Phospho-PRKCA (T638) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Production and Mechanism

This antibody is generated using recombinant technology:

  • Immunogen Design: A synthetic peptide mimicking phosphorylated T638 is used to immunize rabbits or HEK293F cells .

  • Cloning and Expression: Heavy and light chain genes are isolated, cloned into mammalian expression vectors, and transfected into host cells (e.g., HEK293F) for high-yield production .

  • Purification: Affinity chromatography ensures high specificity and purity .

The antibody binds selectively to phosphorylated T638, a critical site for PKC-α activation. PKC-α is a serine/threonine kinase activated by calcium and diacylglycerol, playing roles in cell adhesion, migration, and survival .

Research Applications

The antibody is validated for three primary techniques:

ApplicationPurposeRecommended DilutionKey Uses
Western Blot (WB)Detect phosphorylated PKC-α in lysates1:500–1:5000 Confirming phosphorylation status
Immunofluorescence (IF)Localize PKC-α in fixed cells1:20–1:200 Studying subcellular distribution
ELISAQuantify phosphorylated PKC-α levelsN/A (supplier-specific)High-throughput screening

Example: In hepatocellular carcinoma studies, the antibody has been used to demonstrate PKC-α T638 phosphorylation in HepG2 cells via IF, highlighting its role in tumorigenesis .

PKC-α T638 Phosphorylation

  • Activation Mechanism: Phosphorylation at T638 stabilizes PKC-α’s active conformation, enabling substrate binding and kinase activity .

  • Disease Implications: Dysregulated PKC-α signaling is linked to cancer (e.g., breast, liver), cardiovascular diseases, and inflammatory disorders .

Key Findings

  1. Cancer Research:

    • PKC-α T638 phosphorylation correlates with metastatic potential in breast cancer models .

    • PRKCA gene mutations are implicated in pigment epithelial melanoma and glioblastoma .

  2. Cardiovascular Function:

    • PKC-α regulates cardiac contractility and calcium handling in myocytes .

  3. Therapeutic Targets:

    • Inhibitors targeting PKC-α phosphorylation are under investigation for cancer treatment .

Comparison with Polyclonal Antibodies

While recombinant monoclonal antibodies offer superior consistency, polyclonal alternatives (e.g., Boster Bio’s A00743T638) may show broader reactivity (human, mouse, rat) but lack the precision of epitope-specific binding .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

The vectors expressing anti-PRKCA antibody were constructed through a series of steps: immunizing an animal with a synthesized peptide derived from human Phospho-PRKCA (T638), isolating the positive splenocyte and extracting RNA, obtaining DNA by reverse transcription, sequencing and screening PRKCA antibody gene, and amplifying heavy and light chain sequences by PCR and cloning them into plasma vectors. Subsequently, the vector clones were transfected into mammalian cells for production. The final product is the recombinant PRKCA antibody. Recombinant PRKCA antibody in the culture medium was purified using affinity-chromatography. This antibody reacts with PRKCA protein from Human and is used in ELISA, WB, and IF applications.

The PRKCA gene is extensive, comprising 17 exons spanning 0.5Mb of genomic DNA. It encodes the PKC alpha protein, a cytoplasmic serine/threonine kinase belonging to the AGC (PKA, PKG, PKC) family. According to several studies, PRKCA exhibits the following characteristics:
PRKCA functions as a repeatedly mutated tumor gene in human cancers, potentially revealing therapeutic targets for this uncommon brain tumor. Multipoint SNP analysis indicated an association between PRKCA and its telomeric flanking regions in both populations, and combined SNP haplotype and genotype analysis revealed an allelic variant of PRKCA. PRKCA fusions are highly diagnostic for PGNT, and rare fusion partners can be identified by RNA-sequencing detection. Genomic analysis of pigment epithelial melanoma reveals recurrent alterations in PRKAR1A and PRKCA genes.

