The antibody recognizes phosphorylated RPS6KB1 (p70 S6K), a serine/threonine kinase activated downstream of mTORC1. Phosphorylation at T421 and S424 is essential for RPS6KB1’s kinase activity, enabling its role in translating mRNAs encoding ribosomal proteins and elongation factors . The recombinant antibody is engineered for enhanced specificity and sensitivity, leveraging synthetic peptides derived from human p70 S6K phosphorylated at T421/S424 as immunogens .
The antibody is validated for multiple techniques, with optimized dilutions for each:
WB Specificity: Preincubation with phosphopeptides (T421/S424) abolishes signals, confirming target specificity .
IP Efficiency: Outperforms rabbit control IgG in immunoprecipitation assays .
Cross-Reactivity: Reacts with rat tissues but not confirmed for zebrafish .
The antibody has been instrumental in studying RPS6KB1’s role in:
mTORC1 → RPS6KB1 → Protein Synthesis: Phosphorylates EIF4B, RPS6, and EEF2K to drive translation .
Feedback Regulation: Inhibits mTORC2 via phosphorylation of RICTOR, modulating AKT signaling .
The antibody is produced via mammalian cell transfection with a recombinant plasma vector encoding the anti-RPS6KB1 sequence, ensuring:
High Batch Consistency: Reduced lot-to-lot variability compared to traditional hybridoma-derived antibodies .
Animal-Free Production: Aligns with ethical and sustainability standards .
Purification: Affinity chromatography yields >95% pure IgG .
Cross-Reactivity: While primarily human-reactive, rat cross-reactivity is documented . Zebrafish reactivity remains unvalidated .
Controls: Phosphopeptide competition is critical to exclude nonspecific binding .
Storage: Repeated freeze-thaw cycles degrade antibody performance; aliquot for long-term use .
This recombinant RPS6KB1 antibody is produced in mammalian cells transfected with a recombinant plasma vector. The vector was engineered to express the antibody against RPS6KB1. Following expression, the recombinant RPS6KB1 antibody is purified from the cell culture medium using affinity chromatography. This antibody exhibits reactivity with samples containing RPS6KB1 protein from human sources and has been validated for use in ELISA, Western blot, Immunofluorescence, and Immunoprecipitation applications.
The T421/S424 phospho-RPS6KB1 antibody specifically recognizes the RPS6KB1 protein phosphorylated at T421/S424 residues. RPS6KB1, also known as S6K1 or p70S6K, is a serine/threonine kinase crucial for ribosomal protein phosphorylation, a process essential for protein translation and elongation. It is an integral component of the PI3K/mTOR signaling pathway. Growth factors and hormones activate RPS6KB1 by phosphorylating its multiple serine and threonine sites in a sequential manner. Active RPS6KB1 phosphorylates ribosomal protein S6, leading to selective translation of the 5'-terminal oligopyrimidine tract mRNAs that code for ribosomal proteins and elongation factors.
Serine/threonine-protein kinase that functions downstream of mTOR signaling in response to growth factors and nutrients, thereby promoting cell proliferation, growth, and progression through the cell cycle. It regulates protein synthesis by phosphorylating EIF4B, RPS6, and EEF2K, and contributes to cell survival by inhibiting the pro-apoptotic function of BAD. Under conditions of nutrient depletion, the inactive form associates with the EIF3 translation initiation complex. Upon mitogenic stimulation, phosphorylation by the mammalian target of rapamycin complex 1 (mTORC1) leads to dissociation from the EIF3 complex and activation. The active form then phosphorylates and activates several substrates in the pre-initiation complex, including the EIF2B complex and the cap-binding complex component EIF4B.
Additionally, it controls translation initiation by phosphorylating a negative regulator of EIF4A, PDCD4, targeting it for ubiquitination and subsequent proteolysis. It promotes the initiation of the pioneer round of protein synthesis by phosphorylating POLDIP3/SKAR. In response to IGF1, it activates translation elongation by phosphorylating EEF2 kinase (EEF2K), leading to its inhibition and thus activation of EEF2. It also plays a role in feedback regulation of mTORC2 by mTORC1 through phosphorylation of RICTOR, resulting in the inhibition of mTORC2 and AKT1 signaling.
RPS6KB1 mediates cell survival by phosphorylating the pro-apoptotic protein BAD and suppressing its pro-apoptotic function. It phosphorylates mitochondrial URI1, leading to the dissociation of a URI1-PPP1CC complex. The free mitochondrial PPP1CC can then dephosphorylate RPS6KB1 at Thr-412, which is proposed to be a negative feedback mechanism for the RPS6KB1 anti-apoptotic function. RPS6KB1 mediates TNF-alpha-induced insulin resistance by phosphorylating IRS1 at multiple serine residues, resulting in accelerated degradation of IRS1.
