Phospho-PRKD1 (Ser738) Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Properties and Development

The Phospho-PRKD1 (Ser738) Antibody is a rabbit-derived polyclonal antibody generated using a synthetic phosphopeptide immunogen corresponding to the sequence around Ser738 (E-K-S(p)-F-R) in human PRKD1 . Key characteristics include:

PropertyDetails
Host SpeciesRabbit
ReactivityHuman, Mouse, Rat
ClonalityPolyclonal
ApplicationsWestern Blot (WB), ELISA, Immunohistochemistry (IHC), Immunofluorescence (IF)
Recommended DilutionsWB: 1:500–1:1000; ELISA: 1:2000–1:10,000; IF: 1:100–1:200
Storage1.0 mg/mL in PBS with 50% glycerol; store at -20°C or -80°C

The antibody undergoes stringent purification via affinity chromatography to remove non-phospho-specific cross-reactivity, ensuring high specificity for phosphorylated Ser738 .

Biological Significance of PRKD1 Phosphorylation

Phosphorylation at Ser738 (and the adjacent Ser742 in PRKD1/2/3 isoforms) regulates PRKD1 kinase activity, influencing:

  • Cellular Signaling: Modulates MAPK/JNK, Ras, and NF-κB pathways .

  • Golgi Function: Maintains Golgi membrane integrity and vesicle transport .

  • Disease Mechanisms:

    • Promotes cardiac hypertrophy via HDAC5 phosphorylation .

    • Enhances VEGF-induced angiogenesis .

    • Mediates genotoxic stress-induced apoptosis through caspase-3 cleavage .

A. Western Blot Validation

The antibody detects endogenous PRKD1 phosphorylation in cell lines like A549 (lung carcinoma) and MCF7 (breast cancer) under PMA stimulation . Preabsorption with the immunizing peptide abolishes signal, confirming specificity .

B. Functional Studies

  • Cardiac Research: Identifies PRKD1 activation in hypertrophic cardiomyocytes .

  • Cancer Biology: Links PRKD1 phosphorylation to EGFR and MAPK/ERK signaling in tumor proliferation .

  • Neuroscience: Visualizes PRKD1 roles in dendritic arborization via IF in hippocampal cells .

Validation Data

Figure 1: Western blot of PMA-treated A549 cells shows a strong band at ~110 kDa (PRKD1’s molecular weight). Signal elimination with competitive peptide confirms specificity .
Figure 2: Immunofluorescence in HeLa cells reveals cytoplasmic and trans-Golgi network localization .

Limitations and Considerations

  • Isoform Cross-Reactivity: Some antibodies (e.g., PACO23993) detect phospho-Ser738/742 in PRKD1/2/3, requiring isoform-specific validation .

  • Species Restrictions: Not validated for non-mammalian systems .

Product Specs

Form
Supplied at 1.0mg/mL in phosphate buffered saline (without Mg2+ and Ca2+), pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Lead Time
Typically, we can ship your orders within 1-3 business days after receiving them. Delivery time may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery timeframes.
Synonyms
KPCD1_HUMAN antibody; nPKC D1 antibody; nPKC mu antibody; nPKC-D1 antibody; nPKC-mu antibody; nPKCD1 antibody; nPKCmu antibody; PKC antibody; PKC MU antibody; PKCM antibody; PKCmu antibody; PKD 1 antibody; PKD antibody; PKD1 antibody; PRKCM antibody; PRKD 1 antibody; Prkd1 antibody; Protein kinase C mu antibody; Protein kinase C mu type antibody; Protein kinase D antibody; Protein kinase D1 antibody; Serine/threonine protein kinase D1 antibody; Serine/threonine-protein kinase D1 antibody
Target Names
Uniprot No.

Target Background

Function
Protein kinase D1 (PRKD1) is a serine/threonine-protein kinase that converts transient diacylglycerol (DAG) signals into prolonged physiological effects downstream of PKC. It plays a crucial role in regulating various cellular processes, including:
  • MAPK8/JNK1 and Ras signaling
  • Golgi membrane integrity and trafficking
  • Cell survival through NF-kappa-B activation
  • Cell migration
  • Cell differentiation by mediating HDAC7 nuclear export
  • Cell proliferation via MAPK1/3 (ERK1/2) signaling

PRKD1 is involved in several physiological and pathological processes such as cardiac hypertrophy, VEGFA-induced angiogenesis, genotoxic-induced apoptosis, and flagellin-stimulated inflammatory response.