Form
Liquid
Lead Time
Typically, we can dispatch products within 1-3 business days after receiving your order. Delivery time may vary depending on the chosen purchasing method or location. Please consult your local distributor for specific delivery timelines.
Synonyms
KPCA_HUMAN antibody; PKC alpha antibody; PKC beta antibody; PKC delta antibody; PKC epsilon antibody; PKC gamma antibody; PKC zeta antibody; PKC-A antibody; PKC-alpha antibody; PKC2 antibody; PKCA antibody; PKCB antibody; PKCD antibody; PKCE antibody; PKCG antibody; PRKCA antibody; PRKCB antibody; PRKCB1 antibody; PRKCB2 antibody; PRKCD antibody; PRKCE antibody; PRKCG antibody; PRKCZ antibody; Protein kinase C alpha antibody; Protein kinase C alpha type antibody; Protein kinase C antibody; Protein kinase C beta antibody; Protein kinase C delta antibody; Protein kinase C epsilon antibody; Protein kinase C gamma antibody; Protein kinase C zeta antibody
Target Names
Uniprot No.

Target Background

Function

Calcium-activated, phospholipid- and diacylglycerol (DAG)-dependent serine/threonine-protein kinase that plays a crucial role in both positive and negative regulation of cellular processes. These processes include cell proliferation, apoptosis, differentiation, migration and adhesion, tumorigenesis, cardiac hypertrophy, angiogenesis, platelet function, and inflammation. PRKCA exerts these effects by directly phosphorylating targets such as RAF1, BCL2, CSPG4, TNNT2/CTNT, or activating signaling cascades involving MAPK1/3 (ERK1/2) and RAP1GAP.

PRKCA's involvement in cell proliferation and cell growth arrest is multifaceted. It can promote cell growth by phosphorylating and activating RAF1, which subsequently mediates the activation of the MAPK/ERK signaling cascade. Alternatively, PRKCA can upregulate CDKN1A, facilitating the formation of active cyclin-dependent kinase (CDK) complex in glioma cells. In intestinal cells stimulated by the phorbol ester PMA, PRKCA can trigger a cell cycle arrest program associated with the accumulation of the hyper-phosphorylated growth-suppressive form of RB1 and induction of the CDK inhibitors CDKN1A and CDKN1B.

PRKCA exhibits anti-apoptotic function in glioma cells, protecting them from apoptosis by suppressing the p53/TP53-mediated activation of IGFBP3. In leukemia cells, PRKCA mediates anti-apoptotic action through phosphorylation of BCL2.

During macrophage differentiation induced by macrophage colony-stimulating factor (CSF1), PRKCA translocates to the nucleus, contributing to macrophage development. Following wounding, PRKCA translocates from focal contacts to lamellipodia, participating in the modulation of desmosomal adhesion. PRKCA plays a role in cell motility by phosphorylating CSPG4, leading to the association of CSPG4 with extensive lamellipodia at the cell periphery and cell polarization accompanied by increased cell motility.

In chemokine-induced CD4(+) T cell migration, PRKCA phosphorylates CDC42-guanine exchange factor DOCK8, resulting in its dissociation from LRCH1 and activation of GTPase CDC42. PRKCA is highly expressed in numerous cancer cells where it acts as a tumor promoter and is implicated in malignant phenotypes of various tumors, including gliomas and breast cancers.

PRKCA negatively regulates myocardial contractility and positively regulates angiogenesis, platelet aggregation, and thrombus formation in arteries. It mediates hypertrophic growth of neonatal cardiomyocytes, partially through a MAPK1/3 (ERK1/2)-dependent signaling pathway. Upon PMA treatment, PRKCA is required to induce cardiomyocyte hypertrophy leading to heart failure and death by increasing protein synthesis, protein-DNA ratio, and cell surface area. PRKCA regulates cardiomyocyte function by phosphorylating cardiac troponin T (TNNT2/CTNT), which induces significant reduction in actomyosin ATPase activity, myofilament calcium sensitivity, and myocardial contractility.

In angiogenesis, PRKCA is required for complete endothelial cell migration, adhesion to vitronectin (VTN), and vascular endothelial growth factor A (VEGFA)-dependent regulation of kinase activation and vascular tube formation. PRKCA is involved in the stabilization of VEGFA mRNA at the post-transcriptional level and mediates VEGFA-induced cell proliferation.

In the regulation of calcium-induced platelet aggregation, PRKCA mediates signals from the CD36/GP4 receptor for granule release, and activates the integrin heterodimer ITGA2B-ITGB3 through the RAP1GAP pathway for adhesion.