In cells lacking functional TSC1-2 complex, RPS6KB1 constitutively phosphorylates and inhibits GSK3B. It may be involved in cytoskeletal rearrangement through binding to neurabin. It phosphorylates and activates the pyrimidine biosynthesis enzyme CAD, downstream of MTOR. Following activation by mTORC1, it phosphorylates EPRS, thereby playing a key role in fatty acid uptake by adipocytes and likely in interferon-gamma-induced translation inhibition.
RPS6KB1 (Ribosomal Protein S6 Kinase, 70kDa, Polypeptide 1), also known as p70S6K, is a serine/threonine kinase that plays a crucial role in protein synthesis through the phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling pathway. It functions by activating the substrate ribosomal protein S6, which induces protein synthesis by translating the 5'-terminal oligopyrimidine tract mRNAs. This kinase is essential for cellular growth, proliferation, and various physiological processes, including muscle development and tissue regeneration .
The phosphorylation of RPS6KB1 at T421+S424 sites represents a critical regulatory mechanism in the activation process of this kinase. These phosphorylation sites are located in the C-terminal autoinhibitory domain of the protein. When phosphorylated at these sites, the inhibitory effect of the C-terminal domain on the catalytic domain is released, which is an essential first step in the complete activation of RPS6KB1. This phosphorylation event precedes the subsequent phosphorylation at Thr389 by mTOR, which is required for full kinase activity . Therefore, the T421+S424 phosphorylation serves as an important regulatory switch in the RPS6KB1 activation cascade.
Several serine/threonine kinases have been identified as responsible for the phosphorylation of RPS6KB1 at T421+S424 sites. JNK1 (c-Jun N-terminal kinase 1) has been experimentally demonstrated to phosphorylate S424 in the RPS6KB1 C-terminal domain. This was confirmed through kinase assays with purified S6K as a substrate and in cellular studies where JNK1 was activated by TNF-α or MEKK1 expression . Other kinases reported to participate in this first step of phosphorylation include ERK (for T421/S424), p38, and CDC2 (for S411). These findings highlight the complexity of the RPS6KB1 regulation network and the involvement of multiple MAPK pathway members in its activation process .
For optimal Western blotting results with Phospho-RPS6KB1 (T421+S424) antibodies, follow this methodological approach:
Sample preparation: Lyse cells in a buffer containing phosphatase inhibitors (such as sodium orthovanadate, sodium fluoride, and β-glycerophosphate) to preserve phosphorylation status.
Antibody dilution: Start with a 1:1000 dilution of the primary antibody, though optimal concentration may vary by lot and experimental conditions .
Positive control: Include a positive control such as NIH/3T3 cells treated with phosphatase inhibitors like calyculin A (50 nM) and okadaic acid (500 nM) for 30 minutes, which enhances phosphorylation signals .
Validation: Perform peptide inhibition assays to confirm specificity, using both phosphorylated and non-phosphorylated peptides of the target region.
Detection optimization: Use enhanced chemiluminescence with exposure times adjusted based on signal intensity; gradient exposures may be necessary to capture optimal signal without saturation.
This protocol emphasizes preservation of phosphorylation status and confirmation of antibody specificity, which are critical for reliable results when studying signaling pathway dynamics.
Phospho-RPS6KB1 (T421+S424) recombinant monoclonal antibodies are versatile tools suitable for multiple research applications:
Western Blotting (WB): Provides quantitative analysis of phosphorylation levels in different experimental conditions or disease states .
Immunohistochemistry (IHC): Enables visualization of phosphorylated RPS6KB1 distribution in tissue sections, valuable for pathological studies such as cancer research .
Immunofluorescence (IF): Offers high-resolution subcellular localization of phosphorylated RPS6KB1, important for understanding spatial regulation .
Immunocytochemistry (ICC): Allows detailed examination of phosphorylated RPS6KB1 in cultured cells, useful for in vitro modeling .
ELISA: Provides quantitative measurement of phosphorylated RPS6KB1 levels in cell or tissue lysates .
Each application requires specific optimization of antibody concentration, incubation conditions, and detection methods. For cross-validation of results, it is recommended to use multiple approaches when investigating phosphorylation-dependent signaling pathways.
When encountering weak or absent signals with Phospho-RPS6KB1 (T421+S424) antibodies, consider the following methodological troubleshooting steps:
Phosphorylation state preservation: Ensure immediate cell lysis after stimulation and include fresh phosphatase inhibitors in all buffers. Flash-freeze tissues immediately after collection.