PRKD1 exerts its diverse functions through a range of mechanisms:

  • Phosphorylation of the epidermal growth factor receptor (EGFR) on dual threonine residues, leading to the suppression of epidermal growth factor (EGF)-induced MAPK8/JNK1 activation and subsequent JUN phosphorylation.
  • Phosphorylation of RIN1, inducing RIN1 binding to 14-3-3 proteins YWHAB, YWHAE, and YWHAZ, thereby increasing competition with RAF1 for binding to GTP-bound form of Ras proteins (NRAS, HRAS, and KRAS).
  • Acting downstream of the heterotrimeric G-protein beta/gamma-subunit complex to maintain the structural integrity of the Golgi membranes, which is essential for protein transport along the secretory pathway.
  • Regulating the fission of transport vesicles in the trans-Golgi network (TGN) that are destined for the plasma membrane.
  • Activation of the lipid kinase phosphatidylinositol 4-kinase beta (PI4KB) at the TGN to locally synthesize phosphorylated inositol lipids, which induce a sequential production of DAG, phosphatidic acid (PA), and lyso-PA (LPA) that are essential for membrane fission and generation of specific transport carriers to the cell surface.
  • Phosphorylation at Tyr-463 via SRC-ABL1 under oxidative stress, contributing to cell survival by activating IKK complex and subsequent nuclear translocation and activation of NFKB1.
  • Regulation of integrin alpha-5/beta-3 recycling and promotion of its recruitment in newly forming focal adhesion, thereby playing a role in cell migration.
  • Mediating the bone morphogenetic protein 2 (BMP2)-induced nuclear export of HDAC7 in osteoblast differentiation, leading to the inhibition of HDAC7 transcriptional repression of RUNX2.
  • Regulating the biogenesis of TGN-derived dendritic vesicles and contributing to the maintenance of dendritic arborization and Golgi structure in hippocampal cells, highlighting its crucial role in neuronal polarity.
  • Potentiation of mitogenesis induced by the neuropeptide bombesin or vasopressin by mediating an increase in the duration of MAPK1/3 (ERK1/2) signaling, resulting in the accumulation of immediate-early gene products such as FOS that stimulate cell cycle progression.
  • Crucial involvement in the proliferative response induced by low calcium in keratinocytes through sustained activation of the MAPK1/3 (ERK1/2) pathway.
  • Participation in cardiac hypertrophy downstream of novel PKC signaling by phosphorylating HDAC5, which triggers XPO1/CRM1-dependent nuclear export of HDAC5, MEF2A transcriptional activation, and induction of downstream target genes that promote myocyte hypertrophy and pathological cardiac remodeling.
  • Mediation of cardiac troponin I (TNNI3) phosphorylation at the PKA sites, leading to reduced myofilament calcium sensitivity and accelerated crossbridge cycling kinetics.
  • Involvement in angiogenesis through the PRKD1-HDAC5 pathway by mediating VEGFA-induced specific subset of gene expression, cell migration, and tube formation.
  • Necessity for HDAC7 phosphorylation in response to VEGFA, which induces HDAC7 nuclear export and endothelial cell proliferation and migration.
  • Cleavage by caspase-3 at Asp-378 during apoptosis induced by cytarabine and other genotoxic agents, resulting in activation of its kinase function and increased sensitivity of cells to the cytotoxic effects of genotoxic agents.
  • Essential role in transducing flagellin-stimulated inflammatory responses in epithelial cells by binding and phosphorylating TLR5, contributing to MAPK14/p38 activation and production of inflammatory cytokines.
  • Potential involvement in inflammatory response by mediating activation of NF-kappa-B.
  • Possible role in pain transmission by directly modulating TRPV1 receptor.
  • Regulation of activated KRAS-mediated stabilization of ZNF304 in colorectal cancer (CRC) cells.
  • Regulation of nuclear translocation of transcription factor TFEB in macrophages upon live S.enterica infection.
Gene References Into Functions
  1. PKD1 regulates the hypoxic glycolytic metabolism of cancer cells via regulation of the expression of HIF-1alpha and glycolytic enzymes. PMID: 29901206
