of 34 correlated traits. Expression of both SCHIP1 and PDE8A was clearly demonstrated in the developing mouse face using whole-mount in situ hybridization and RNA-seq. PMID: 27560698
  2. PDE8A supports HIV-1 replication in macrophages. PMID: 25295610
  3. Phosphodiesterases play a novel and crucial role in modulating local cAMP concentrations in microdomains and signal resetting. PMID: 25229150
  4. PDE8A binds tightly to Raf-1, regulates Raf-1 phosphorylation at S259, and, in doing so, controls the cross-talk node where cAMP exerts an inhibitory effect on Raf-1 signaling, slowing down subsequent ERK phosphorylation and activation. PMID: 23509299
  5. PDE8 activity can be modulated by a kinase. PMID: 22673573
  6. Polymorphism in PDE8A affects HIV-1 replication in primary macrophages. PMID: 21920574
  7. Comparison of enzymatic characterization and gene organization of PDE8B and PDE8A. PMID: 12681444
  8. In vitro and in vivo experiments demonstrated that association with IkappaB significantly enhanced the enzyme activity of PDE8A1. PMID: 15596729
  9. Kinetic properties of the highly active PDE8A1 catalytic domain prepared from refolding and its crystal structures in the unliganded and 3-isobutyl-1-methylxanthine (IBMX) bound forms at 1.9 and 2.1 A resolutions. PMID: 18983167
  10. The findings exclude a significant role for PDE8A as a Polycystic Ovary Syndrome (PCOS) candidate gene and as a major determinant of androgen levels in women. PMID: 19482904

Show More

Hide All

Database Links

HGNC: 8793

OMIM: 602972

KEGG: hsa:5151

STRING: 9606.ENSP00000311453

UniGene: Hs.9333

Protein Families
Cyclic nucleotide phosphodiesterase family, PDE8 subfamily
Tissue Specificity
Expressed in most tissues except thymus and peripheral blood leukocytes. Highest levels in testis, ovary, small intestine and colon.

Q&A

What is PDE8A and why are antibodies against it important for research?

PDE8A (phosphodiesterase 8A) is a high-affinity cAMP-specific and IBMX-insensitive 3',5'-cyclic phosphodiesterase that belongs to the cyclic nucleotide phosphodiesterase family. It plays a critical role in the modulation of compartmentalized cAMP and PKA activity during β-adrenergic receptor activation . PDE8A hydrolyzes the second messenger cAMP, which regulates many important physiological processes. Unlike other PDE families, PDE8A is uniquely insensitive to the non-selective inhibitor IBMX, making it biochemically distinct .

PDE8A is expressed in most tissues except thymus and peripheral blood leukocytes, with highest expression in testis, ovary, small intestine, and colon . At the molecular level, PDE8A contains PAC (PAS-associated C-terminal) and PAS (PER-ARNT-SIM) domains that may contribute to its regulatory properties . Recent research has revealed that PDE8A directly interacts with Raf-1 kinase with remarkably high affinity (Kd <61 pM), establishing a novel mechanism for cross-talk between cAMP and MAP kinase signaling pathways .

Antibodies against PDE8A are essential for studying its tissue-specific expression patterns, subcellular localization, protein-protein interactions, and roles in compartmentalized signaling. They enable researchers to investigate how this enzyme contributes to specific physiological processes like cardiac excitation-contraction coupling and airway smooth muscle function .

What are the optimal applications for PDE8A antibodies?

Based on extensive validation studies, PDE8A antibodies have demonstrated utility across multiple experimental applications, though performance may vary by specific antibody clone and experimental conditions.