During response to lipopolysaccharides (LPS), PRKCA may regulate selective LPS-induced macrophage functions involved in host defense and inflammation. However, in some inflammatory responses, PRKCA may negatively regulate NF-kappa-B-induced genes, through IL1A-dependent induction of NF-kappa-B inhibitor alpha (NFKBIA/IKBA).

Upon stimulation with 12-O-tetradecanoylphorbol-13-acetate (TPA), PRKCA phosphorylates EIF4G1, modulating EIF4G1 binding to MKNK1 and potentially playing a role in the regulation of EIF4E phosphorylation.

PRKCA phosphorylates KIT, leading to inhibition of KIT activity.

PRKCA phosphorylates ATF2, promoting cooperation between ATF2 and JUN, activating transcription.

PRKCA phosphorylates SOCS2 at 'Ser-52', facilitating its ubiquitination and proteosomal degradation.

Gene References Into Functions
  1. D463H mutation highly specific to chordoid glioma, enhances proliferation of astrocytes and tanycytes PMID: 29915258
  2. Modeling of the different conformations of PRKACA-DNAJB1 Chimeric Kinase revealed no obvious steric interactions of the J-domain with the rest of the RIIbeta holoenzyme. PMID: 29335433
  3. PKC activation triggers down-regulation of Kv1.3 by inducing a clathrin-mediated endocytic event that targets the channel to lysosomal-degradative compartments. PMID: 28186199
  4. Protein kinase C acts as a tumor suppressor.Cancer-associated mutations in protein kinase C are generally loss-of-function mutations.[review] PMID: 28476658
  5. results could not only better explain the role of PI-PLCbeta1/PKC-alpha signaling in erythropoiesis but also lead to a better comprehension of the lenalidomide effect on del(5q) MDS and pave the way to innovative, targeted therapies. PMID: 28970249
  6. A characteristic di-leucine motif (SVRPLL) in the C-terminal cytoplasmic region of ATP11C becomes functional upon PKCalpha activation. Moreover, endocytosis of ATP11C is induced by Ca(2+)-signaling via Gq-coupled receptors. PMID: 29123098
  7. The haplotype carrying rs9909004 influences PRKCA expression in the heart and is associated with traits linked to heart failure, potentially affecting therapy of heart failure PMID: 28120175
  8. our results demonstrate that Pc-induced expression of HO-1 is mediated by the PKCA-Nrf-2/HO-1 pathway, and inhibits UVB-induced apoptotic cell death in primary skin cells. PMID: 29470442
  9. Regulation of vascular smooth muscle cell calcification by syndecan-4/FGF-2/PKCalpha signalling and cross-talk with TGF-beta1. PMID: 29016732
  10. this study reveals a protective role for miR-706 by blocking the oxidative stress-induced activation of PKCalpha/TAOK1. Our results further identify a major implication for miR-706 in preventing hepatic fibrogenesis and suggest that miR-706 may be a suitable molecular target for anti-fibrosis therapy. PMID: 27876854
  11. We also discuss the contribution of PKC enzymes to pancreatic diseases, including insulin resistance and diabetes mellitus, as well as pancreatitis and the development and progression of pancreatic cancer PMID: 28826907
  12. data provided evidence that increased Rack1-mediated upregulation of PKC kinase activity may be responsible for the development of chemoresistance in T-ALL-derived cell line potentially by reducing FEM1b and Apaf-1 level. PMID: 27644318
  13. Regulation of insulin exocytosis by calcium-dependent protein kinase C in beta cells has been summarized. (Review) PMID: 29029784
  14. these data propose a mechanism where CD82 membrane organization regulates sustained PKCalpha signaling that results in an aggressive leukemia phenotype. These observations suggest that the CD82 scaffold may be a potential therapeutic target for attenuating aberrant signal transduction in acute myeloid leukemia (AML). PMID: 27417454
  15. MiR-3148 may play an important role in the development of CTEPH. The key mechanisms for this miRNA may be hsa-miR-3148-AR-pathways in cancer or hsa-miR-3148-PRKCA-pathways in cancer/glioma/ErbB signaling pathway. PMID: 28904974
  16. the spatial organization of cPKCs bound to the plasma membrane, is reported. PMID: 27808106
  17. PRKCA is a recurrently mutated oncogene in human chordoid glioma. PMID: 29476136
  18. Our study showed that PKCalpha modulated cell resistance to apoptosis by stimulating NF-kappaB activation and thus promoted the tumorigenesis of bladder cancer. PMID: 28629334
  19. PKCalpha translocation may occur as an early event in radiation-induced bystander responses. PMID: 27165942
  20. our study indicated that PKC alpha and beta appeared coping with oncogenic Ras or mutated Akt to maintain the balance of the homeostasis in cancer cells. Once these PKC isoforms were suppressed, the redox state in the cancer cells was disrupted, which elicited persistent oncogenic stress and subsequent apoptotic crisis. PMID: 28415683
  21. high expression of both PLCE1 and PRKCA is significantly associated with poor outcomes of the patients with esophageal cancers. PMID: 28402280
  22. In nasopharyngeal carcinoma, PKCalpha is linked to the invasion of adjacent tissues, especially in the skull base. Down-regulation of PKCalpha is a risk factor for regional lymph node metastasis. PMID: 28084179
  23. LAV-BPIFB4 isoform modulates eNOS signalling through Ca2+/PKC-alpha-dependent mechanism. PMID: 28419216