  2. This research elucidates a novel mechanism governing PRKD1 gene expression in pancreatic ductal adenocarcinoma, establishing a functional link between oncogenic KRas, NF-kappaB, and expression of PRKD1. PMID: 27649783
  3. The p110alpha subunit of PI3K and PKD mediate YAP activation in response to insulin and neurotensin in pancreatic cancer cells. Inhibitors of PI3K or PKD disrupt crosstalk between insulin receptor and GPCR signaling systems by blocking YAP/TEAD-regulated gene expression in pancreatic cancer cells. PMID: 28360038
  4. High PRKD1 expression is associated with drug resistance in breast cancer. PMID: 26895471
  5. Our findings directly associate the AR/NCOA1 complex with PRKD1 regulation and cellular migration, supporting the concept of therapeutic inhibition of NCOA1 in prostate cancer. PMID: 27255895
  6. No Polymorphous low-grade adenocarcinoma (PLGA) lacking PRKD1 somatic mutations or PRKD gene family rearrangements harbored somatic mutations in the kinase domains of the PRKD2 or PRKD3 genes. PMID: 26426580
  7. A single nucleotide polymorphism located within the fourth intron of PRKD1 (rs57803087) was strongly associated with DPP-4 inhibitor response in patients with type 2 diabetes. PMID: 28160554
  8. Mutation in the PRKD1 gene is associated with congenital heart defects. PMID: 27479907
  9. Bradykinin stimulates myofibroblast migration through protein kinase D-mediated activation of COX-2 and Hsp27. PMID: 28032559
  10. Lysophosphatidic acid/PKD-1 signaling leads to nuclear accumulation of histone deacetylase 7, where it interacts with forkhead box protein O1 to suppress endothelial CD36 transcription and mediates silencing of the antiangiogenic switch, resulting in proangiogenic and proarteriogenic reprogramming. PMID: 27013613
  11. This study identified and characterized a novel, highly conserved N-terminal domain, comprising 92 amino acids, which mediates dimerization of Protein Kinase D (PKD) isoforms, PKD1, PKD2, and PKD3 monomers. PMID: 27662904
  12. Mast cell (MC) stimulation by physical contact with T cells results in PKD activation, leading to the phosphorylation of p38, degranulation, and release of cytokines. Understanding the molecular events associated with T cell-induced MC activation might lead to therapeutic approaches for controlling T cell-mediated inflammatory processes in which MC participate. PMID: 28049203
  13. Data suggest the role of the phospholipase C epsilon-Protein kinase D-PEA15 protein-ribosomal S6 kinase-IkappaB-NF-kappa B pathway in facilitating inflammation and inflammation-associated carcinogenesis in the colon. PMID: 27053111
  14. PRKD1 Mutation is not associated with Solid Tumors and Leukemias. PMID: 26518775
  15. Knockdown of PKD1 did not affect NMDAR internalization but prevented the phosphorylation and inhibition of remaining surface NMDARs and NMDAR-mediated synaptic functions. PMID: 26584860
  16. Studies indicate that the loss of protein kinase D PKD1 is thought to promote invasion and metastasis, while PKD2 and upregulated PKD3 are positive regulators of proliferation. PMID: 26253275
  17. It is highly possible that PKD1 plays a critical role in signal transduction from the PKC pathway to the tyrosine kinase pathway. PMID: 26338704
  18. Positional mapping of PRKD1, NRP1, and PRDM1 as novel candidate disease genes in truncus arteriosus. PMID: 25713110
  19. Protein kinase D is increased and activated in lung epithelial cells and macrophages in idiopathic pulmonary fibrosis. PMID: 25000413
  20. A positive relationship between L1 and pPKD1 was observed in both cultured cerebellar neurons and human cerebellar tissue, suggesting that L1 functions in the modulation of PKD1 phosphorylation. PMID: 25445362
  21. Results demonstrate a putative tumor-suppressor function of PKD1 in colon tumorigenesis via modulation of beta-catenin functions in cells. PMID: 25149539
  22. PRKD1 is aberrantly methylated and silenced in its expression in invasive breast cancer. PMID: 23971832
  23. A novel and recurrent gene rearrangements in PRKD1-3 primarily in cribriform adenocarcinoma of minor salivary gland is described, suggesting a possible pathogenetic dichotomy from "classic" polymorphous low-grade adenocarcinoma. PMID: 24942367
  24. PRKD1 hotspot mutations encoding p.Glu710Asp in 72.9% of polymorphous low-grade adenocarcinomas, but not in other salivary gland tumors. PMID: 25240283