ApplicationValidated ApplicationsRecommended DilutionsNotes
Western Blot (WB)Widely validated1:500-1:2400Detects bands at 93 kDa and 88 kDa
Immunoprecipitation (IP)Strong performance0.5-4.0 μg for 1.0-3.0 mg lysateEffective for studying protein interactions
Immunohistochemistry (IHC)Validated1:20-1:200May require antigen retrieval with TE buffer pH 9.0
Immunofluorescence (IF)Validated1:250Used for subcellular localization studies
Immunocytochemistry (ICC)Validated1:250Effective for cultured cell studies
ELISAValidated1:1000-1:10,000High sensitivity with proper dilution

For optimal results, each application should be carefully optimized. For instance, in Western blotting, PDE8A antibodies have been successfully tested in diverse tissue and cell samples including mouse testis, mouse spleen, and SH-SY5Y cells . For immunohistochemistry, human colon cancer tissue has served as a reliable positive control .

How should researchers optimize experimental conditions for detecting PDE8A in Western blots?

Successful detection of PDE8A in Western blots requires attention to several critical parameters:

  • Sample preparation and loading:

    • Fresh tissue/cell lysates yield optimal results, with 15-50 μg total protein typically sufficient for cell lines like HeLa or 293T

    • Mouse testis and spleen tissues provide reliable positive controls

    • Include protease inhibitors in lysis buffers to prevent degradation

  • Gel electrophoresis and transfer:

    • Use 7-10% polyacrylamide gels for optimal separation around the 93 kDa range

    • Standard wet transfer protocols are effective for PDE8A

  • Blocking and antibody incubation:

    • Primary antibody dilutions typically range from 1:500-1:2400

    • Overnight incubation at 4°C generally yields optimal signal-to-noise ratio

    • Expected bands appear at 93 kDa and sometimes at 88 kDa, reflecting different isoforms

  • Visualization and analysis:

    • Standard chemiluminescence detection with 30-second exposure is generally sufficient

    • For challenging samples, consider signal enhancement systems

When interpreting bands, note that PDE8A knockout/knockdown samples serve as crucial negative controls to confirm specificity . In samples with low expression, an immunoprecipitation step prior to Western blotting can enrich the target protein, improving detection sensitivity .

How can researchers use PDE8A antibodies to investigate its interaction with Raf-1 kinase?

The PDE8A-Raf-1 interaction represents a novel mechanism for cross-talk between cAMP and MAPK signaling pathways. Several methodological approaches utilizing PDE8A antibodies can effectively investigate this interaction:

  • Co-immunoprecipitation studies:

    • Immunoprecipitate Raf-1 and probe for PDE8A, or vice versa

    • Both endogenous and epitope-tagged (Myc-Raf-1, Flag-PDE8A) proteins can be successfully co-immunoprecipitated

    • Include appropriate controls (non-specific IgG immunoprecipitation) to confirm specificity

    • The interaction appears independent of intracellular cAMP levels, persisting even after forskolin treatment

  • Direct binding assessment:

    • Surface plasmon resonance analysis has demonstrated extremely high-affinity binding (Kd <61 pM) between purified proteins

    • In vitro pull-down assays with GST-Raf-1 and MBP-PDE8A confirm direct interaction

  • Functional consequences assessment:

    • PDE8A regulates the inhibitory phosphorylation of Raf-1 at Ser-259

    • Compare phospho-Ser-259 levels in cells overexpressing wild-type versus dominant-negative PDE8A

    • Investigate downstream effects on ERK activation using phospho-specific antibodies

  • Interaction disruption approaches:

    • A cell-permeable peptide derived from the PDE8A Raf-1-docking sequence (residues R454-T465) can disrupt the interaction

    • Compare effects of disruptor peptide versus scrambled control peptide on complex formation and signaling outcomes

This interaction has proven functionally significant, as disruption attenuates EGF-induced morphological changes and affects Raf-1-dependent ERK signaling in multiple cell types .

What methodologies can researchers employ to study PDE8A's role in compartmentalized cAMP signaling?

