PKN3 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

Definition and Core Characteristics

PKN3 Antibody, FITC conjugated is a rabbit polyclonal antibody chemically linked to fluorescein isothiocyanate (FITC), enabling fluorescence-based detection of PKN3. This kinase is implicated in prostate cancer invasiveness and cellular signaling pathways .

ParameterSpecification
TargetSerine/threonine-protein kinase N3 (UniProt ID: Q6P5Z2)
Host SpeciesRabbit
ConjugationFITC (Ex/Em: 495/519 nm)
ReactivityHuman ; predicted cross-reactivity with pig, bovine, horse, and dog
ApplicationsWestern blot (WB), immunofluorescence (IF), intracellular flow cytometry
Storage-20°C in 50% glycerol, 0.01 M PBS, and 0.03% Proclin-300
Purity>95% (Protein G purified)

Immunogen Design

  • Derived from recombinant human PKN3 protein (amino acids 1–252) .

  • Recognizes epitopes in the N-terminal regulatory domain, critical for kinase activity modulation .

Biological Relevance

  • Role in Cancer: PKN3 promotes invasiveness in malignant prostate cancer cells (e.g., LNCaP and PC-3 lines) .

  • Apoptosis Link: Caspase-3 cleaves PKN during apoptosis, generating constitutively active fragments (e.g., AF3) .

Western Blotting

  • Detects PKN3 at ~99–112 kDa in human prostate adenocarcinoma (PC-3) lysates .

  • Validated in mouse and rat spleen lysates, demonstrating cross-species reactivity .

Flow Cytometry

  • Used for intracellular staining in LNCaP cells, showing PKN3 localization in the perinuclear region .

  • Example protocol:

    1. Fix cells with 4% paraformaldehyde .

    2. Incubate with antibody (1:300 dilution) .

    3. Detect using anti-rabbit IgG secondary antibodies (e.g., Alexa Fluor® 488) .

Immunofluorescence

  • Localizes PKN3 to the nucleus and Golgi apparatus in fixed cell lines .

Validation and Quality Control

  • Specificity: No cross-reactivity with PKN1 or PKN2 isoforms .

  • Performance:

    • Signal-to-Noise Ratio: >10:1 in flow cytometry (vs. isotype controls) .

    • Lot Consistency: Batch-specific validation data provided by manufacturers .

Limitations and Considerations

  • Species Restrictions: Limited reactivity in non-mammalian systems (e.g., zebrafish, Xenopus) .

  • Phosphorylation Sensitivity: PKN3’s phosphorylation state affects antibody binding (e.g., phosphatase treatment alters migration patterns) .

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 order within 1-3 business days of receiving it. Delivery time may vary depending on the method of purchase and the location. Please consult your local distributor for specific delivery times.
Synonyms
pkn3 antibody; PKN3_HUMAN antibody; PKNBETA antibody; Protein kinase C related kinase 3 antibody; Protein kinase N3 antibody; Protein kinase PKN beta antibody; Protein kinase PKN-beta antibody; Protein-kinase C-related kinase 3 antibody; RP11-545E17.1 antibody; Serine/threonine protein kinase N3 antibody; Serine/threonine-protein kinase N3 antibody
Target Names
Uniprot No.

Target Background

Function
PKN3 antibody, FITC conjugated, plays a crucial role in promoting invasiveness in malignant prostate cancer.
Gene References Into Functions
  1. Kinetic analysis of PKN1-3 demonstrates a sequential ordered Bi-Bi kinetic mechanism where peptide substrate binding occurs before ATP binding. This mechanism is supported by additional kinetic studies regarding product inhibition and the affinity of small molecule inhibitors. PMID: 27919031
  2. PKN3 is the primary regulator of angiogenesis in both humans and mice, as well as tumor metastasis in mice. PMID: 26742562
  3. PKN3 is a newly identified protein involved in remodeling the actin-adherens junction, potentially by linking ICAM-1 signaling with actin/AJ dynamics. PMID: 22609186
  4. PKN isoforms are not simply redundant in supporting migration, but rather appear to be connected through isoform-specific regulatory domain properties to selective upstream signals. PMID: 21754995
  5. PKN3 could serve as a promising target for therapeutic interventions in cancers lacking the tumor suppressor PTEN function or relying on chronic activation of phosphoinositide 3-kinase. PMID: 15282551