  25. PKD1 may impair cancer cell motility and invasive properties by specific interaction with SSH1L at the cell periphery and phosphorylation of the Ser-978 substrate motif. PMID: 24336522
  26. PRKD1 mRNA was significantly upregulated in esophageal squamous cell carcinoma compared to non-tumorous tissue. PMID: 23621299
  27. Protein kinase D1 is essential for Ras-induced senescence and tumor suppression by regulating senescence-associated inflammation. PMID: 24828530
  28. High PRKD1 along with positive nodal status correlate with the recurrence of primary laryngeal cancer. PMID: 23950933
  29. This review addresses the role of PKD in the organization of the actin cytoskeleton with a particular emphasis on the substrates associated with this function. PKD regulates cancer cell migration and invasion. [review] PMID: 23688773
  30. PKD1 directly phosphorylates VASP at two serine residues, Ser-157 and Ser-322. These phosphorylations occur in response to RhoA activation and mediate VASP re-localization from focal contacts to the leading edge region. PMID: 23846685
  31. These results suggest that respiratory syncytial virus-induced airway epithelial barrier disruption involves PKD-dependent actin cytoskeletal remodeling, possibly dependent on cortactin activation. PMID: 23926335
  32. These results indicate that PKD is downstream of PLD and suggest that PKD is one of the mechanisms through which PLD promotes aldosterone production in response to AngII in adrenal glomerulosa cells. PMID: 23178798
  33. Neuregulin mediates F-actin-driven cell migration through inhibition of protein kinase D1 via Rac1 protein. PMID: 23148218
  34. The PKD pathway couples receptor tyrosine kinase signaling to an integrin switch via Rabaptin-5 phosphorylation. PMID: 22975325
  35. The role of PKD is found to mediate the regulation of vascular morphogenesis. PMID: 22855295
  36. Snail1 and its phosphorylation at Ser-11 were required and sufficient to control PKD1-mediated anchorage-independent growth and anchorage-dependent proliferation of different tumor cells. PMID: 22791710
  37. CERT is at a convergence point of non-vesicular and vesicular transport processes and plays a central role within the PKD signaling network at the Golgi complex. (Review) PMID: 22226883
  38. PKCmicro isoform is an important factor in the abnormal growth of vascular endothelial cells induced by 1,2-dimethylthdrazine. PMID: 22664730
  39. PKD1 overexpression increases the aggressiveness of MCF-7 breast cancer cells through enhancing their oncogenic properties. PMID: 22245102
  40. Results describe PKD as a novel Vps34 kinase that functions as an effecter of autophagy under oxidative stress. PMID: 22095288
  41. Protein kinase D regulates RhoA activity via phosphorylation rhotekin at Ser-435. PMID: 22228765
  42. Data showed that regulation of SNAI1 through PKD1 occurs in vivo in normal breast ductal tissue and is decreased or lost in invasive ductal carcinoma. PMID: 22276203
  43. It is increasingly apparent that PKD1 is a key player in the regulation of cardiac hypertrophy, most likely through its effect on the transcriptional regulation of fetal gene programming via the phosphorylation of HDAC5. [Review] PMID: 22260707
  44. Downregulation of PKD1 expression may determine the behavior of gastric tumor cells, promoting an invasive phenotype and potentially resulting in a general poor prognosis. PMID: 22217708
  45. Agonist-dependent increases in diacylglycerol accumulation lead to the activation of protein kinase C and PKC-dependent phosphorylation of PKD1 at two conserved serine residues in the activation loop; this modification increases PKD1 catalytic activity. PMID: 22188925
  46. PAR(1) and PAR(2) are involved in WM9 cell proliferation and secretion of IL-8 by activation of PKD1. PMID: 21993564
  47. Serine 1884 is essential for the regulation of hCaV1.2 by PKD. PMID: 22100296
  48. Protein kinase D activity is essential for exercise-induced MEF2-dependent skeletal muscle remodelling in vivo. PMID: 21848513
  49. PolyI:C-dependent barrier disruption is mediated by disassembly of epithelial apical junctions, which is dependent on PKD signaling. PMID: 21996340