  24. Studied interactions between protein kinase C alpha (PKCalpha), FOXC2, and p120-catenin (CTNND1) in breast cancer, cell migration/ invasion; found PKCalpha acts as an upstream regulator of FOXC2, which in turn represses the expression of p120-catenin, in both in endocrine resistant ER+breast cancer and basal A triple negative breast cancer PMID: 29216867
  25. Phosphorylated PKCalpha is elevated in epidermis genetically deleted of DLX3 and the hyperproliferative response to TPA is increased, suggesting that the homeobox protein indirectly regulates the activity in the pathway, possibly through an effect on reduced phosphatase expression PMID: 28186503
  26. Results show that PKCalpha expression is under the regulation of miR-142-3p contributing to reduced osteoclasts survival. PMID: 27113904
  27. Molecular Determinants for the Binding Mode of Alkylphosphocholines in the C2 Domain of PKCalpha. PMID: 27490031
  28. Studies suggest that rare deleterious variants of PARD3 in the aPKC-binding region contribute to human cranial neural tube defect (NTD). PMID: 27925688
  29. study identified PKCalpha as hepatitis E virus HEV in host defense PMID: 28077314
  30. ADP inhibits mesothelioma cell proliferation via PKC-alpha/ERK/p53 signaling. PMID: 28777435
  31. This study provides evidences of a new PKCalpha/GAP-43 nuclear signalling pathway that controls neuronal differentiation in Human Periodontal Ligament Stem Cells. PMID: 27478064
  32. protein kinase Calpha (PKCalpha) gain of function mutations may promote synaptic defects in Alzheimer's disease PMID: 27165780
  33. Some polyphenols exert their antioxidant properties by regulating the transcription of the antioxidant enzyme genes through PKC signaling. Regulation of PKC by polyphenols is isoform dependent PMID: 27369735
  34. Data suggest that phosphorylation activity of PRKCA stems from conformational flexibility in region C-terminal to phosphorylated Ser/Thr residues; flexibility of substrate-kinase interaction enables an Arg/Lys two to three amino acids C-terminal to phosphorylated Ser/Thr to prime a catalytically active conformation, facilitating phosphoryl transfer to substrate. PMID: 28821615
  35. these results provide evidence for inherent deficits in the cystic fibrosis macrophage oxidative burst caused by decreased phosphorylation of NADPH oxidase cytosolic components that are augmented by Burkholderia PMID: 28093527
  36. The interplay between intracellular progesterone receptor and PRKCA-PRKCD plays a key role in migration and invasion of human glioblastoma cells. PMID: 27717886
  37. PRKCA SNPs are associated with neuropathic pain post total joint replacement. PMID: 28051079
  38. These findings provide the first evidence linking PKC activation to suppression of Kv7 currents, membrane depolarization, and Ca(2+) influx via L-type voltage-sensitive Ca(2+) channels as a mechanism for histamine-induced bronchoconstriction. PMID: 28283479
  39. pseudosubstrate and C1a domains, however, are minimally essential for maintaining the inactivated state. Furthermore, disrupting known interactions between the C1a and other regulatory domains releases the autoinhibited interaction and increases basal activity PMID: 28049730
  40. In polymorphism PRKCA rs9892651, HDL-C levels were lower in carriers of CC and TC genotypes that were more frequent in current-wheezers Vs TT genotype (52.2 and 52.7 Vs 55.2 mg/dl, p-value = 0.042 and p-value for trend = 0.02). PMID: 27411394
  41. Ca(2+)-PKC-MARCKS-PIP2-PI3K-PIP3 system functions as an activation module in vitro PMID: 27119641
  42. phosphorylation of TIMAP on Ser331 by PKC represents a new mechanism of endothelial barrier regulation, through the inhibition of phospho-ERM dephosphorylation PMID: 27939168
  43. PKCalpha-GSK3beta-NF-kappaB signaling pathway involvement in TRAIL-induced apoptosis PMID: 27219672
  44. Curcumin inhibited phorbol ester-induced membrane translocation of protein kinase C-epsilon (PKCepsilon) mutants, in which the epsilonC1 domain was replaced with alphaC1, but not the protein kinase C-alpha (PKCalpha) mutant in which alphaC1 was replaced with the epsilonC1 domain, suggesting that alphaC1 is a determinant for curcumin's inhibitory effect. PMID: 27776404
  45. a library of FRET sensors to monitor these transient complexes, specifically examining weak interactions between the catalytic domain of protein kinase Calpha and 14 substrate peptides. PMID: 27555323
  46. calpain and protein kinase Calpha abnormal release promotes a constitutive release of matrix metalloproteinase 9 in peripheral blood mononuclear cells from cystic fibrosis patients PMID: 27349634
  47. protein kinase C modulates alpha1B-adrenergic receptor transfer to late endosomes and that Rab9 regulates this process and participates in G protein-mediated signaling turn-off. PMID: 28082304
  48. Protein kinase C enhances the swelling-induced chloride current in human atrial myocytes. PMID: 27376808
  49. these results confirm the correlation between AXL and PKCalpha, and suggest PKCalpha-AXL signaling may be a treatment target, particularly in malignant cancer cells. PMID: 27357025
  50. After inhibition of the PKC/ERK signalling pathway, the effects of DOR on breast cancer were significantly attenuated in vivo and in vitro. In summary, DOR is highly expressed in breast cancer and is closely related to its progression. These results suggest that DOR may serve as a potential biomarker for the early diagnosis of breast cancer and may be a viable molecular target for therapeutic intervention. PMID: 27665747

