RPS6KA1/RPS6KA3/RPS6KA2/RPS6KA6 Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Characteristics

The antibody is generated in rabbits using synthesized peptides derived from conserved regions of the RSK family. Key features include:

PropertyDetails
Host SpeciesRabbit
ClonalityPolyclonal
ReactivityHuman, Mouse, Rat
Tested ApplicationsWestern Blot (WB), Immunohistochemistry (IHC), Immunofluorescence (IF), ELISA
Recommended DilutionsWB: 1:500–1:2000; IHC: 1:50–1:200; IF: 1:50–1:200
ImmunogenPeptide sequences around non-phosphorylation sites (e.g., S221/227/S218/232)
StoragePBS with 50% glycerol, 0.5% BSA, and 0.02% sodium azide at -20°C

The antibody’s specificity for multiple RSK isoforms enables broad utility in studying overlapping and distinct roles of these kinases in signaling pathways .

Target Proteins: Structure and Function

The RSK family (RPS6KA1–4) comprises serine/threonine kinases with two catalytic domains. Key functional insights include:

  • Activation Mechanism: Phosphorylated by ERK1/2 in response to growth factors, hormones, or stress signals .

  • Substrates: Regulate transcription factors (e.g., CREB1, NFκB), translation machinery (e.g., RPS6, EIF4B), and apoptosis mediators (e.g., BAD) .

  • Cellular Roles:

    • RPS6KA1 (RSK1): Linked to cancer progression, inflammation, and drug resistance in acute myeloid leukemia (AML) .

    • RPS6KA3 (RSK2): Mutations cause Coffin-Lowry syndrome; implicated in cognitive function .

    • RPS6KA2 (RSK3): Associated with ovarian cancer suppression via p53 activation .

    • RPS6KA6 (RSK4): Low expression in healthy tissues but elevated in certain cancers .

3.1. Cancer Biology

  • Myeloproliferative Neoplasms (MPN) and AML:

    • RSK1 inhibition (via shRNA or small molecules like PMD-026) reduces colony formation in AML cells and dampens pro-inflammatory cytokine production (e.g., TNF, IL-6) .

    • RSK1 overexpression correlates with venetoclax/azacitidine resistance in AML; combinatorial inhibition restores drug sensitivity .

  • Ovarian Cancer: RPS6KA2 acts as a tumor suppressor by activating p53-dependent apoptosis pathways .

3.2. Signaling Pathway Analysis

  • The antibody facilitates studies on RSK-mediated phosphorylation events, such as NFκB activation in monocytes and mTORC1 regulation .

  • Identified substrates include RanBP3 (nuclear transport), PDCD4 (translation suppression), and TRIM33 (transcriptional regulation) .

Therapeutic Implications

  • RSK1 Inhibition: Reduces leukemic cell proliferation and inflammatory cytokine secretion in MPN/AML models .

  • Resistance Reversal: Pharmacological RSK1 inhibition restores venetoclax/azacitidine efficacy in resistant AML cells .

  • Inflammatory Diseases: Targeting RSK1 in monocytes suppresses TNF/NFκB pathways, suggesting utility in inflammation-driven pathologies .

Future Directions

  • Isoform-Specific Functions: Further studies are needed to dissect unique roles of RSK1–4 in tissue-specific contexts .

  • Clinical Translation: Develop isoform-selective inhibitors to minimize off-target effects in cancer therapy .

Product Specs

Buffer
The antibody is provided as a liquid solution in phosphate-buffered saline (PBS) containing 50% glycerol, 0.5% bovine serum albumin (BSA), and 0.02% sodium azide.
Form
Liquid
Lead Time
We typically ship orders within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. For specific delivery estimates, please contact your local distributor.
Target Names
RPS6KA1/RPS6KA3/RPS6KA2/RPS6KA6
Uniprot No.

Q&A

What are the key differences among RPS6KA family members and how does this impact antibody selection?