Show More

Hide All

Database Links

HGNC: 9407

OMIM: 605435

KEGG: hsa:5587

STRING: 9606.ENSP00000333568

UniGene: Hs.508999

Involvement In Disease
Congenital heart defects and ectodermal dysplasia (CHDED)
Protein Families
Protein kinase superfamily, CAMK Ser/Thr protein kinase family, PKD subfamily
Subcellular Location
Cytoplasm. Cell membrane. Golgi apparatus, trans-Golgi network. Note=Translocation to the cell membrane is required for kinase activation.

Q&A

What are PRKD1, PRKD2, and PRKD3 proteins and what role does phosphorylation at Ser738/742 play?

PRKD1, PRKD2, and PRKD3 are protein kinases that belong to the protein kinase C (PKC) family, with PRKD1 also known as PKCμ. These serine/threonine kinases play crucial roles in multiple cellular functions including cell proliferation, differentiation, and apoptosis. The phosphorylation at serine residues 738 and 742 is particularly significant as it regulates their kinase activity and influences downstream signaling cascades. This phosphorylation serves as an activation mechanism that enables these kinases to participate in mitogenic signal transduction pathways, including the activation of p42/p44 MAPK leading to Elk1-mediated gene transcription. Understanding this phosphorylation is essential for characterizing normal cellular function and pathological conditions where these kinases are implicated.

What are the key applications for Phospho-PRKD1 (Ser738) Antibody in research?

Phospho-PRKD1 (Ser738) Antibody serves multiple critical applications in signaling research:

  • Western Blot (WB): Enables detection and quantification of phosphorylated PRKD1 at approximately 115kDa. Recommended dilutions range from 1:500-1:2000, allowing researchers to monitor activation states across different experimental conditions.

  • Enzyme-Linked Immunosorbent Assay (ELISA): Facilitates high-throughput screening with dilutions of 1:2000-1:10000, suitable for large-scale phosphorylation studies.

  • Immunohistochemistry (IHC): Used at dilutions of 1:100-1:300 to visualize phosphorylated PRKD1 in tissue sections, providing spatial information about kinase activation.

  • Immunofluorescence (IF): Applied at dilutions of 1:100-1:200 to localize phosphorylated PRKD1 in cells, enabling subcellular distribution studies.

These applications collectively provide researchers with tools to investigate PRKD1 activation in various experimental contexts, from protein expression to subcellular localization.

What species reactivity can be expected with Phospho-PRKD1/PRKD2/PRKD3 antibodies?

Phospho-PRKD1/PRKD2/PRKD3 antibodies demonstrate cross-reactivity with samples from multiple mammalian species. Based on the available data, these antibodies have been validated for:

  • Human samples

  • Mouse samples

  • Rat samples

This multi-species reactivity is particularly valuable for comparative studies and translational research where findings from animal models need to be verified in human systems. The conservation of the phosphorylation sites (Ser738/742) across these species indicates the evolutionary importance of these regulatory mechanisms. Researchers should note that while reactivity has been confirmed in these three species, optimization may still be required when using these antibodies in specific tissue types or under particular experimental conditions.

How does phosphorylation at Ser738 relate to activation of PRKD1 compared to other phosphorylation sites?

The phosphorylation of PRKD1 at Ser738 represents a critical activation step that distinguishes it from other regulatory phosphorylation events. While multiple phosphorylation sites exist on PRKD1, Ser738 (along with Ser742) phosphorylation occurs within the activation loop and serves as a direct indicator of kinase activation status. This phosphorylation is typically mediated by upstream PKC isoforms in a PKC-dependent pathway, creating a hierarchical signaling cascade. Unlike phosphorylation events at other sites (such as those in regulatory domains), Ser738 phosphorylation directly correlates with catalytic activity and the ability to phosphorylate downstream substrates.

Research has demonstrated that Ser738/742 phosphorylation precedes and enables the activation of p42/p44 MAPK pathways and subsequent Elk1-mediated transcriptional activation. When studying PRKD1 activation dynamics, monitoring Ser738 phosphorylation provides a more direct measurement of kinase activity than assessing total protein levels or phosphorylation at regulatory sites. This distinction is particularly important when investigating temporal aspects of signaling or when determining the effects of potential inhibitors on the PKC-PRKD axis.

What are the methodological considerations for comparing phospho-specific versus total PRKD1 detection in experimental design?