Show More

Hide All

Database Links

HGNC: 17999

OMIM: 610714

KEGG: hsa:29941

STRING: 9606.ENSP00000291906

UniGene: Hs.300485

Protein Families
Protein kinase superfamily, AGC Ser/Thr protein kinase family, PKC subfamily
Subcellular Location
Nucleus. Cytoplasm, perinuclear region. Note=Nuclear and perinuclear Golgi region.
Tissue Specificity
Expressed in prostate tumors and various cancer cell lines. Not expressed in adult tissues.

Q&A

What is PKN3 and why is it an important research target?

PKN3 (Serine/threonine-protein kinase N3) functions as a protein kinase C-related molecule that mediates malignant cell growth downstream of activated PI3K pathway. It is particularly significant because it is required for invasive prostate cell growth as demonstrated in 3D cell culture assays and orthotopic mouse tumor models . PKN3 is regulated by PI3K at both expression and catalytic activity levels, making it a potential therapeutic target in cancers lacking tumor suppressor PTEN function or those dependent on chronic PI3K activation . As a member of the AGC family of protein kinases, understanding PKN3's role provides insight into fundamental cellular signaling mechanisms controlling proliferation and invasion.

What are the optimal storage conditions for PKN3 antibody with FITC conjugation to maintain sensitivity?

PKN3 antibodies with FITC conjugation should be stored at -20°C or -80°C upon receipt . For long-term storage, -20°C is recommended while avoiding repeated freeze-thaw cycles . When storing as a concentrated solution, brief centrifugation prior to opening the vial is advised to ensure collection of all material . The storage buffer typically consists of preservatives (e.g., 0.03% Proclin 300) and stabilizers (e.g., 50% Glycerol, 0.01M PBS, pH 7.4) . For proper aliquoting to prevent freeze-thaw damage, divide the antibody into multiple small volumes based on typical experimental usage—this preserves the FITC fluorescence intensity which can diminish with repeated temperature fluctuations.

How can I verify the specificity of a PKN3 antibody in my experimental system?

Verifying antibody specificity requires multiple complementary approaches. First, utilize positive controls known to express PKN3, such as HeLa cell lysate as recommended for commercial PKN3 antibodies . Implement a western blot validation using recombinant PKN3 protein alongside tissue/cell samples to confirm the antibody detects a band of the expected molecular weight (approximately 100 kDa for full-length PKN3). For FITC-conjugated antibodies, perform fluorescence microscopy on cells with confirmed PKN3 expression versus those with PKN3 knockdown via siRNA or CRISPR to demonstrate specific staining patterns.

For additional validation, conduct peptide competition assays using the specific immunogen peptide—in the case of some commercial PKN3 antibodies, this would be the recombinant human serine/threonine-protein kinase N3 protein (amino acids 1-252) . The peptide should block antibody binding if the antibody is specific. Cross-referencing your staining pattern with published subcellular localization data (cytoplasm and cell membrane for PKN3) provides further confirmation of specificity.

What are the optimal experimental conditions for using PKN3 antibody in fluorescence-based applications?

For optimal fluorescence-based applications, consider the following methodology:

First, for immunofluorescence staining of cells or tissues, fix samples using 4% paraformaldehyde (10-15 minutes at room temperature) to preserve cellular architecture while maintaining antigen accessibility. For PKN3 detection, permeabilize with 0.1-0.3% Triton X-100 since PKN3 has both cytoplasmic and membrane localization . The FITC-conjugated PKN3 antibody should be diluted in blocking buffer (typically 1-5% BSA in PBS) at concentrations determined through titration experiments (starting with manufacturer recommendations, usually 1-10 μg/ml).