The RPS6KA family consists of four main members with distinct functions and tissue distributions:

  • RPS6KA1 (RSK1): Mediates mitogenic and stress-induced activation of transcription factors CREB1, ETV1/ER81, and NR4A1/NUR77

  • RPS6KA3 (RSK2): Associated with Coffin-Lowry Syndrome, important for learning-related synaptic plasticity

  • RPS6KA2 (RSK3): Shares structural similarities with other family members but has distinct regulatory functions

  • RPS6KA6 (RSK4): Has unique expression patterns compared to other RSK proteins

When selecting antibodies, researchers should consider:

  • Specific phosphorylation sites (e.g., Ser221/227/S218/232) that may indicate activation status

  • Cross-reactivity between family members

  • Requirements for specific isoform detection or pan-RSK detection

Research indicates that despite high sequence homology, these proteins have distinct functions, which necessitates careful antibody selection for accurate experimental outcomes .

How can phospho-specific RPS6KA antibodies be validated to ensure specificity and accuracy in experimental protocols?

Comprehensive validation of phospho-specific RPS6KA antibodies should include:

  • Phosphatase treatment controls: Lambda phosphatase-treated samples should show reduced signal compared to untreated samples

  • Stimulation-dependent phosphorylation: Treatment with activators (e.g., EGF, PMA) should increase phospho-specific signals

  • Peptide competition assays: Using synthetic phosphopeptides and non-phosphopeptides to confirm specificity

  • Knockout/knockdown validation: Testing in cell lines with genetic deletion or RNAi-mediated knockdown of target proteins

  • Cross-reactivity testing: Evaluating antibody performance across multiple RSK family members

For example, the phospho-RPS6KA1 (Thr359/Ser363) antibody specificity can be verified by comparing western blot results between normal and λ phosphatase-treated HeLa cells, where phosphatase treatment should eliminate the signal .

A rigorous validation protocol enhances experimental reproducibility and data reliability, particularly for multiplex analysis of phosphorylation events within the RSK family.

What are the optimal experimental conditions for detecting RPS6KA family phosphorylation in different cell types?

Detection of RPS6KA phosphorylation varies by cell type and experimental objective:

Cell TypeRecommended Lysis BufferStimulation ProtocolAntibody Dilution (WB)Incubation Conditions
Cancer cell lines (K-562, HeLa)PBS with 0.02% sodium azide, 50% glycerolSerum starvation (12h) followed by EGF (100ng/ml, 10 min)1:500-1:2000Overnight at 4°C
Primary neuronsRIPA with phosphatase inhibitorsBDNF (50ng/ml, 30 min)1:500-1:1000Overnight at 4°C
Immune cellsNP-40 buffer with protease inhibitorsPMA (100nM, 15 min)1:1000-1:50002h at room temperature

For optimal detection:

  • Always include phosphatase inhibitors in lysis buffers

  • Quickly process samples to prevent dephosphorylation

  • Consider cell-specific stimuli that activate MAPK pathways

  • Use BSA (not milk) for blocking when detecting phospho-epitopes

Research shows that phosphorylation of Ser221/227/S218/232 sites is particularly sensitive to growth factor stimulation, while Thr359/Ser363 phosphorylation may require different activation conditions .

How can RPS6KA phosphorylation states be accurately quantified in multiplex assays?

For accurate multiplex quantification of RPS6KA phosphorylation:

  • Sequential immunoblotting approach:

    • Strip and reprobe membranes with total RPS6KA antibodies after phospho-detection

    • Calculate phospho/total protein ratios for normalization

    • Use loading controls that do not overlap with RSK family members by molecular weight

  • Flow cytometry-based quantification:

    • For intracellular phospho-RPS6KA detection, use the validated concentration of 0.06 μg per 10^6 cells in a 100 μl suspension

    • Include λ phosphatase-treated controls

    • Use fluorescence minus one (FMO) controls to define positive populations

  • Phospho-proteomic analysis:

    • Enrich phosphopeptides using TiO₂ or IMAC

    • Target specific RPS6KA phosphosites (Ser221/227/S218/232, Thr359/Ser363, etc.)