When designing experiments to investigate PRKD1 phosphorylation status, several methodological considerations must be addressed to ensure accurate interpretation:

Sample preparation protocol:

  • Phosphatase inhibitors must be included in lysis buffers to prevent dephosphorylation during sample preparation

  • Standardized protein extraction methods should be employed to ensure comparable phosphorylation states

  • Sample processing time should be minimized to preserve labile phosphorylation sites

Dual detection approach:

  • Parallel blots or sequential probing with phospho-specific and total PRKD1 antibodies

  • Calculation of phospho-to-total PRKD1 ratios to normalize for expression level variations

  • Use of appropriate loading controls for both phosphorylated and total protein quantification

Controls and validation:

  • Positive controls using PKC activators (e.g., PMA) to induce Ser738 phosphorylation

  • Negative controls using PKC inhibitors to demonstrate specificity

  • Phosphatase treatment of duplicate samples to confirm phospho-specificity

This methodological framework enables researchers to distinguish between changes in PRKD1 activation versus alterations in total protein expression, a critical distinction when studying signaling dynamics or drug responses.

How can researchers distinguish between PRKD1, PRKD2, and PRKD3 phosphorylation when using antibodies that recognize all three isoforms?

Distinguishing between PRKD1, PRKD2, and PRKD3 phosphorylation requires a strategic experimental approach when using antibodies that recognize the conserved phosphorylation sites across all three isoforms:

Molecular weight discrimination:

  • PRKD1: ~115 kDa

  • PRKD2: ~105 kDa

  • PRKD3: ~110 kDa

Combinatorial antibody approaches:

  • Use the pan-phospho-PRKD1/2/3 antibody to detect all phosphorylated isoforms

  • In parallel experiments, employ isoform-specific antibodies against total PRKD1, PRKD2, or PRKD3

  • Perform immunoprecipitation with isoform-specific antibodies followed by detection with the phospho-specific antibody

Genetic manipulation techniques:

  • siRNA/shRNA knockdown of specific isoforms to identify band disappearance

  • Overexpression of individual isoforms to confirm band identity

  • CRISPR/Cas9 knockout validation to establish definitive isoform identification

This multi-faceted approach allows researchers to confidently assign phosphorylation signals to specific PRKD isoforms, critical for understanding their differential roles in signaling pathways.

What are the optimal conditions for antibody storage and handling to maintain phospho-specificity?

Maintaining phospho-specificity of PRKD1/PRKD2/PRKD3 antibodies requires adherence to specific storage and handling protocols:

Storage conditions:

  • Long-term storage: -20°C is recommended for all antibody formats

  • Avoid repeated freeze-thaw cycles (limit to <5 cycles)

  • Store in small aliquots to minimize freeze-thaw exposure

  • Store in the original buffer containing 50% glycerol, which prevents freezing damage

Handling guidelines:

  • When removing from storage, thaw antibodies on ice

  • Centrifuge briefly before opening to collect solution at the bottom of the tube

  • Use sterile technique when accessing antibody solutions

  • Return to -20°C immediately after use

Stability considerations:

  • Antibodies remain stable for at least one year when stored properly at -20°C

  • Working solutions should be prepared fresh and used within 24 hours

  • Monitor performance periodically through positive controls

The phosphorylated epitope recognized by these antibodies is particularly sensitive to storage conditions. Proper maintenance of storage temperature and minimizing exposure to degradative enzymes are essential for preserving specificity for the phosphorylated versus non-phosphorylated forms of the protein.

What methodologies can be employed to validate the specificity of phospho-PRKD1 (Ser738) antibody detection?

Validating the specificity of phospho-PRKD1 (Ser738) antibody detection requires a multi-pronged approach:

Biochemical validation:

  • Lambda phosphatase treatment: Sample splitting and treating one portion with lambda phosphatase should eliminate the signal detected by the phospho-specific antibody

  • Blocking peptide competition: Pre-incubation of the antibody with the phosphorylated immunogen peptide should abolish specific signals

  • Non-phosphorylated peptide competition: Pre-incubation with the non-phosphorylated version of the same peptide should not affect signal intensity

Biological validation:

  • PKC activation: Treatment of cells with PKC activators (e.g., PMA) should increase the phospho-PRKD1 signal

  • PKC inhibition: Pre-treatment with PKC inhibitors should block stimulus-induced increases in phospho-PRKD1 signal

  • Genetic manipulation: Mutation of the Ser738 site to alanine should eliminate antibody recognition

Analytical validation:

  • Side-by-side comparison with other commercially available phospho-PRKD1 (Ser738) antibodies

  • Correlation of phospho-signal with functional outputs known to depend on PRKD1 activation

  • Mass spectrometry confirmation of phosphorylation status at Ser738

How can researchers address inconsistent Western blot results when using phospho-PRKD1 (Ser738) antibodies?