PDE8A plays a crucial role in compartmentalized cAMP signaling, particularly in cardiac myocytes and airway smooth muscle cells. Several complementary approaches can investigate this function:

  • Genetic manipulation strategies:

    • PDE8A knockout mice show altered calcium handling in ventricular myocytes without changes in global PKA activity, suggesting compartment-specific effects

    • shRNA knockdown of PDE8A in human airway smooth muscle cells enhances cAMP responses to forskolin

    • Compare effects of wild-type versus catalytically inactive (dominant-negative) PDE8A overexpression

  • Pharmacological approaches:

    • PF-04957325, a PDE8-selective inhibitor, increases basal cAMP levels and enhances isoproterenol-induced inhibition of cell proliferation in airway smooth muscle cells

    • Unlike other PDE families, PDE8A is insensitive to IBMX, providing a useful pharmacological distinction

  • Subcellular localization studies:

    • Lipid raft fractionation reveals PDE8A immunoreactivity in buoyant fractions containing caveolin-1 and AC5/6

    • This localization explains its selective regulation of β2-adrenergic receptor-AC6 signaling without affecting AC2 signaling by E prostanoid receptors in non-raft domains

  • Real-time cAMP dynamics:

    • Fluorescence-based cAMP sensors in live cells can track compartmentalized cAMP changes

    • PDE8A inhibition accelerates cAMP production after β2-adrenergic stimulation but has no effect on prostaglandin E2 responses

These methodologies collectively demonstrate that PDE8A regulates specific subcellular pools of cAMP rather than global levels, highlighting the importance of precisely localized signaling modules in cellular function.

How do experimental approaches differ when studying PDE8A in cardiac versus airway smooth muscle systems?

PDE8A functions in tissue-specific signaling contexts, requiring tailored experimental approaches:

  • Cardiac system investigations:

    • In ventricular myocytes, PDE8A regulates excitation-contraction coupling and calcium handling

    • Experimental readouts include action potential-evoked calcium transients, calcium spark frequency, and L-type calcium channel currents

    • PDE8A knockout mice show higher calcium transients and increased L-type calcium channel current after isoproterenol stimulation

    • These cardiac effects occur without changes in global PKA activity, indicating compartmentalized regulation

  • Airway smooth muscle studies:

    • In human airway smooth muscle cells, PDE8A specifically modulates β2-adrenergic receptor-AC6 signaling in lipid raft microdomains

    • Key experimental readouts include cAMP accumulation, cell proliferation, and real-time cAMP dynamics using fluorescence sensors

    • PDE8A knockdown evokes twofold greater cAMP responses to forskolin in the presence of IBMX

    • AC6 overexpression (but not AC2) increases these responses by an additional 80%, confirming pathway specificity

  • Common methodological considerations:

    • Both systems benefit from combining genetic approaches (knockout/knockdown) with selective pharmacological inhibition

    • Both require careful assessment of compartmentalized versus global cAMP/PKA effects

    • Both demonstrate the importance of analyzing PDE8A in the context of its signaling partners (Raf-1, adenylyl cyclase isoforms)

These tissue-specific approaches have revealed that while the biochemical function of PDE8A remains constant (cAMP hydrolysis), its physiological roles are highly context-dependent, regulating different downstream effectors in cardiac versus airway smooth muscle cells.

Why might researchers observe multiple bands when detecting PDE8A by Western blot?

Multiple bands during PDE8A Western blot analysis are common and can arise from several biological and technical factors:

  • Multiple isoforms:

    • PDE8A has several splice variants that may appear as distinct bands

    • Observed molecular weights include both 93 kDa and 88 kDa species

    • Different tissues may express varying isoform profiles

  • Post-translational modifications:

    • Phosphorylation states can alter protein migration patterns

    • PDE8A function is regulated by phosphorylation in some contexts

  • Proteolytic degradation:

    • Insufficient protease inhibitors or sample mishandling may produce degradation fragments

    • Compare fresh samples with stored lysates to identify potential degradation patterns

  • Non-specific binding:

    • Some antibodies may exhibit cross-reactivity with related proteins

    • Optimize blocking conditions and antibody dilution to minimize this issue

    • Test multiple antibodies targeting different epitopes to confirm specificity

To differentiate authentic signals from artifacts, researchers should:

  • Include positive controls from tissues known to express PDE8A (testis, spleen)

  • Compare patterns with recombinant PDE8A standards

  • Validate specificity using PDE8A knockout or knockdown samples

  • Test multiple antibodies targeting different epitopes within PDE8A

What controls are essential when using PDE8A antibodies for protein interaction studies?