    • Use isotope-labeled internal standards for absolute quantification

The fold change in phosphorylation should be calculated relative to basal conditions and normalized to total protein expression to account for variations in protein abundance .

What strategies can be employed to study the differential roles of RPS6KA family members in specific signaling pathways?

To dissect the specific roles of RPS6KA family members:

  • Isoform-specific knockdown/knockout approaches:

    • Use siRNA or CRISPR-Cas9 to target individual family members

    • Validate specificity using antibodies that recognize unique epitopes

    • Assess compensatory mechanisms by other RSK family members

  • Domain-specific inhibitors:

    • Use BI-D1870 as an RSK inhibitor to study functional outcomes

    • Compare with MEK inhibitors to distinguish direct vs. upstream effects

    • Titrate inhibitor concentrations to achieve selective inhibition

  • Phospho-mutant expression:

    • Generate phospho-mimetic (S→D/E) or phospho-deficient (S→A) mutants

    • Express in knockout backgrounds to avoid interference from endogenous proteins

    • Assess functional rescues using phenotypic assays

  • Interaction studies:

    • Use co-immunoprecipitation to identify isoform-specific binding partners

    • For structural studies, the critical interaction between SPRED2 and RSK2 involves hydrogen bonds that can be disrupted by specific mutations (e.g., SPRED2 F145A)

This approach revealed that RSK2 specifically regulates long-term synaptic facilitation, while other family members may have distinct neuronal functions .

How does sample preparation affect the detection of RPS6KA family members in different experimental applications?

Sample preparation significantly impacts RPS6KA detection across different applications:

For Western Blotting:

  • Use fresh samples whenever possible; freeze-thaw cycles reduce phospho-epitope detection

  • Include both phosphatase inhibitors (e.g., sodium fluoride, sodium orthovanadate) and protease inhibitors

  • Optimal lysis buffers vary by experiment: RIPA for total protein, NP-40 for preserving protein interactions

  • Heating samples can destroy some phospho-epitopes; consider sample preparation at 70°C instead of 95°C

For Immunohistochemistry:

  • Antigen retrieval is critical - use TE buffer pH 9.0 for optimal results

  • Alternative method: citrate buffer pH 6.0

  • Fixation should be brief (10-15 minutes) to prevent epitope masking

  • Dilution range: 1:50-1:500 depending on tissue type

For Flow Cytometry:

  • Permeabilization requires optimization for intracellular kinase detection

  • For RPS6KA1 (Thr359/Ser363), use 0.06 μg per 10^6 cells in suspension

  • Include viability dyes to exclude dead cells that may show nonspecific binding

Experimental validation shows that immunohistochemistry of human liver cancer tissue requires specific antigen retrieval conditions for optimal detection of RPS6KA1 .

What are the key considerations when using RPS6KA family antibodies for studying disease mechanisms, particularly in cancer research?

When employing RPS6KA antibodies in disease research:

  • Expression analysis in different cancer types:

    • RPS6KA1 is overexpressed in head and neck squamous cell carcinoma (HNSCC), particularly in advanced stage (III-IV) disease

    • Expression correlates with HPV status and clinical stage in HNSCC (p < 0.001)

    • Expression patterns differ across cancer types, necessitating tissue-specific protocols

  • Correlation with immune microenvironment:

    • RPS6KA1 expression positively correlates with tumor immune score and B cells in HNSCC

    • Flow cytometry validation of immune cell populations requires specific gating strategies

    • Analysis should include CD19+ B cells, CD3+ T cells, CD4+ T lymphocytes, and CD3+CD8+ T lymphocytes

  • Therapeutic resistance mechanisms:

    • RPS6KA1 mediates resistance to venetoclax/azacitidine in acute myeloid leukemia (AML)

    • RPS6KA1 inhibition with BI-D1870 can restore drug sensitivity in resistant cells

    • Antibodies can monitor treatment effects on phosphorylation status

  • Biomarker potential:

    • Phospho-specific antibodies may detect activation status that correlates with drug response

    • Western blot and qPCR validation should be performed on patient-derived samples

    • ROC curve analysis should be used to determine diagnostic threshold values

These approaches have identified RPS6KA1 as a potential diagnostic biomarker and therapeutic target in HNSCC and AML .