Stimulation protocols: Verify that your experimental conditions actually activate the pathways leading to T421+S424 phosphorylation. Consider using positive controls such as treatment with calyculin A/okadaic acid .
Antibody validation: Confirm antibody activity using a positive control lysate from cells with known high phosphorylation levels.
Cross-reactivity assessment: Test for potential cross-reactivity with other phosphorylated proteins by running peptide competition assays.
Signal enhancement techniques: Try longer primary antibody incubation (overnight at 4°C), increased antibody concentration, or more sensitive detection systems.
Sample processing: Consider phospho-protein enrichment methods prior to analysis for low-abundance targets.
These systematic approaches will help identify whether the issue lies with the experimental conditions, the sample preparation, or the detection methodology.
The phosphorylation of RPS6KB1 occurs in a sequential, multi-step process with distinct functional consequences:
T421+S424 phosphorylation (first stage):
Thr389 phosphorylation (second stage):
The temporal relationship between these phosphorylation events is crucial; T421+S424 phosphorylation typically precedes Thr389 phosphorylation, creating a regulatory mechanism that prevents premature activation. This sequential phosphorylation creates a sophisticated control system that integrates multiple cellular signals before fully activating the kinase.
Research has revealed a complex relationship between RPS6KB1 phosphorylation and non-small cell lung cancer (NSCLC) progression:
These findings highlight the critical distinction between protein expression and phosphorylation status, emphasizing that hyperphosphorylation, rather than merely increased expression, drives the oncogenic activity of RPS6KB1 in NSCLC.
To effectively investigate the complex interplay between different kinases in RPS6KB1 phosphorylation, consider this comprehensive experimental design approach:
Kinase inhibition studies:
Employ specific inhibitors for JNK1, ERK, p38, and mTOR individually and in combination
Use genetic approaches (siRNA, CRISPR-Cas9) to confirm inhibitor specificity
Monitor changes in RPS6KB1 phosphorylation at multiple sites (T421, S424, T389) simultaneously
Phosphorylation site mutants:
Generate phospho-mimetic (S/T→D/E) and phospho-deficient (S/T→A) mutants
Examine how these mutations affect subsequent phosphorylation steps
Assess functional consequences through downstream substrate activation
Temporal dynamics analysis:
Implement time-course experiments after stimulus application
Use phospho-specific antibodies for different sites to establish phosphorylation sequence
Apply mathematical modeling to understand kinetics and threshold effects
Context-specific regulation:
Compare phosphorylation patterns across different cell types
Investigate the effect of microenvironment factors
Examine phosphorylation in normal versus disease states (e.g., NSCLC)
This multi-faceted approach will provide insights into the hierarchical relationships between kinases and help establish both necessary and sufficient conditions for RPS6KB1 activation in different cellular contexts.
A rigorous validation strategy for Phospho-RPS6KB1 (T421+S424) antibodies should include multiple complementary approaches:
Peptide competition assays: Pre-incubate the antibody with phosphorylated and non-phosphorylated peptides corresponding to the target epitope region. A specific antibody will show signal reduction only with the phosphorylated peptide .
Phosphatase treatment: Treat half of your sample with lambda phosphatase before immunoblotting. A phospho-specific antibody should show signal only in the untreated fraction.
Kinase modulators: Compare samples from cells treated with pathway activators versus inhibitors that affect RPS6KB1 phosphorylation.
Genetic approaches: Use cells with RPS6KB1 knockdown/knockout or cells expressing phospho-deficient mutants (T421A/S424A) as negative controls.
Cross-platform validation: Confirm results using complementary techniques (e.g., mass spectrometry-based phosphoproteomics) to verify the detected phosphorylation events.
These validation steps are crucial for ensuring that experimental observations reflect true biological events rather than artifacts or cross-reactivity with other phosphoproteins.
To effectively study RPS6KB1 phosphorylation at T421+S424, researchers should consider these optimized stimulation protocols:
Physiological stimuli:
Pharmacological inducers:
Experimental considerations:
Cell confluency: Maintain at 70-80% for optimal signaling
Serum starvation: Usually 16-24 hours before stimulation to reduce baseline phosphorylation
Time course: Include multiple timepoints (5, 15, 30, 60 minutes) to capture both rapid and sustained phosphorylation events
Cell type variations:
These protocols provide a foundation for investigating both the mechanisms and functional consequences of RPS6KB1 phosphorylation in various research contexts.