Inconsistent Western blot results when using phospho-PRKD1 (Ser738) antibodies can be systematically addressed through the following troubleshooting approach:

Sample preparation issues:

  • Ensure complete and consistent inhibition of phosphatases by using fresh phosphatase inhibitor cocktails

  • Standardize the time between cell lysis and protein denaturation to prevent variability in phosphorylation status

  • Verify protein concentration measurement accuracy using multiple methods (e.g., Bradford and BCA)

Technical parameters:

  • Optimize antibody dilution within the recommended range (1:500-1:2000 for Western blot)

  • Extend primary antibody incubation time (overnight at 4°C) to improve signal consistency

  • Adjust blocking conditions to reduce background while maintaining specific signal

Signal detection refinement:

  • Evaluate different membrane types (PVDF vs. nitrocellulose) for optimal signal-to-noise ratio

  • Test various detection systems (chemiluminescence vs. fluorescence-based methods)

  • Implement quantitative analysis with appropriate normalization to loading controls

Experimental design adjustments:

  • Include positive controls (PMA-treated cells) and negative controls (PKC inhibitor-treated cells) in each experiment

  • Run time-course experiments to identify optimal time points for phosphorylation detection

  • Consider the impact of cell density and growth conditions on baseline phosphorylation levels

This systematic approach allows researchers to isolate and address specific factors contributing to inconsistent results, leading to more reliable and reproducible detection of phospho-PRKD1.

What are the key considerations for quantitative analysis of phospho-PRKD1 (Ser738) immunoblotting data?

Conducting rigorous quantitative analysis of phospho-PRKD1 (Ser738) immunoblotting data requires attention to several critical factors:

Normalization strategies:

Normalization ApproachAdvantagesLimitations
Total PRKD1 normalizationDirectly accounts for variation in PRKD1 expressionRequires stripping and reprobing or parallel blots
Loading control normalization (e.g., β-actin, GAPDH)Simple implementationMay not account for PRKD1-specific expression changes
Phospho/Total ratio determinationMost accurate representation of activation stateRequires consistent detection of both forms

Statistical considerations:

  • Perform experiments with sufficient biological replicates (minimum n=3)

  • Use appropriate statistical tests based on data distribution (parametric vs. non-parametric)

  • Report both fold-change and statistical significance

  • Consider time-dependent changes when comparing activation states

Technical recommendations:

  • Ensure signal is within the linear range of detection for accurate quantification

  • Avoid saturation of signal which prevents accurate quantification

  • Use standard curves with recombinant phosphorylated proteins for absolute quantification

  • Employ image analysis software that can account for background and normalize band intensities

Reporting standards:

  • Present both representative blot images and quantification graphs

  • Include all experimental conditions in a single graph for direct comparison

  • Report variability measures (standard deviation or standard error)

  • Document exact exposure conditions and image acquisition parameters

Adherence to these quantitative analysis principles ensures reliable interpretation of phospho-PRKD1 (Ser738) data and enables meaningful comparisons across experimental conditions.

How can contradictory results between phosphorylation status and functional outcomes be reconciled?

Reconciling contradictory results between PRKD1 phosphorylation status and functional outcomes requires systematic investigation of multiple factors that may explain the discrepancy:

Temporal dynamics assessment:

  • Phosphorylation may be transient while functional outcomes persist

  • Time-course experiments with fine temporal resolution can reveal disconnects between immediate phosphorylation and delayed functional responses

  • Consider kinetics of phosphorylation versus dephosphorylation rates at different sites

Multi-site phosphorylation analysis:

  • While Ser738 phosphorylation is important, additional regulatory phosphorylation sites may influence function

  • Comprehensive phospho-profiling using mass spectrometry can identify other relevant sites

  • Ser742 phosphorylation often occurs in conjunction with Ser738 and may be required for full activation

Subcellular localization factors:

  • Phosphorylated PRKD1 may require proper subcellular localization to access substrates

  • Combine phospho-detection with subcellular fractionation or immunofluorescence to track localization

  • Consider the role of scaffolding proteins in facilitating or preventing functional interactions