When designing fluorescence microscopy experiments, account for FITC's excitation maximum at approximately 495 nm and emission maximum at about 519 nm. Be aware of potential photobleaching—minimize exposure to light and consider anti-fade mounting media for preserved signal. For multi-color imaging, select far-separated fluorophores to avoid spectral overlap with FITC (avoid PE or GFP which have similar emission spectra).

For flow cytometry applications, optimal cell density is typically 1×10^6 cells/ml with antibody concentrations titrated to maximize the separation index between positive and negative populations while minimizing background. Include appropriate single-color controls and fluorescence-minus-one (FMO) samples for accurate compensation and gating.

How can I design experiments to investigate PKN3 phosphorylation status using the phospho-specific antibodies?

Designing experiments to investigate PKN3 phosphorylation requires careful consideration of activation conditions and controls. Begin by treating cells with PI3K pathway activators, as PKN3 is regulated downstream of PI3K signaling . Include both positive controls (cells with known PKN3 activation) and negative controls (PKN3 knockdown cells or cells treated with PI3K inhibitors).

For western blot analysis of phosphorylation status, lysate preparation must include phosphatase inhibitors (e.g., sodium orthovanadate, sodium fluoride, and β-glycerophosphate) to preserve phosphorylation states. Run parallel gels to probe with both phospho-specific PKN3 antibodies and total PKN3 antibodies to calculate phosphorylation/total protein ratios.

When using FITC-conjugated phospho-specific antibodies (such as those targeting specific residues like Tyr9) , implement a phosphatase treatment control group to confirm the specificity of phosphorylation detection. This is particularly important as PKN3 fragments can show multiple bands representing different phosphorylation states, as demonstrated for related PKN family proteins .

For quantitative analysis, consider implementing a NanoBRET assay as described for related kinases . This technique allows for real-time monitoring of PKN3 activity in living cells by measuring bioluminescence resonance energy transfer between a NanoLuciferase-tagged PKN3 and an ATP-competitive fluorescent tracer. This approach has been validated to correlate with enzyme inhibition potency in cellular contexts .

What methods can be used to validate PKN3 antibody results across different experimental techniques?

Validating PKN3 antibody results across techniques requires a multi-platform approach:

First, implement orthogonal detection methods—if using the FITC-conjugated antibody for immunofluorescence, confirm findings with western blot using the same antibody or an unconjugated version. Compare subcellular localization observed in microscopy with fractionation studies and subsequent western blotting.

For functional validation, correlate antibody-based protein detection with mRNA expression using RT-qPCR. Additionally, implement genetic approaches by creating PKN3 knockdown or knockout models using siRNA, shRNA, or CRISPR-Cas9 systems to confirm signal specificity. For instance, research has demonstrated that PKN3 is required for invasive prostate cell growth through inducible expression of short hairpin RNA (shRNA) .

When examining PKN3 activity, especially in cancer contexts, correlate antibody results with functional assays such as 3D cell culture invasion assays that have been established as readouts for PKN3 activity . This approach helps validate whether the detected protein correlates with expected biological function.

Finally, for comprehensive validation, implement proximity ligation assays (PLA) to confirm protein-protein interactions suggested by co-localization studies, particularly examining PKN3 interactions with components of the PI3K pathway or targeted substrates.

What are common causes of weak signal when using FITC-conjugated PKN3 antibodies and how can they be addressed?

Weak fluorescence signals when using FITC-conjugated PKN3 antibodies may stem from multiple causes requiring systematic troubleshooting:

Antibody integrity issues often cause signal problems. FITC is sensitive to light exposure and pH fluctuations, leading to photobleaching and diminished fluorescence. Store the antibody in dark containers at recommended temperatures (-20°C or -80°C) , and prepare fresh working dilutions for each experiment. Avoid repeated freeze-thaw cycles by aliquoting the stock solution upon receipt.

Fixation protocols significantly impact epitope availability. If using paraformaldehyde fixation, limit exposure to 10-15 minutes as over-fixation can mask epitopes. Test alternative fixation methods like methanol or acetone for cytoplasmic and membrane proteins like PKN3 . For antigen retrieval in tissue sections, optimize heat-induced epitope retrieval (HIER) conditions using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0).

