Detects endogenous RSK1 phosphorylation in EGF-treated HepG2 cells .
Shows nuclear localization in prostate cancer bone metastasis specimens .
Blocks NFκB-mediated inflammation in myeloproliferative neoplasms when RSK1 is inhibited .
MAPK/ERK Signaling: Phosphorylates CREB1, ETV1, and NR4A1 to drive mitogenic responses .
mTOR Regulation: Modulates TSC2 and RPTOR to influence mTORC1 activity .
Cell Survival: Suppresses apoptosis by phosphorylating BAD and DAPK1 .
Cancer Progression: Elevated phospho-RSK1 (T359+S363) correlates with prostate cancer bone metastasis (40% positivity in clinical specimens) .
Inflammatory Disorders: RSK1 inhibition reduces pro-inflammatory cytokines (TNF, IL6, CCL3) in myeloproliferative neoplasms .
Specificity: No cross-reactivity with non-phosphorylated RSK1 .
Lot Consistency: Recombinant production ensures batch-to-batch reproducibility .
The Anti-phospho-RPS6KA1 (T359+S363) antibody is a recombinant monoclonal antibody that specifically recognizes human RPS6KA1 phosphorylated at Thr359 and Ser363 residues. This antibody is generated from tissue culture supernatant (TCS) of cell lines expressing the human phospho-RPS6KA1 (T359+S363) monoclonal antibody gene. Subsequently, it undergoes affinity-chromatography purification. This antibody is a rabbit IgG and is suitable for various applications such as ELISA, Western blotting (WB), immunofluorescence (IF), and immunoprecipitation (IP) using human samples.
RPS6KA1 (RSK1) is a serine/threonine kinase regulated by growth factors, playing a crucial role in the MAPK and PI3K signaling pathways. The C-terminal kinase domain of RPS6KA1 is involved in autophosphorylation, while the N-terminal kinase domain is responsible for phosphorylating various substrates. It controls cellular proliferation and differentiation through phosphorylation of transcription factors, signaling kinases, and pro-apoptotic proteins.
RPS6KA1 (RSK1), a serine/threonine-protein kinase, functions downstream of ERK (MAPK1/ERK2 and MAPK3/ERK1) signaling, mediating mitogenic and stress-induced activation of transcription factors CREB1, ETV1/ER81, and NR4A1/NUR77. It regulates translation via phosphorylation of RPS6 and EIF4B. RSK1 further modulates cellular proliferation, survival, and differentiation by influencing mTOR signaling and suppressing pro-apoptotic function of BAD and DAPK1.
In fibroblasts, RSK1 is essential for EGF-stimulated CREB1 phosphorylation, leading to subsequent transcriptional activation of immediate-early genes. Upon mitogenic stimulation (EGF and PMA), it phosphorylates and activates transcription factors NR4A1/NUR77 and ETV1/ER81, along with the cofactor CREBBP.
In response to insulin-derived signals, RSK1 indirectly affects gene transcription regulation by phosphorylating GSK3B at Ser-9, inhibiting its activity. It phosphorylates RPS6 in response to serum or EGF through an mTOR-independent mechanism, facilitating the assembly of the pre-initiation complex and promoting translation initiation. RSK1 also phosphorylates EIF4B upon insulin stimulation, enhancing its affinity for the EIF3 complex and stimulating cap-dependent translation.
RSK1 plays a role in the mTOR nutrient-sensing pathway by directly phosphorylating TSC2 at Ser-1798, potently inhibiting its ability to suppress mTOR signaling. Furthermore, it mediates phosphorylation of RPTOR, regulating mTORC1 activity and potentially promoting rapamycin-sensitive signaling independently of the PI3K/AKT pathway.
RSK1 promotes cell survival by phosphorylating the pro-apoptotic proteins BAD and DAPK1, suppressing their pro-apoptotic functions. It contributes to the survival of hepatic stellate cells by phosphorylating CEBPB in response to the hepatotoxin carbon tetrachloride (CCl4). RSK1 mediates the induction of hepatocyte proliferation by TGFA through phosphorylation of CEBPB.
RSK1 is involved in cell cycle regulation by phosphorylating the CDK inhibitor CDKN1B, promoting its association with 14-3-3 proteins and preventing its nuclear translocation and inhibition of G1 progression.
RSK1 phosphorylates EPHA2 at Ser-897, and this RSK-EPHA2 signaling pathway controls cell migration.