Pathway crosstalk and compensatory mechanisms:

  • Parallel signaling pathways may compensate for changes in PRKD1 activity

  • Inhibitor panels targeting multiple pathways can identify compensatory mechanisms

  • Systems biology approaches may help model complex signaling networks

Experimental validation strategies:

  • Use constitutively active and dominant-negative PRKD1 mutants to confirm causal relationships

  • Employ phosphomimetic (S738D) and phospho-deficient (S738A) mutants to directly test the role of phosphorylation

  • Utilize CRISPR/Cas9 genome editing to modify endogenous phosphorylation sites

This comprehensive approach enables researchers to identify the mechanistic basis for apparent contradictions between phosphorylation status and functional outcomes, leading to more nuanced understanding of PRKD1 signaling complexity.

How can phospho-PRKD1 (Ser738) antibodies be incorporated into high-throughput screening approaches?

Incorporating phospho-PRKD1 (Ser738) antibodies into high-throughput screening (HTS) approaches enables large-scale investigations of PRKD1 activation in response to various stimuli or inhibitors:

ELISA-based screening platforms:

  • Develop sandwich ELISA with capture antibody against total PRKD1 and detection antibody against phospho-Ser738

  • Optimize antibody dilutions (1:2000-1:10000) for maximal signal-to-noise ratio in 96- or 384-well formats

  • Implement automated liquid handling systems for consistent reagent addition and washing steps

Cell-based assay adaptation:

  • In-cell Western techniques using fluorescently-labeled secondary antibodies

  • High-content imaging with phospho-PRKD1 (Ser738) antibodies to simultaneously assess phosphorylation and subcellular localization

  • Phospho-flow cytometry for single-cell resolution of PRKD1 activation in heterogeneous populations

Multiplexed detection systems:

  • Multiplex with other phospho-specific antibodies to map pathway activation networks

  • Combine with phenotypic readouts to correlate PRKD1 phosphorylation with functional outcomes

  • Implement bead-based multiplex assays for simultaneous detection of multiple analytes

Data analysis and integration:

  • Develop phosphorylation signatures associated with specific cellular responses

  • Machine learning approaches to identify patterns in complex phosphorylation datasets

  • Integration with other -omics data (transcriptomics, metabolomics) for systems-level understanding

These HTS approaches provide researchers with powerful tools to identify novel regulators of PRKD1 activation, screen for pathway-specific inhibitors, and understand the position of PRKD1 in complex signaling networks across large sample sets or compound libraries.

What are the emerging applications for studying phospho-PRKD1 (Ser738) in disease models and therapeutic development?

The study of phospho-PRKD1 (Ser738) in disease models and therapeutic development represents an expanding frontier with several important applications:

Cancer research applications:

  • Monitor PRKD1 activation status across cancer types and stages to identify dysregulated signaling

  • Correlate phospho-PRKD1 levels with tumor aggressiveness and treatment response

  • Evaluate PRKD1 inhibition as a therapeutic strategy in cancer types with hyperactivated signaling

Cardiovascular disease investigations:

  • Study the role of PRKD1 phosphorylation in cardiac hypertrophy and heart failure models

  • Assess PRKD1 activation in vascular smooth muscle cells during atherosclerosis progression

  • Develop therapeutic approaches targeting PRKD1 activation in cardiac remodeling

Inflammatory and immune response research:

  • Monitor PRKD1 phosphorylation during T-cell activation and inflammatory responses

  • Investigate the role of PRKD1 in cytokine production and inflammatory signaling cascades

  • Develop immunomodulatory strategies based on PRKD1 pathway regulation

Therapeutic development considerations:

  • Use phospho-PRKD1 (Ser738) as a pharmacodynamic biomarker in drug development

  • Screen for compounds that selectively modulate phosphorylation at specific sites

  • Develop combination therapies targeting multiple nodes in PRKD1-related signaling networks

Technical innovations:

  • Implement tissue microarray analysis with phospho-PRKD1 antibodies for large-scale patient sample analysis

  • Develop imaging techniques to monitor PRKD1 activation in living systems

  • Create patient-derived organoid models to study PRKD1 phosphorylation in personalized medicine applications

These emerging applications highlight the translational potential of phospho-PRKD1 research from basic cellular mechanisms to clinical applications, positioning PRKD1 as an important target for therapeutic intervention across multiple disease states.

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.