How can antibodies against RPS6KA family members be effectively used to study protein-protein interactions?

To study RPS6KA protein-protein interactions:

  • Co-immunoprecipitation optimization:

    • Pre-clearing step is critical: Incubate 500 μg protein lysate with 1.5 μg normal IgG for 2 hours, followed by Protein A/G Plus incubation

    • For phosphorylation-dependent interactions, maintain samples at 4°C throughout

    • Include phosphatase inhibitors to preserve interaction status

    • Always include negative control using normal IgG instead of primary antibody

  • Proximity ligation assays:

    • Use pairs of antibodies raised in different species against target proteins

    • Ensure antibodies recognize distinct, accessible epitopes when proteins interact

    • Include single antibody controls to establish background signal levels

  • Structure-based interaction studies:

    • The RSK2-SPRED2 complex forms nine hydrogen bonds at the interface, burying 940 Ų of RSK2 surface area

    • Critical residues like SPRED2 F145 are essential for binding; F145A mutation abolishes interaction

    • The binding pocket is conserved across the RSK family, supporting cross-family interactions

  • Interaction domain mapping:

    • Generate domain deletion constructs to identify binding regions

    • Use specific antibodies against different domains to confirm results

    • Consider post-translational modifications that may regulate interactions

These approaches revealed that p-rpS6 structurally interacts with Arp3, Eps8, and actin, suggesting its role in regulating F-actin organization and blood-testis barrier dynamics .

What controls should be included when using phospho-specific antibodies against RPS6KA family members?

Essential controls for phospho-specific RPS6KA antibody experiments:

Control TypePurposeImplementationExpected Result
Phosphatase treatmentVerify phospho-specificityTreat lysates with λ phosphataseLoss of signal in Western blot/IHC/flow cytometry
Stimulation controlConfirm antibody sensitivityTreat cells with activators (PMA, growth factors)Increased signal intensity
Inhibitor controlValidate signaling pathwayPretreat with MEK inhibitors (U0126, PD98059)Reduced phospho-signal
Knockdown/knockoutConfirm antibody specificitysiRNA or CRISPR targeting specific RSKReduced/absent signal
Peptide competitionVerify epitope specificityPre-incubate antibody with phospho/non-phospho peptidesPhospho-peptide should block signal; non-phospho should not
Cross-reactivity testAssess family member specificityTest on samples overexpressing specific RSK isoformsSignal intensity should correlate with expression patterns

For RPS6KA1/RPS6KA3/RPS6KA2/RPS6KA6 phospho-antibodies, manufacturers recommend confirming specificity by showing that non-phospho-specific antibodies can be removed by chromatography using non-phosphopeptide .

Flow cytometry validation for phospho-RPS6KA1 (Thr359/Ser363) should include λ phosphatase-treated HeLa cells as a negative control .

How can RPS6KA antibodies be used to study the role of these kinases in neurological disorders?

For studying RPS6KA in neurological disorders:

  • Coffin-Lowry Syndrome (CLS) models:

    • CLS is caused by mutations in RPS6KA3 (RSK2) gene

    • Antibodies can assess RSK2 expression and phosphorylation in patient-derived cells

    • Knockout/knockdown models can be validated using specific anti-RSK2 antibodies

    • Aplysia sensorimotor culture system provides a valuable model for studying RSK2 in synaptic plasticity

  • Long-term synaptic facilitation (LTF) assessment:

    • RSK2 inhibition reduces CREB1 phosphorylation and impairs LTF

    • Antibodies against phospho-CREB and phospho-RSK2 can monitor this pathway

    • Knockout of RSK by RNAi impairs LTF but can be rescued by computationally designed spaced training protocols

  • Blood-brain barrier studies:

    • RPS6KA family members regulate barrier dynamics in various tissues

    • Similar to the blood-testis barrier regulation by rpS6 affecting F-actin organization

    • Antibodies can be used to study kinase localization at barrier junctions

  • Neurodevelopmental processes:

    • RSK2 regulates neuronal excitability and memory formation

    • Phospho-specific antibodies can track activation during various developmental stages

    • IHC applications require specific optimization for neural tissues

These approaches have established that RSK is required for learning-related synaptic plasticity and enhancement in neuronal excitability .

What are the methodological considerations for multiplexing different RPS6KA family antibodies in the same experiment?

For successful multiplexing of RPS6KA family antibodies:

  • Antibody selection strategy:

    • Choose antibodies raised in different host species (e.g., rabbit anti-RPS6KA1, mouse anti-RPS6KA3)

    • For same-species antibodies, use directly conjugated primary antibodies

    • Select antibodies targeting different domains or phosphorylation sites

    • Verify lack of cross-reactivity before multiplexing

  • Sequential immunodetection protocol:

    • Strip and reprobe strategy: Document complete stripping using secondary antibody only

    • Order matters: detect low-abundance targets first, then more abundant proteins

    • For phospho/total protein pairs, always detect phospho-form first

  • Spectral separation for immunofluorescence:

    • Choose fluorophores with minimal spectral overlap

    • Include single-stain controls for compensation

    • Use nuclear counterstain as reference point for co-localization

  • Cross-validation approaches:

    • Confirm multiplexed results with individual staining experiments

    • Use alternative detection methods (e.g., mass spectrometry) to validate findings

    • Include cell type-specific markers when working with heterogeneous samples

This approach has been validated in studies examining the phosphorylation status of multiple RSK family members in response to various stimuli and inhibitors .

What techniques can be used to study the temporal dynamics of RPS6KA phosphorylation in living cells?

To study temporal dynamics of RPS6KA phosphorylation:

  • Live-cell imaging approaches:

    • FRET-based biosensors incorporating specific phospho-binding domains

    • Design sensors with RSK consensus phosphorylation sequences

    • Include appropriate controls (non-phosphorylatable mutants)

    • Time-resolution should be optimized based on the expected kinetics

  • Time-course immunoblotting optimization:

    • Rapid sample collection and processing is crucial

    • Recommended time points: 0, 5, 15, 30, 60, 120 minutes post-stimulation

    • Process all samples simultaneously to minimize technical variation

    • Quantify phospho/total protein ratios at each time point

  • Microfluidic-based single-cell analysis:

    • Enables capturing cell-to-cell variability in signaling dynamics

    • Requires optimized fixation and permeabilization protocols

    • Cell-specific antibody concentrations may need adjustment

    • Include unstimulated control cells in each experiment

  • Inhibitor wash-out studies:

    • Pre-treat with RSK inhibitors (e.g., BI-D1870)

    • Rapidly wash out inhibitor and monitor re-activation kinetics

    • Provides insights into negative feedback mechanisms

These approaches have revealed distinct temporal patterns of RSK activation in response to various stimuli, with implications for understanding signaling specificity in different cellular contexts .

How can RPS6KA antibodies be used to identify novel therapeutic targets in cancer research?