When using PDE8A antibodies to investigate protein-protein interactions, particularly with Raf-1 kinase, several critical controls ensure data reliability:

  • Input controls:

    • Analyze a fraction (5-10%) of pre-immunoprecipitation lysate to confirm target protein presence

    • Include positive control tissues/cells with known PDE8A expression (testis, spleen)

  • Immunoprecipitation controls:

    • Non-specific IgG from the same species as the PDE8A antibody to assess non-specific binding

    • Research has demonstrated that control IgG immunoprecipitates show no co-immunoprecipitating species

    • For tagged proteins, include antibodies against unrelated tags (e.g., vesicular stomatitis virus)

  • Reciprocal co-immunoprecipitation:

    • Perform bidirectional analysis (IP Raf-1 → detect PDE8A; IP PDE8A → detect Raf-1)

    • This approach confirms the interaction from both perspectives

  • Negative control lysates:

    • When available, use PDE8A knockout or knockdown cells/tissues

    • This control can identify non-specific bands and confirm antibody specificity

  • Functional validation:

    • Assess PDE activity in immunoprecipitates (PDE8A activity is uniquely IBMX-insensitive)

    • Measure effects of overexpression, knockdown, or peptide-mediated disruption on functional outcomes

These controls collectively strengthen data interpretation and ensure that observed interactions represent authentic biological phenomena rather than experimental artifacts.

How can researchers validate PDE8A antibody specificity across different experimental systems?

Thorough validation of PDE8A antibody specificity is crucial for experimental rigor and reproducibility:

  • Genetic validation approaches:

    • Test in PDE8A knockout tissues/cells – a 95 kDa band corresponding to PDE8A is absent in knockout samples

    • Use siRNA or shRNA knockdown to reduce target expression

    • Overexpress PDE8A in low-expressing cell lines to confirm signal increase

  • Epitope-specific validation:

    • Compare antibodies targeting different PDE8A regions (N-terminal, C-terminal, internal epitopes)

    • Perform peptide competition assays using the immunizing peptide

    • For C-terminal antibodies, the target sequence "VSNPCRPLQYCIEWAARISEEYFSQTDEEKQQGLPVVMPVFDRNTCSIPK" can be used for validation

  • Cross-species reactivity assessment:

    • PDE8A sequences show high conservation across species: 100% in human, mouse, cow, pig; 93% in dog, horse; 92% in guinea pig and zebrafish

    • Testing in multiple species increases confidence in antibody specificity

  • Multi-application validation:

    • Demonstrate consistent detection across different techniques (WB, IP, IHC, IF)

    • Each application provides complementary information on specificity

  • Functional validation:

    • Immunoprecipitate PDE8A and measure PDE activity that is insensitive to IBMX but inhibited by dipyridimole (PDE8-selective inhibitor)

    • Correlate protein detection with known expression patterns across tissues

Comprehensive validation should include multiple approaches, as no single method is sufficient to establish absolute specificity. Researchers should document validation results and specify which validation approaches were used when reporting experimental findings.

How can PDE8A antibodies be used to investigate compartmentalized cAMP signaling in disease models?

PDE8A antibodies enable investigation of compartmentalized cAMP signaling disruptions in various disease contexts:

  • Cardiovascular disease models:

    • PDE8A regulates cardiac excitation-contraction coupling through control of L-type calcium channel currents and sarcoplasmic reticulum calcium release

    • Antibodies can track PDE8A expression, localization, and interactions in heart failure or hypertrophy models

    • Combining immunofluorescence with calcium imaging can correlate PDE8A distribution with functional alterations

  • Respiratory disease approaches:

    • In human airway smooth muscle, PDE8A specifically modulates β2-adrenergic receptor-AC6 signaling, affecting cell proliferation