Phosphorylation at T359/S363 sites is crucial for the activation and regulation of p90RSK (RPS6KA1), a key signaling protein involved in cell growth, survival, and proliferation pathways . These specific phosphorylation events occur within the activation loop of the N-terminal kinase domain and are necessary for full catalytic activity of RPS6KA1. This phosphorylation is typically mediated by ERK1/2 following growth factor stimulation and represents a critical step in the MAPK signaling cascade . Functionally, this phosphorylation enables RPS6KA1 to subsequently phosphorylate downstream substrates involved in transcriptional regulation, protein synthesis, and cell cycle progression, making it a pivotal regulatory point in cellular signaling networks .
Phospho-RPS6KA1 (T359/S363) antibodies have been validated for several experimental applications with varying optimal dilutions:
When selecting the appropriate application, researchers should consider their specific experimental goals. Western blotting provides quantitative measurement of phosphorylation status across different experimental conditions, while flow cytometry allows for analysis at the single-cell level, which is particularly valuable for heterogeneous cell populations . All applications require proper controls, including λ phosphatase-treated samples to confirm antibody specificity for the phosphorylated form of the protein .
RPS6KA1 (RSK1) functions as a downstream effector of the MAPK pathway, particularly ERK1/2 signaling . Following growth factor stimulation, the signaling cascade progresses as follows:
Growth factors bind to receptor tyrosine kinases, activating the RAS-RAF-MEK-ERK pathway
Activated ERK1/2 phosphorylates RPS6KA1 at multiple sites, including T359/S363, which are critical for activation
Once activated, RPS6KA1 mediates several cellular processes by phosphorylating various downstream substrates:
This positions RPS6KA1 as a crucial mediator that translates extracellular signals into specific cellular responses related to growth, differentiation, and survival . Understanding this pathway is essential for interpreting experimental results when working with phospho-specific antibodies targeting this protein.
Proper controls are essential for validating the specificity of phospho-specific antibodies:
Validation should include side-by-side comparison between stimulated and unstimulated conditions to demonstrate the dynamic range of phosphorylation detection. For Western blotting, treatment with λ phosphatase is particularly important as it removes phosphate groups, allowing researchers to confirm that the observed signal is truly phosphorylation-dependent rather than non-specific binding . Based on available data, the expected molecular weight of phosphorylated RPS6KA1 is approximately 83-90 kDa when detected on Western blots .
Preservation of phosphorylation status is critical when working with phospho-specific antibodies:
Rapid sample processing: Harvest and process cells quickly to minimize dephosphorylation by endogenous phosphatases
Phosphatase inhibitor cocktails: Always include fresh phosphatase inhibitors in lysis buffers:
Serine/threonine phosphatase inhibitors (e.g., sodium fluoride, β-glycerophosphate)
Tyrosine phosphatase inhibitors (e.g., sodium orthovanadate)
Broad-spectrum inhibitors (e.g., sodium pyrophosphate, microcystin-LR)
Cold temperature maintenance: Keep samples on ice throughout processing
Optimal lysis buffer composition:
Storage considerations: Aliquot lysates and store at -80°C; avoid repeated freeze-thaw cycles
For phosphorylation analysis, timing of sample collection post-stimulation is crucial as T359/S363 phosphorylation typically peaks within 5-15 minutes following growth factor treatment. Sample preparation techniques should be thoroughly documented in research protocols to ensure reproducibility and reliable phosphorylation detection .
To effectively induce and detect RPS6KA1 T359/S363 phosphorylation:
Stimulation Agent | Concentration | Duration | Cell Types |
---|---|---|---|
Epidermal Growth Factor (EGF) | 50-100 ng/mL | 5-15 min | Epithelial cells, fibroblasts |
Serum | 10-20% | 10-30 min | Most adherent cell lines |
Phorbol esters (PMA/TPA) | 100-200 nM | 15-30 min | Various cell types |
Insulin | 100 nM | 10-20 min | Metabolically responsive cells |
Cell line selection is important as expression and phosphorylation patterns of RPS6KA1 vary across tissue types. HeLa cells have been validated as a positive control system for detecting T359/S363 phosphorylation . For experimental design, researchers should establish a time course of stimulation, as phosphorylation events are often transient. When testing new cell systems, researchers should verify RPS6KA1 expression levels before proceeding with phosphorylation studies, as expression levels impact detection sensitivity. Additionally, serum starvation (0.1-0.5% serum for 16-24 hours) prior to stimulation often enhances detection of induced phosphorylation by reducing baseline signaling activity .