Permeabilization efficiency affects antibody accessibility to intracellular targets. Increase Triton X-100 concentration incrementally (0.1% to 0.5%) or try saponin (0.1-0.5%) which creates smaller pores and may better preserve membrane-associated PKN3. For flow cytometry applications, test commercial permeabilization kits specifically designed for intracellular kinases.

Signal amplification techniques can overcome weak signals: implement tyramide signal amplification (TSA), use anti-FITC secondary antibodies conjugated to brighter fluorophores like Alexa Fluor 488, or consider using a biotin-streptavidin system for signal enhancement.

How can I optimize blocking conditions to reduce background when using PKN3 antibody in immunofluorescence?

Optimizing blocking conditions requires systematic evaluation of blocking agents and protocols:

Start by identifying the source of background. Autofluorescence can be assessed by examining unstained samples; non-specific antibody binding requires testing with isotype controls matching the PKN3 antibody's host species (typically rabbit IgG for PKN3 polyclonal antibodies) .

For blocking optimization, test different blocking solutions systematically:

  • BSA at varying concentrations (1%, 3%, and 5%) in PBS

  • Normal serum (5-10%) from the species in which the secondary antibody was raised

  • Commercial blocking reagents specifically designed for fluorescence applications

  • Combinations such as 2% BSA with 5% normal serum and 0.1% Tween-20

Blocking duration and temperature significantly impact efficacy. Extend blocking time from 30 minutes to 1-2 hours at room temperature, or overnight at 4°C for tissues with high background. Additionally, include 0.1-0.3% Triton X-100 in the blocking solution to reduce hydrophobic interactions contributing to background.

For samples with persistent background, pre-incubate with unconjugated Fab fragments matching the host species of your primary antibody. Add 0.1% Tween-20 to all washing and incubation buffers to reduce non-specific hydrophobic interactions, and increase washing duration and frequency (5-6 washes of 5-10 minutes each).

What strategies can resolve issues with non-specific binding of PKN3 antibody in western blotting and immunofluorescence?

Resolving non-specific binding requires a multi-faceted approach targeting each potential source:

For western blotting, implement gradient gel electrophoresis (4-20%) to improve separation of PKN3 (approximately 100 kDa) from potentially cross-reactive proteins. Increase the stringency of blocking with 5% non-fat dry milk or 5% BSA in TBST, and extend blocking time to 2 hours at room temperature or overnight at 4°C. Test different detergents in wash buffers, comparing 0.1% Tween-20 versus 0.1% Triton X-100 for reduced background.

Antibody validation is crucial for specificity. Conduct peptide competition assays using the specific immunogen (recombinant human PKN3 protein amino acids 1-252) to confirm binding specificity. Implement additional controls including the target protein in knockout/knockdown samples and recombinant proteins as positive controls.

For immunofluorescence, dilution optimization is key. Titrate the FITC-conjugated PKN3 antibody beginning at the manufacturer's recommended dilution, then test 2-fold and 5-fold more dilute solutions to find the optimal signal-to-noise ratio. Reduce primary antibody incubation time if non-specific binding persists.

Cross-adsorbed antibodies can improve specificity. If available, use PKN3 antibodies that have been cross-adsorbed against related kinases, particularly other PKN family members (PKN1, PKN2) which share structural homology with PKN3.

How can PKN3 antibody be used to investigate protein-protein interactions in the PI3K signaling pathway?

Investigating PKN3 protein-protein interactions requires sophisticated methodological approaches:

Implement co-immunoprecipitation (Co-IP) using FITC-conjugated PKN3 antibodies with anti-FITC beads for pull-down, followed by immunoblotting for potential interaction partners within the PI3K pathway. This approach is particularly valuable for examining PKN3's interactions with upstream regulators and downstream effectors in the PI3K signaling cascade .