To distinguish between the specific effects of T421+S424 phosphorylation and other RPS6KB1 phosphorylation sites, implement these methodological approaches:
Site-specific mutant constructs:
Generate single and combined phosphosite mutants:
T421A+S424A (prevent phosphorylation at these sites)
T389A (prevent mTOR-mediated phosphorylation)
Combinations of these mutations
Express these constructs in cells with RPS6KB1 knockdown background
Compare phenotypic and signaling outcomes to isolate site-specific effects
Temporal analysis with site-specific antibodies:
Perform time-course experiments after stimulation
Use antibodies specific for different phosphorylation sites (T421+S424, T389)
Correlate the timing of each phosphorylation event with downstream effects
Pharmacological approach:
Substrate specificity analysis:
Measure phosphorylation of different RPS6KB1 substrates
Correlate substrate activation with specific phosphorylation events
Identify substrates uniquely dependent on particular phosphorylation patterns
Phospho-RPS6KB1 (T421+S424) antibodies offer valuable applications for cancer biomarker research through several methodological approaches:
Prognostic marker development:
Immunohistochemical evaluation of patient tissue microarrays to correlate phosphorylation status with clinical outcomes
Multivariate analysis incorporating phosphorylation data with other clinicopathological factors to develop comprehensive prognostic models
Longitudinal studies to assess changes in phosphorylation patterns during disease progression
Therapeutic response prediction:
Pre- and post-treatment analysis of phosphorylation status to identify predictive patterns for drug sensitivity
Correlation of baseline phosphorylation with response to mTOR/PI3K pathway inhibitors
Development of companion diagnostic assays for targeted therapies
Monitoring treatment efficacy:
Quantitative analysis of phosphorylation changes as pharmacodynamic markers
Serial biopsy analysis to track pathway modulation during treatment
Correlation of phosphorylation changes with clinical response metrics
Cancer subtyping:
Integration of phosphorylation data with other molecular markers for refined cancer classification
Identification of patient subgroups with hyperactivated RPS6KB1 signaling that might benefit from specific therapeutic approaches
Research in NSCLC has already demonstrated the superior prognostic value of phosphorylated RPS6KB1 compared to total protein expression, highlighting the potential clinical utility of phosphorylation-specific detection in cancer management .
The relationship between RPS6KB1 phosphorylation and response to mTOR pathway inhibitors reveals important mechanistic and clinical insights:
Mechanistic relationship:
RPS6KB1 phosphorylation at T389 is directly mediated by mTOR and serves as a primary pharmacodynamic marker of mTOR activity
Phosphorylation at T421+S424 occurs upstream or in parallel pathways, primarily through MAPK family kinases including JNK1 and ERK
Inhibition of mTOR with rapamycin or rapalogs directly blocks T389 phosphorylation while potentially leaving T421+S424 phosphorylation intact
Second-generation mTOR inhibitors (Torin, PP242) more completely suppress all RPS6KB1 phosphorylation events
Resistance mechanisms:
Clinical implications:
Monitoring both T389 and T421+S424 phosphorylation provides more comprehensive assessment of pathway inhibition
Patients whose tumors maintain T421+S424 phosphorylation during treatment may benefit from combination therapies targeting both mTOR and MAPK pathways
The presence of hyperphosphorylated RPS6KB1 in NSCLC correlates with poorer prognosis, suggesting potential benefit from pathway inhibition
Understanding these complex relationships enables more rational design of targeted therapies and improved prediction of treatment responses in cancers with aberrant mTOR/S6K signaling.
Cell-specific contexts significantly impact experimental outcomes when studying RPS6KB1 phosphorylation, requiring careful consideration in experimental design:
Tissue-specific phosphorylation patterns:
NSCLC tissues demonstrate significantly higher p-RPS6KB1 levels (61.25%) compared to normal lung tissues (41.86%)
Different cancer subtypes show variable dependencies on RPS6KB1 signaling (e.g., lung adenocarcinoma vs. squamous cell carcinoma)
Normal physiological processes like muscle development show context-specific RPS6KB1 activation patterns
Cell line considerations:
Adenocarcinoma cell line A549 shows greater apoptotic response to RPS6KB1 dephosphorylation compared to squamous cell line SK-MES-1
Baseline pathway activation status varies across cell types, affecting stimulation requirements
Genetic background (mutation status of PI3K, PTEN, RAS) alters RPS6KB1 regulation
Microenvironmental factors:
Nutrient availability affects mTOR activity and subsequent RPS6KB1 phosphorylation
Hypoxia modulates phosphorylation patterns through AMPK-mediated pathways
Cell-cell interactions in 3D cultures versus 2D monolayers yield different phosphorylation dynamics
Experimental recommendations:
Include multiple cell lines or primary cells representing different tissues or disease subtypes
Document genetic background of experimental models that might affect RPS6KB1 regulation
Consider physiological relevance of culture conditions (nutrients, oxygen, matrix interactions)
Validate key findings in multiple systems to ensure generalizability
This contextual awareness is essential for accurate interpretation of experimental results and their translation to physiological or pathological significance.