RPS6KA1 activation involves a complex sequence of phosphorylation events at multiple sites:
The activation of RPS6KA1 follows a specific order: ERK1/2 first phosphorylates T573 in the C-terminal kinase domain, followed by phosphorylation of T359/S363 in the N-terminal kinase domain. The C-terminal kinase domain then phosphorylates S380, creating a docking site for PDK1, which phosphorylates S221, completing the activation process . Researchers investigating RPS6KA1 should consider analyzing multiple phosphorylation sites simultaneously to gain a comprehensive understanding of activation status. The interconnected nature of these phosphorylation events means that disruption at any step (through mutations or inhibitors) can affect the entire activation cascade, highlighting the importance of phospho-specific antibodies that target distinct sites .
The RSK family consists of four isoforms (RSK1-4) with high sequence homology, presenting challenges for isoform-specific phosphorylation detection:
Sequence similarity around phosphorylation sites: The T359/S363 region has high conservation among RSK family members, potentially causing cross-reactivity
Verification strategies:
Knockout/knockdown validation: Use siRNA or CRISPR targeting specific RSK isoforms
Isoform-specific expression: Overexpress individual RSK family members
Mass spectrometry: For definitive identification of phosphorylated peptides
Antibody validation data: Review manufacturer's cross-reactivity testing data
Peptide competition assays: Confirm specificity using blocking peptides containing the phosphorylated epitope
When investigating RPS6KA1-specific functions, researchers should employ multiple approaches to confirm isoform specificity. This could include using cell lines with differential expression of RSK family members or verifying results with multiple antibodies targeting different epitopes. For advanced studies, considering the use of phosphatase-resistant phosphomimetic mutants (T359D/S363D) can help distinguish the functional consequences of phosphorylation at these specific sites versus other regulatory sites .
For comprehensive signaling pathway analysis:
Multiplex analysis approaches:
Sequential blotting with antibodies against multiple pathway components
Phospho-proteomics to identify global phosphorylation changes
Kinase activity assays to measure functional outcomes
Key pathway components to analyze alongside RPS6KA1:
Upstream: ERK1/2 phosphorylation status
Parallel: AKT, p38 MAPK, and JNK activation
Downstream: S6, CREB, and BAD phosphorylation
Inhibitor studies to dissect pathway connectivity:
MEK inhibitors (U0126, PD98059) to block ERK1/2-mediated RPS6KA1 phosphorylation
RSK inhibitors (BI-D1870, SL0101) to distinguish RPS6KA1-specific effects
mTOR inhibitors (rapamycin) to differentiate RPS6KA1 from S6K1 effects on downstream targets
Temporal analysis:
Time-course experiments to map activation sequence
Pulse-chase approaches to determine phosphorylation stability
By examining multiple components simultaneously, researchers can place RPS6KA1 T359/S363 phosphorylation in proper context within the signaling network . This integrated approach also helps identify potential compensatory mechanisms or feedback loops that might impact experimental outcomes when targeting RPS6KA1 specifically.
When troubleshooting phosphorylation-specific detection, researchers should systematically evaluate each step of their protocol. For Western blotting, transferring phosphorylated proteins efficiently may require optimization of transfer conditions. For flow cytometry applications, fixation and permeabilization protocols significantly impact epitope preservation and accessibility. Additionally, certain cell types may exhibit different baseline phosphorylation levels or activation kinetics, necessitating protocol adjustments .
For accurate quantification of phosphorylation changes:
Normalization approaches:
Ratio of phospho-RPS6KA1 to total RPS6KA1 (preferred method)
Normalization to housekeeping proteins (GAPDH, β-actin) when comparing total lysate loading
Use of loading controls specific to subcellular fractions if performing fractionation
Quantification methods:
Densitometry for Western blots with linear range validation
Mean fluorescence intensity (MFI) measurements for flow cytometry
ELISA-based quantification for high-throughput analysis
Statistical considerations:
Perform at least three independent biological replicates
Use appropriate statistical tests for the experimental design
Consider the dynamic range of the assay and avoid saturation
Technical validation:
Include a stimulation dose-response to demonstrate proportional signal changes
Use both positive (stimulated) and negative (unstimulated or phosphatase-treated) samples as references
These approaches help ensure that observed changes in phosphorylation are biologically meaningful rather than technical artifacts. When reporting quantitative changes, researchers should clearly describe their normalization approach and include both representative images and quantitative data with statistical analysis .