Show More

Hide All

Database Links

HGNC: 9393

OMIM: 176960

KEGG: hsa:5578

STRING: 9606.ENSP00000408695

UniGene: Hs.531704

Protein Families
Protein kinase superfamily, AGC Ser/Thr protein kinase family, PKC subfamily
Subcellular Location
Cytoplasm. Cell membrane; Peripheral membrane protein. Mitochondrion membrane; Peripheral membrane protein. Nucleus.

Q&A

What is PRKCA and what is the significance of the T638 phosphorylation site?

Protein Kinase C alpha (PRKCA or PKCα) is a serine/threonine kinase that undergoes multisite phosphorylation for its regulation. The threonine residue at position 638 (T638) represents a critical carboxy-terminal phosphorylation site that plays an essential role in protein stability and function. Unlike the activation loop phosphorylation site (T497), which is required for catalytic competence, T638 phosphorylation is not essential for the catalytic activity of PRKCA per se, but instead serves to control the duration of activation by regulating the rate of dephosphorylation and inactivation of the protein . Through functional interaction with the T497 site, phosphorylation at T638 helps maintain PRKCA in an active, stable conformation. When T638 is not phosphorylated, the protein becomes significantly more susceptible to phosphatase action, leading to accelerated inactivation .

How does phospho-T638 status affect PRKCA conformation and stability?

Phosphorylation at T638 significantly impacts the structural integrity and stability of PRKCA in several measurable ways:

  • Thermal stability: Mutation studies with amino acid substitutions at the T638 site have demonstrated that phosphorylation at this position enhances thermal stability of the protein .