    • Antibodies can assess PDE8A expression and compartmentalization in asthma models

    • Immunohistochemical analysis of patient samples can correlate PDE8A expression with disease severity or treatment responsiveness

  • Cancer signaling studies:

    • PDE8A-Raf-1 interaction affects ERK signaling and EGF-induced responses

    • Given RAF pathway importance in cancer, antibodies can investigate PDE8A's role in tumor cell signaling

    • Immunoprecipitation approaches can determine if PDE8A-Raf-1 interactions are altered in malignant versus normal tissues

  • Methodological integration:

    • Combine PDE8A immunodetection with phospho-specific antibodies for downstream effectors

    • Correlate PDE8A localization with real-time cAMP measurements in disease models

    • Use proximity ligation assays to visualize PDE8A interactions with signaling partners in situ

These approaches allow researchers to not only correlate PDE8A expression with disease states but also to mechanistically understand how alterations in compartmentalized cAMP signaling contribute to pathophysiology.

What are the advantages and limitations of different PDE8A antibody types for mechanistic studies?

Different PDE8A antibody types offer distinct advantages and limitations for mechanistic investigations:

Antibody TypeAdvantagesLimitationsBest Applications
Polyclonal (C-terminal)Detects multiple epitopes; High sensitivity; Works in multiple species Batch-to-batch variation; Potential cross-reactivityWestern blot; IP; IHC
MonoclonalConsistent performance; High specificity; Reduced backgroundMay lose reactivity if epitope is modified; Limited epitope recognitionWestern blot; Highly quantitative applications
HRP-conjugatedDirect detection without secondary antibody; Reduced assay time Limited signal amplification; Fixed detection systemELISA; Rapid Western blot
Epitope-specificCan distinguish isoforms; Target specific domainsMay miss isoforms lacking the epitopeIsoform-specific detection

For mechanistic studies investigating PDE8A-Raf-1 interactions, unconjugated polyclonal antibodies recognizing the C-terminal region have proven particularly effective . This region contains parts of the Raf-1 interaction domain, making these antibodies valuable for studying how this interaction is regulated.

When selecting antibodies for specific applications, researchers should consider:

  • The specific PDE8A domain/region being studied

  • Required detection sensitivity

  • Need for consistent performance in quantitative studies

  • Compatibility with sample types (fresh frozen vs. fixed tissues)

  • Previous validation in similar experimental systems

How should researchers interpret conflicting data when studying PDE8A in different cell types?

When encountering conflicting data regarding PDE8A across different cell types, systematic analysis can resolve apparent contradictions:

  • Cell-type specific expression patterns:

    • PDE8A expression varies dramatically across tissues, with highest levels in testis, ovary, small intestine, and colon

    • Expression level differences may explain varying functional importance

  • Subcellular localization differences:

    • In human airway smooth muscle cells, PDE8A localizes in lipid raft microdomains with caveolin-1 and AC5/6

    • In other cell types, distribution may differ, affecting which signaling pathways it regulates

    • Use fractionation studies and co-localization immunofluorescence to resolve differences

  • Signaling partner availability:

    • PDE8A interacts with Raf-1 with extremely high affinity (Kd <61 pM)

    • Differential expression of Raf-1 or other partners across cell types affects PDE8A function

    • Assess expression of key interacting proteins in each cell type

  • Methodological considerations:

    • Different detection methods have varying sensitivities

    • Knockout/knockdown efficiency varies across cell types

    • Standardize methods across cell types when possible

  • Physiological context integration:

    • In cardiac myocytes, PDE8A regulates calcium handling without affecting global PKA activity

    • In airway smooth muscle, it selectively regulates β2-adrenergic receptor-AC6 signaling without affecting prostaglandin E2 responses

    • These distinct roles reflect tissue-specific signaling architectures rather than contradictions

When reporting seemingly conflicting results, researchers should explicitly describe cell type-specific conditions, highlight methodological differences, and consider how tissue-specific signaling environments may produce different functional outcomes despite conserved biochemical mechanisms.

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.