Proximity ligation assay (PLA) offers superior sensitivity for detecting protein interactions. Combine anti-PKN3 antibody with antibodies against suspected interaction partners (e.g., components of the PI3K pathway), followed by species-specific secondary antibodies linked to complementary oligonucleotides. When proteins interact (<40 nm apart), these oligonucleotides enable rolling circle amplification and fluorescent probe binding, visualizing interactions as distinct puncta.

For live-cell dynamics, implement Förster resonance energy transfer (FRET) by co-expressing PKN3 fused to a donor fluorophore (e.g., CFP) and potential interaction partners fused to an acceptor fluorophore (e.g., YFP). Alternatively, adapt the NanoBRET approach described for related kinases , using NanoLuciferase-tagged PKN3 and fluorophore-tagged interaction partners.

Fluorescence recovery after photobleaching (FRAP) with FITC-conjugated PKN3 antibody can assess how protein complexes affect PKN3 mobility within cellular compartments. This is particularly relevant given PKN3's role in cancer cell migration and invasion downstream of PI3K activation .

How can researchers use PKN3 antibodies to investigate its role in cancer progression and therapeutic resistance?

Investigating PKN3's role in cancer progression requires integration of cellular, molecular, and in vivo approaches:

For cellular models, implement stable expression of fluorescently-tagged PKN3 in combination with FITC-conjugated antibodies against phosphorylated forms to visualize both total and activated PKN3 in real-time during cancer cell invasion assays. This approach can be extended to patient-derived organoids to assess PKN3 expression and activation in clinically relevant models.

Therapeutic resistance studies should examine PKN3 expression and activation before and after treatment with targeted therapies, particularly PI3K pathway inhibitors. Use FITC-conjugated PKN3 antibodies for flow cytometry to quantify changes in PKN3 expression at the single-cell level within heterogeneous tumor populations. This can identify resistant subpopulations where PKN3 remains active despite therapy.

For in vivo studies, develop xenograft models using cells with manipulated PKN3 expression (overexpression, knockdown via shRNA) . Harvest tissues for immunohistochemical analysis using PKN3 antibodies to correlate PKN3 expression with invasion patterns, metastatic potential, and treatment response.

Pharmacological studies can employ small molecule PKN3 inhibitors such as those identified in screening campaigns (e.g., 4-anilino-quin(az)oline derivatives) alongside FITC-conjugated PKN3 antibodies to monitor target engagement and downstream effects. The NanoBRET assay described for related kinases offers a valuable method to assess inhibitor efficacy in living cells .

What methodologies can be used to study PKN3 activation dynamics using phospho-specific antibodies in response to different stimuli?

Studying PKN3 activation dynamics requires temporal resolution and quantitative approaches:

Time-course experiments should examine PKN3 phosphorylation following stimulation with growth factors or inhibition of phosphatases. Collect samples at multiple timepoints (0, 5, 15, 30, 60, 120 minutes) post-stimulation and analyze using western blotting with phospho-specific antibodies. Compare these phosphorylation patterns to known PKN cleavage patterns during cellular processes like apoptosis, where PKN family proteins generate constitutively active fragments .

For single-cell resolution, implement high-content imaging using FITC-conjugated phospho-specific PKN3 antibodies. This allows quantification of phosphorylation intensity, subcellular localization changes, and population heterogeneity in response to stimuli. Complement this with flow cytometry to quantify phosphorylation levels across large cell populations.

Real-time measurements of PKN3 activity can be achieved using genetically encoded FRET-based biosensors. Design these with PKN3 substrate sequences flanked by fluorescent proteins that undergo FRET changes upon phosphorylation. This approach permits continuous monitoring of PKN3 activity in living cells following various stimuli.

For in-depth mechanistic studies, combine phospho-specific antibodies with phosphatase treatment controls and kinase inhibitors targeting the PI3K pathway. This helps distinguish between direct phosphorylation events and downstream consequences. As demonstrated with other PKN family members, proteolytic cleavage by caspases can generate constitutively active kinase fragments , suggesting that PKN3 activation dynamics may involve both phosphorylation and proteolytic processing.

How should researchers quantify and normalize fluorescence data from experiments using FITC-conjugated PKN3 antibodies?