Several cutting-edge technologies are poised to revolutionize the study of site-specific RPS6KB1 phosphorylation:
Mass spectrometry-based approaches:
Targeted phosphoproteomics for absolute quantification of specific phosphosites
Parallel reaction monitoring (PRM) for simultaneous tracking of multiple phosphorylation events
Crosslinking mass spectrometry to identify phosphorylation-dependent protein interactions
Live-cell imaging techniques:
Genetically encoded biosensors based on phospho-specific binding domains
FRET-based reporters to monitor RPS6KB1 phosphorylation in real-time
Super-resolution microscopy to track subcellular localization of differentially phosphorylated RPS6KB1
Single-cell analysis methods:
Single-cell phospho-flow cytometry for heterogeneity assessment
Mass cytometry (CyTOF) for simultaneous detection of multiple phosphorylation events
Single-cell RNA-seq combined with phosphoprotein analysis to correlate signaling with transcriptional changes
Protein engineering approaches:
Expanded genetic code systems to incorporate phosphomimetic non-canonical amino acids
Synthetic phospho-switch protein domains to control RPS6KB1 activity optogenetically
CRISPR-based endogenous tagging for physiological phosphorylation monitoring
These technologies will enable more precise temporal and spatial resolution of phosphorylation events, facilitating deeper understanding of how specific phosphosites contribute to RPS6KB1 regulation in complex cellular environments.
Advanced understanding of RPS6KB1 phosphorylation mechanisms offers promising avenues for novel therapeutic development:
Targeted inhibition strategies:
Site-specific inhibitors targeting T421+S424 phosphorylation rather than catalytic activity
Dual-specificity compounds blocking both mTOR-mediated and MAPK-mediated phosphorylation events
Allosteric modulators that stabilize inactive conformations maintained by dephosphorylation
Rational combination therapies:
Biomarker-guided precision medicine:
Development of diagnostic assays to identify patients with hyperphosphorylated RPS6KB1
Phosphorylation pattern analysis to predict optimal treatment selection
Monitoring phosphorylation dynamics to detect resistance mechanisms early
Novel drug delivery approaches:
Nanoparticle-based delivery of inhibitors to tissues with RPS6KB1 hyperphosphorylation
Phosphorylation-responsive drug release systems
Proteolysis-targeting chimeras (PROTACs) directed against phosphorylated RPS6KB1 forms
The prognostic significance of p-RPS6KB1 in cancers like NSCLC underscores the therapeutic potential of targeting these specific phosphorylation events, potentially offering more selective intervention than broad kinase inhibition approaches .
Current research on phospho-RPS6KB1 faces several methodological limitations that require innovative solutions:
Antibody specificity challenges:
Current limitation: Cross-reactivity between similar phosphorylation motifs across proteins
Solutions: Development of recombinant antibodies with enhanced specificity; complementary mass spectrometry validation; synthetic nanobodies with improved epitope recognition
Temporal resolution limitations:
Current limitation: Difficulty capturing rapid phosphorylation/dephosphorylation events
Solutions: Development of real-time biosensors; optimization of rapid cell fixation protocols; microfluidic platforms for millisecond-scale stimulation and fixation
Contextual dependency understanding:
Current limitation: Variable results across experimental systems and cellular contexts
Solutions: Systematic comparison across multiple cell types; development of more physiologically relevant 3D culture systems; in situ phosphorylation analysis in intact tissues
Stoichiometry determination:
Current limitation: Difficulty quantifying the proportion of RPS6KB1 molecules phosphorylated at specific sites
Solutions: Absolute quantification methods using isotope-labeled phosphopeptides; single-molecule approaches to detect phosphorylation states; improved computational models
Functional significance assessment:
Current limitation: Challenges connecting phosphorylation events to specific biological outcomes
Solutions: Development of rapid, reversible, site-specific phosphatase or kinase systems; phosphomimetic approaches with enhanced physiological relevance; systems biology frameworks linking phosphorylation to downstream effects
Addressing these limitations will require interdisciplinary approaches combining advanced protein engineering, synthetic biology, and analytical technologies to develop more comprehensive understanding of RPS6KB1 regulation and function.