For identifying novel therapeutic targets using RPS6KA antibodies:

  • Target validation in drug resistance mechanisms:

    • RPS6KA1 was identified among the most significantly depleted sgRNA-genes in venetoclax/azacitidine-treated AML cells

    • Addition of the RPS6KA1 inhibitor BI-D1870 to venetoclax/azacitidine decreased proliferation and colony formation

    • BI-D1870 completely restored sensitivity in resistant OCI-AML2 cells

    • Antibodies can monitor changes in phosphorylation status after treatment

  • Biomarker identification protocol:

    • Analyze RPS6KA expression patterns across cancer types

    • In HNSCC, RPS6KA1 expression is significantly higher in stage III-IV compared to stage I-II

    • Correlate expression with clinical outcomes using tissue microarrays

    • Validate findings using multiple antibodies targeting different epitopes

  • Pathway analysis approach:

    • Use phospho-specific antibodies to map activation states

    • Combine with inhibitor studies to identify critical nodes

    • Correlate with patient response to targeted therapies

    • RPS6KA1 has certain predictive value for anti-PD-1 or CTLA-4 reactivity

  • Immunotherapy correlations:

    • RPS6KA1 expression positively correlates with immune score and B cells in HNSCC

    • Flow cytometry validation requires specific antibody combinations

    • Analyze relationships between RPS6KA activation and immune checkpoint expression

These approaches have identified RPS6KA1 as both a potential biomarker and therapeutic target, particularly in combination therapies for AML and HNSCC .

What are the technical challenges and solutions for detecting low-abundance phosphorylated RPS6KA proteins in primary tissue samples?

Addressing challenges in detecting phosphorylated RPS6KA in primary tissues:

  • Optimized tissue handling protocol:

    • Collect samples in phosphatase inhibitor-containing buffers

    • Flash-freeze within 10 minutes of collection

    • Process frozen sections rather than paraffin when possible

    • For FFPE samples, extended antigen retrieval may be necessary

  • Signal amplification strategies:

    • Tyramide signal amplification can enhance detection sensitivity

    • Polymer-based detection systems provide better signal-to-noise ratio

    • Longer primary antibody incubation (overnight at 4°C) improves detection

    • For IHC applications in human liver cancer tissue, dilutions of 1:50-1:200 are recommended

  • Phospho-enrichment methods:

    • Immunoprecipitate total RPS6KA first, then probe for phospho-forms

    • Use phospho-peptide enrichment strategies prior to mass spectrometry

    • Employ sequential extraction to separate nuclear and cytoplasmic fractions

    • Multiple antibodies targeting different phospho-sites provide complementary data

  • Quantification approaches:

    • Use digital pathology platforms for standardized quantification

    • Include phosphatase-treated control sections

    • Establish tissue-specific positive controls (e.g., mitotic cells for active kinases)

    • Digital image analysis should include background subtraction and normalization

These techniques have been validated in studies of RPS6KA expression in clinical samples, including HNSCC tissues where Western blot and qPCR confirmed upregulation of RPS6KA1 .

How can researchers troubleshoot common issues with RPS6KA family antibodies in experimental applications?

Troubleshooting guide for RPS6KA antibody applications:

IssuePossible CausesSolutionsValidation Method
No signal in Western blotDegraded phospho-epitopeAdd fresh phosphatase inhibitors, process samples quicklyInclude positive control lysate
High backgroundInsufficient blocking, antibody concentration too highOptimize blocking (use 5% BSA instead of milk), titrate antibodyTest dilution series (1:500-1:50000)
Multiple bandsCross-reactivity with other RSK family membersUse isoform-specific antibodies, validate with knockout samplesPeptide competition assay
Inconsistent resultsVariable phosphorylation statusStandardize stimulation protocols, control incubation timesInclude time-course experiments
Poor immunoprecipitationEpitope masked by protein interactionsTry different antibodies targeting different epitopesValidate with overexpression systems
No signal in IHCIneffective antigen retrievalTest both TE buffer pH 9.0 and citrate buffer pH 6.0 Include positive control tissues
Loss of signal in stored samplesPhospho-epitope degradationAliquot antibodies, avoid freeze-thaw cyclesCompare fresh vs. stored samples
Variable flow cytometry resultsInconsistent permeabilizationStandardize fixation/permeabilization protocolUse calibration beads

For phospho-specific antibodies, non-phospho-specific antibodies should be removed by chromatography using non-phosphopeptide during production , which can be verified through appropriate controls.

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.