Quantifying and normalizing fluorescence data requires rigorous methodological approaches:

For microscopy data, implement intensity-based quantification using open-source software like ImageJ/FIJI or CellProfiler. Define regions of interest (ROIs) based on cellular compartments where PKN3 localizes (cytoplasm, cell membrane) . Calculate the mean fluorescence intensity (MFI) within these ROIs across multiple cells (n>30) per condition. For normalization, use one of several approaches:

  • Background subtraction: Measure and subtract the MFI from regions without cells

  • Internal reference: Normalize to a housekeeping protein labeled with a spectrally distinct fluorophore

  • Total protein normalization: Use total protein stains like SYPRO Ruby or Coomassie prior to immunostaining

For flow cytometry, quantify FITC signal using median fluorescence intensity (MFI) rather than mean, as it is less sensitive to outliers. Normalize using:

  • Fluorescence-minus-one (FMO) controls to account for spectral overlap

  • Calibration beads with known fluorophore molecules to convert arbitrary units to molecules of equivalent soluble fluorochrome (MESF)

  • Isotype controls to establish baseline non-specific binding

Statistical analysis should include tests appropriate for the experimental design (t-test, ANOVA with post-hoc tests) and effect size calculations. For time-course experiments, consider area-under-curve (AUC) analysis to capture the integrated response over time.

What are the key considerations when interpreting results from experiments investigating PKN3 expression and activity in different cancer models?

Interpreting PKN3 data in cancer models requires careful consideration of multiple factors:

Baseline expression variability must be established across normal tissues and cancer subtypes. PKN3 is regulated by the PI3K pathway , and its expression may correlate with PI3K activity and PTEN status. Therefore, characterize the genetic background of your cancer models, particularly PI3K pathway alterations and PTEN status, before interpreting PKN3 expression data.

Cell type heterogeneity within tumors significantly impacts interpretation. PKN3 may be differentially expressed or activated in specific subpopulations like cancer stem cells or invasive front cells. Use multiparameter approaches combining FITC-conjugated PKN3 antibodies with markers for specific cell populations to resolve this heterogeneity.

Activation state versus expression level distinction is crucial. High PKN3 expression doesn't necessarily indicate high activity. Combine total PKN3 detection with phospho-specific antibodies targeting activation-associated residues to distinguish between inactive and active PKN3 pools. Consider the constitutive activity demonstrated for cleaved fragments of PKN family proteins when interpreting activity data.

Functional correlation validation is essential. Correlate PKN3 expression/activation data with functional assays relevant to its known roles in invasion and malignant growth . These include 3D invasion assays, anchorage-independent growth, and in vivo metastasis models. This functional validation strengthens the biological significance of observed PKN3 patterns.

How can researchers distinguish between specific signal and artifacts when analyzing complex tissues with PKN3 antibodies?

Distinguishing specific signal from artifacts in complex tissues requires multiple validation strategies:

Implement rigorous controls including isotype controls matching the PKN3 antibody's host species and concentration, tissues known to be negative for PKN3 expression, and competitive blocking with the immunizing peptide . For FITC-conjugated antibodies, include unstained tissue sections to assess autofluorescence, particularly in tissues rich in collagen, elastin, or lipofuscin.

Multiple detection methods provide orthogonal validation. Compare immunofluorescence results with chromogenic IHC, in situ hybridization for PKN3 mRNA, and western blotting of tissue lysates. Concordance across these methods strengthens confidence in specificity.

Antibody validation through genetic approaches offers definitive evidence of specificity. Use tissues from PKN3 knockout models or those with CRISPR-mediated epitope deletion as negative controls. For human tissues where genetic models aren't available, compare with tissues where PKN3 expression has been characterized by orthogonal methods.

Advanced digital pathology techniques can help distinguish signal from noise. Implement spectral unmixing to separate FITC signal from autofluorescence, particularly in tissues with high background. Apply automated analysis algorithms that can identify subcellular localization patterns consistent with PKN3 biology (cytoplasmic and membrane localization) and exclude non-specific staining patterns.

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.