Understanding the limitations of phospho-specific antibodies across experimental systems is crucial:
Species cross-reactivity considerations:
Cell/tissue-specific challenges:
Primary tissues may require optimized extraction protocols to preserve phosphorylation
High phosphatase activity in certain tissues may necessitate stronger inhibitor cocktails
Autofluorescence in certain tissues may interfere with immunofluorescence/flow cytometry applications
Fixed vs. fresh samples:
Formalin fixation can mask phospho-epitopes or cause epitope degradation over time
Antigen retrieval methods may need optimization for immunohistochemistry
Phosphorylation status may not be preserved in long-term stored samples
Application-specific limitations:
Western blot: Limited spatial information within cell populations
Flow cytometry: Requires single-cell suspensions, limiting tissue architecture information
Immunohistochemistry: More qualitative than quantitative for phospho-epitopes
ELISA: May have cross-reactivity issues unless highly validated
Researchers should validate antibodies in their specific experimental system rather than relying solely on manufacturer data. Preliminary experiments should include appropriate controls to establish sensitivity and specificity boundaries for the chosen application .
RPS6KA1 T359/S363 phosphorylation has significant implications in cancer biology:
Cancer-related phenotypes associated with RPS6KA1 hyperactivation:
Correlation with clinical parameters:
Increased phosphorylation often correlates with more aggressive disease in certain cancers
May serve as a biomarker for MAPK pathway activation
Potential predictive value for response to targeted therapies
Therapeutic implications:
RSK inhibitors under investigation as potential cancer therapeutics
Combination strategies targeting both upstream (MEK/ERK) and downstream (RSK) components
Potential synthetic lethality approaches with other pathway inhibitors
Researchers investigating cancer-related signaling should consider analyzing RPS6KA1 T359/S363 phosphorylation alongside other markers of MAPK pathway activation. The dynamic relationship between RPS6KA1 activation and cellular phenotypes makes phospho-specific antibodies valuable tools for monitoring pathway activity in response to therapeutic interventions .
Phospho-RPS6KA1 analysis reveals important pathway interconnections:
MAPK and mTOR pathway cross-talk:
MAPK and PI3K/AKT pathway integration:
RPS6KA1 and AKT share several downstream substrates (e.g., BAD, GSK3β)
Differential phosphorylation of shared substrates can reveal pathway dominance
Analysis of substrate phosphorylation patterns can identify compensatory mechanisms
Cell cycle regulation cross-talk:
Experimental approaches to study cross-talk:
Combinatorial inhibitor treatments targeting multiple pathways
Temporal analysis of phosphorylation events across pathways
Genetic manipulation of pathway components with phospho-RPS6KA1 readouts
This multi-pathway perspective is crucial for understanding cellular responses to targeted therapies and developing more effective combination strategies. Researchers should design experiments that simultaneously monitor key nodes in interconnected pathways to fully understand the network-level consequences of specific interventions .
Phospho-RPS6KA1 analysis offers several applications in drug development:
Target engagement biomarker:
Direct measure of MAPK pathway inhibition for ERK-targeted therapies
Early pharmacodynamic marker in drug development
Dose-finding tool in preclinical and clinical studies
Resistance mechanism identification:
Persistent RPS6KA1 phosphorylation despite upstream inhibition suggests bypass mechanisms
Temporal changes in phosphorylation patterns may reveal adaptive resistance
Combination therapy rationale development based on phosphorylation profiles
Patient stratification strategies:
Baseline phosphorylation status may predict response to pathway-targeted therapies
Dynamic changes upon treatment could serve as early response indicators
Integration with other biomarkers for refined patient selection
Novel therapeutic approaches:
Identification of synthetic lethal interactions based on phosphorylation status
Development of degraders or inhibitors specifically targeting the phosphorylated form
Combination strategies targeting both the phosphorylation event and downstream effectors
Pharmaceutical researchers can leverage phospho-specific antibodies as tools throughout the drug development process, from target validation to clinical trial biomarker analysis. The specificity of phosphorylation-state detection provides valuable insights into both on-target activity and potential off-target effects of candidate compounds targeting this signaling pathway .