  • Oxidation resistance: Phosphorylated T638 confers protection against oxidative damage to the PRKCA protein .

  • Proteolytic resistance: T638 phosphorylation decreases sensitivity to trypsin digestion, indicating a more compact, protected conformation .

  • Phosphatase resistance: Most critically, T638 phosphorylation provides substantial protection against phosphatase-mediated dephosphorylation, thereby prolonging the active state of PRKCA .

Studies have established that mutation of T638 to alanine (A638) or glutamic acid (E638) results in significantly increased thermal instability and phosphatase sensitivity that mirrors the behavior of the E497 mutant protein, confirming the cooperative relationship between these phosphorylation sites .

What are the common applications for Phospho-PRKCA (T638) antibodies?

Phospho-PRKCA (T638) recombinant monoclonal antibodies are employed in multiple experimental contexts including:

  • Western blotting: For detection and quantification of phosphorylated PRKCA in cell or tissue lysates .

  • Immunohistochemistry-Paraffin (IHC-P): For visualization of phosphorylated PRKCA in fixed tissue sections, particularly valuable in studying diseased tissues versus normal controls .

  • Immunoprecipitation (IP): For isolation of phosphorylated PRKCA protein complexes to study interactions with other signaling molecules .

  • Immunocytochemistry/Immunofluorescence (ICC/IF): For subcellular localization studies of phosphorylated PRKCA, which can translocate between cytosolic and membrane fractions upon activation .

How should I prepare samples for optimal phospho-T638 detection?

Sample preparation is critical for accurate detection of phosphorylated proteins:

  • Tissue samples: For IHC-P applications, high-pressure and high-temperature antigen retrieval using sodium citrate buffer (pH 6.0) is recommended to expose the phospho-T638 epitope .

  • Cell lysates: Rapid lysis in the presence of phosphatase inhibitors (such as calyculin-A) is essential to preserve the phosphorylation status, as T638 is susceptible to dephosphorylation by phosphatases like PPP1CC2 .

  • Protein extraction: Use of a BCA Protein Assay Kit for accurate quantification of protein concentration ensures consistent loading for western blot analysis .

  • Storage considerations: Phosphorylated proteins are generally unstable; therefore, samples should be processed quickly and stored at -80°C with protease and phosphatase inhibitors to preserve phosphorylation status.

How can I validate the specificity of a Phospho-PRKCA (T638) antibody?

Comprehensive validation of phospho-specific antibodies requires multiple approaches:

  • Phosphatase treatment: Treating half of your sample with lambda phosphatase should eliminate the phospho-specific signal while maintaining total PRKCA signal.

  • Mutant controls: Expression of T638A (alanine substitution) mutants provides a negative control as this mutation prevents phosphorylation at this site .

  • Stimulation experiments: Treatments known to modulate PRKCA phosphorylation (like PMA, which initially activates PRKCA but causes downregulation with extended exposure) can demonstrate dynamic changes in T638 phosphorylation .

  • Peptide competition: Pre-incubation of the antibody with the phosphorylated peptide immunogen should block specific signals.

  • Parallel detection methods: Comparison of results using mass spectrometry-based phosphoproteomics can provide orthogonal validation of phosphorylation status .

What signaling pathways affect PRKCA T638 phosphorylation status?

The phosphorylation of PRKCA at T638 is regulated by multiple signaling pathways:

The table below summarizes key regulatory relationships:

Regulatory FactorEffect on PRKCA T638 PhosphorylationMechanism
PRKA activationIncreases phosphorylationPromotes degradation of PPP1CC2
PPP1CC2 activityDecreases phosphorylationDirect dephosphorylation
Short-term PMAIncreases phosphorylationTransient PRKCA activation
Long-term PMADecreases phosphorylationPRKCA degradation
T497 phosphorylationStabilizes T638 phosphorylationCooperative interaction

How does the functional interaction between T638 and T497 phosphorylation affect experimental design?

The cooperative relationship between T638 and T497 phosphorylation sites has significant implications for experimental design:

  • Dual phosphorylation assessment: Experiments should ideally examine both T638 and T497 phosphorylation states simultaneously, as the phosphorylation status of one site influences the other .

  • Mutation studies: When designing PRKCA mutants for functional studies, researchers should consider creating both single site mutants (T638A or T497E) and double mutants to fully assess their cooperative effects .

  • Thermal stability assays: Given that mutation at either T638 or T497 results in thermal instability, thermal shift assays can provide a functional readout of the cooperative phosphorylation effects .

  • Phosphatase sensitivity: Experimental designs should account for the increased phosphatase sensitivity of PRKCA when either T638 or T497 is not phosphorylated, potentially requiring stronger phosphatase inhibition during sample preparation .

  • Kinetics considerations: The temporal dynamics of T638 and T497 phosphorylation may differ, necessitating time-course experiments to fully characterize PRKCA regulation in your system .

What techniques can be used to study phospho-PRKCA (T638) in disease models?

Multiple complementary techniques can be employed to investigate phospho-PRKCA (T638) in disease contexts:

  • Phosphoproteomics: Advanced mass spectrometry approaches using Superbinder resin for phosphorylated tyrosine (pY) enrichment followed by TiO₂ column for phosphorylated serine/threonine (pS/pT) enrichment can identify disease-specific phosphorylation patterns .

  • Parallel Reaction Monitoring (PRM): This targeted mass spectrometry approach allows quantitative analysis of specific phosphopeptides across multiple samples .

  • Immunohistochemistry: Analysis of paraffin-embedded diseased tissues using phospho-specific antibodies can reveal altered localization and expression of phospho-PRKCA (T638) .

  • Functional assays: Assessment of downstream signaling events affected by PRKCA activation, such as cell proliferation (using MTT assays) or apoptosis (using Annexin V staining), can connect phosphorylation status to disease phenotypes .

  • Xenograft models: In vivo assessment of PRKCA phosphorylation mutants in tumor progression models can establish causative relationships .

Why might I observe variability in phospho-T638 detection across experiments?

Variability in phospho-T638 detection can stem from several sources:

  • Rapid dephosphorylation: The T638 site is particularly susceptible to phosphatase activity when the T497 site is not phosphorylated, making proper sample handling with phosphatase inhibitors critical .

  • Conformation-dependent epitope accessibility: The phospho-T638 epitope may be partially masked in certain conformations of PRKCA, affecting antibody recognition.

  • Context-dependent phosphorylation: The activation state of PRKCA varies with cellular conditions and signaling inputs, leading to natural biological variation in T638 phosphorylation .

  • Antibody specificity: Some phospho-specific antibodies may cross-react with similar phosphorylation motifs in related proteins or different phosphorylation sites within PRKCA itself.

  • Degradation kinetics: During certain cellular processes, PRKCA undergoes regulated degradation that may vary across experimental conditions, affecting total and phosphorylated protein levels .

How can I optimize immunohistochemistry protocols for phospho-PRKCA (T638) detection?

Optimizing IHC-P for phospho-PRKCA (T638) requires attention to several critical parameters:

  • Antigen retrieval: High-pressure and high-temperature sodium citrate buffer (pH 6.0) treatment is essential for exposing the phospho-epitope in formalin-fixed, paraffin-embedded tissues .

  • Blocking conditions: Thorough blocking with phosphate-free blocking solutions prevents non-specific binding while preserving phospho-epitopes.

  • Antibody concentration: Titration experiments to determine optimal primary antibody concentration are crucial, as excessive antibody can increase background while insufficient antibody reduces sensitivity.

  • Incubation conditions: Extended incubation times (overnight at 4°C) often provide better signal-to-noise ratios for phospho-specific antibodies.

  • Detection systems: Enhanced detection systems, such as polymer-based secondary antibodies or tyramide signal amplification, can improve detection sensitivity for low-abundance phosphorylated proteins.

What are the best approaches for quantifying changes in PRKCA T638 phosphorylation?

Quantitative assessment of PRKCA T638 phosphorylation requires rigorous methodology:

  • Normalization strategy: Always normalize phospho-PRKCA signal to total PRKCA to account for changes in protein expression versus phosphorylation state.

  • Loading controls: Include appropriate loading controls (β-actin, GAPDH) to normalize for total protein content across samples.

  • Phosphorylation standards: Where possible, include positive control samples with known phosphorylation status to calibrate quantification.

  • Image analysis: For immunoblotting, use linear range capture and analysis software that can accurately quantify band intensity without saturation.

  • Multiple detection methods: Confirm findings using complementary techniques such as ELISA, mass spectrometry, and immunocytochemistry to validate quantitative changes .

How might phospho-PRKCA (T638) antibodies be used to study kinase-phosphatase dynamics?

Investigating kinase-phosphatase dynamics using phospho-PRKCA (T638) antibodies can provide insights into regulatory mechanisms:

  • Temporal analysis: Time-course experiments following stimulation can reveal the dynamics of phosphorylation and subsequent dephosphorylation events .

  • Phosphatase inhibition studies: Selective inhibitors like calyculin-A can block phosphatase activity, allowing assessment of the baseline phosphorylation/dephosphorylation balance .

  • Co-immunoprecipitation: Using phospho-PRKCA (T638) antibodies for IP followed by detection of associated phosphatases can identify specific regulatory interactions, such as the PRKCA-PPP1CC2 complex .

  • Subcellular fractionation: Examining the distribution of phospho-PRKCA between cytosolic and membrane fractions can reveal compartment-specific regulation .

  • Pharmacological manipulation: Using activators like PMA that cause initial activation followed by downregulation can reveal the coordinated dynamics of phosphorylation, dephosphorylation, and degradation .

What role does phospho-PRKCA (T638) play in disease pathways?

While the specific role of PRKCA T638 phosphorylation in diseases requires further investigation, several insights can be derived from existing research:

  • Cancer signaling: Altered PRKC signaling plays crucial roles in various cancers, including breast cancer, where phosphoproteomic profiling has revealed aberrantly activated kinase pathways .

  • Cell proliferation and apoptosis: Phosphorylation status of PRKCA affects downstream pathways that regulate cell proliferation and apoptosis, processes central to cancer development .

  • Protein stability regulation: The T638 phosphorylation site controls protein stability and degradation kinetics, potentially affecting signaling duration in disease states .

  • Interaction with other pathways: PRKCA signaling intersects with other pathways like PI3K, which is frequently dysregulated in diseases, suggesting potential cooperative effects .

  • Therapeutic targeting: Understanding the phosphorylation status of PRKCA could inform therapeutic strategies targeting protein kinase activity in various diseases.

How can recombinant monoclonal antibodies improve reproducibility in phospho-PRKCA research?

Recombinant monoclonal antibodies offer several advantages for phospho-PRKCA research:

  • Increased sensitivity: Recombinant antibodies typically demonstrate higher sensitivity for their target epitopes compared to traditional antibodies .

  • Confirmed specificity: The defined amino acid sequence and production process of recombinant antibodies ensures consistent epitope recognition .

  • Excellent batch-to-batch consistency: Unlike traditional hybridoma-derived antibodies, recombinant antibodies show minimal variation between production lots, enhancing experimental reproducibility .

  • Sustainable supply: Once the antibody sequence is established, production can be maintained indefinitely without concerns about hybridoma stability or animal source limitations .

  • Animal-free production: Modern recombinant antibodies can be produced using animal-free systems, addressing ethical considerations while maintaining performance characteristics .

What are emerging technologies for studying PRKCA phosphorylation dynamics?

Cutting-edge approaches for investigating PRKCA phosphorylation include:

  • Phosphoproteomics with Superbinder technology: Enhanced enrichment methods using engineered SH2 domains (Superbinder resin) provide improved detection of low-abundance phosphoproteins in complex samples .

  • Parallel Reaction Monitoring (PRM): This targeted mass spectrometry approach allows precise quantification of specific phosphopeptides, enabling detailed kinetic studies of phosphorylation events .

  • Phospho-specific biosensors: Genetically encoded FRET-based sensors designed to detect specific phosphorylation events can provide real-time, single-cell resolution of PRKCA phosphorylation dynamics.

  • Proximity ligation assays: These techniques can detect interactions between phosphorylated PRKCA and its binding partners with subcellular resolution in fixed samples.

  • Phospho-proteomic network analysis: Computational approaches integrating multiple phosphorylation events can reveal coordinated signaling networks involving PRKCA .

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 2024 Thebiotek. All Rights Reserved.