The Phospho-NFKBIA (S32) Recombinant Monoclonal Antibody (e.g., CSB-RA015761A32phHU, CAC12469) is produced using recombinant DNA technology. Key steps include:
Immunogen: A synthetic peptide derived from human NFKBIA phosphorylated at S32 .
Host System: Expressed in HEK293F cells or hybridoma systems .
Purification: Affinity chromatography ensures high specificity .
Clonality: Monoclonal (e.g., clone EDG-14, 2D6) , enabling consistent batch-to-batch reproducibility.
Validated applications and recommended dilutions across platforms:
Application | Dilution Range | Species Reactivity | Key Sources |
---|---|---|---|
Western Blot (WB) | 1:500–1:5000 | Human | |
Immunofluorescence | 1:20–1:200 | Human | |
ELISA | 1:100–1:500 | Human | |
Immunoprecipitation | 1:50–1:1000 | Human |
Phosphorylation at S32 (and S36) triggers NFKBIA degradation, activating NF-κB transcription factors involved in immune responses and inflammation . Key functional insights:
Regulatory Role: Phosphorylation disables NFKBIA’s inhibition of NF-κB, enabling nuclear translocation of RELA/p65 .
Disease Relevance: Dysregulation linked to cancer, inflammatory diseases, and viral infections .
Specificity: Confirmed via peptide-blocking assays and reactivity with phosphorylated NFKBIA in TNF-α-treated HeLa cells .
Molecular Weight: Observed ~80 kDa (vs. calculated 35.6 kDa due to post-translational modifications) .
Cross-Reactivity: Theoretical reactivity with rat (90% sequence homology), but dependent on phosphorylation status .
Mechanistic Studies: Used to demonstrate NFKBIA phosphorylation in TNF-α-stimulated U87/U251 glioblastoma cells .
Therapeutic Insights: Applied in studies linking S32 phosphorylation to FK506-mediated NF-κB activation .
This phospho-NFKBIA (S32) recombinant monoclonal antibody is produced using a robust and highly controlled process. Genes encoding the antibody are isolated from rabbits immunized with a synthetic peptide representing human NFKBIA phosphorylated at Serine 32 (S32). These genes are cloned into expression vectors and transfected into host suspension cells. Subsequently, the cells are cultured to express and secrete the antibody. Purification is achieved using affinity chromatography, yielding a highly purified antibody preparation. Rigorous quality control is performed using ELISA, Western blotting (WB), and immunofluorescence (IF) assays to validate the antibody's specificity and functionality for binding to human NFKBIA phosphorylated at S32.
Phosphorylation of NFKBIA at S32 is a crucial regulatory step in the NF-κB signaling pathway. This modification controls NF-κB activation, influencing various cellular processes such as immune responses, inflammation, and cell survival. Dysregulation of this phosphorylation event is implicated in numerous pathological conditions including inflammatory diseases and cancer.
NFKBIA (IκBα) inhibits the activity of dimeric NF-κB/Rel complexes by retaining Rel dimers in the cytoplasm, masking their nuclear localization signals. Upon cellular stimulation by immune and pro-inflammatory signals, NFKBIA undergoes phosphorylation, triggering ubiquitination and subsequent degradation. This allows the RelA dimer to translocate to the nucleus and activate transcription.
The following studies highlight the role of NFKBIA and its regulation in various biological processes:
Phosphorylation of NFKBIA (also known as IκBα) at serine 32 represents a critical regulatory event in the NF-κB signaling pathway. This post-translational modification triggers the ubiquitination and subsequent proteasomal degradation of NFKBIA, allowing the liberation and nuclear translocation of NF-κB transcription factors. This process controls the activation of NF-κB and influences numerous cellular processes including immune responses, inflammation, and cell survival mechanisms . The phosphorylation event serves as a molecular switch that converts extracellular stimuli into gene expression changes, making it a crucial target for investigating signaling dysregulation in various pathologies .
Phospho-NFKBIA (S32) recombinant monoclonal antibodies are validated for multiple experimental applications with specific recommended dilutions:
Application | Recommended Dilution | Purpose |
---|---|---|
Western Blotting (WB) | 1:500-1:5000 | Detecting phosphorylated NFKBIA in cell/tissue lysates |
Immunofluorescence (IF) | 1:20-1:200 | Visualizing subcellular localization of phosphorylated NFKBIA |
The antibody specifically recognizes human NFKBIA protein when phosphorylated at serine 32 position, making it valuable for studying activation states of the NF-κB pathway . For optimal results, researchers should validate the antibody in their specific experimental systems and adjust dilutions accordingly based on signal-to-noise ratios obtained during preliminary experiments.
The production of phospho-NFKBIA (S32) recombinant monoclonal antibodies involves a sophisticated genetic engineering approach that provides advantages over traditional hybridoma technology. The process encompasses:
Immunization of rabbits with a synthesized phospho-peptide derived from human NFKBIA protein phosphorylated at S32
Isolation of antibody-encoding genes from B cells of immunized animals
Cloning of these genes into specialized expression vectors
Transfection of vectors into host suspension cells
Cultivation of transfected cells to express and secrete antibodies
Purification via affinity chromatography
Functional validation through multiple assays (ELISA, WB, IF)
This recombinant approach enables precise control over antibody properties, eliminates hybridoma instability issues, and allows for consistent batch-to-batch production. Unlike traditional hybridoma-derived antibodies, recombinant antibodies maintain identical characteristics across production lots, enhancing experimental reproducibility in long-term research projects .
Optimizing detection of phosphorylated NFKBIA in low-expression samples requires a multifaceted approach:
Phosphatase inhibitor treatment: Include comprehensive phosphatase inhibitor cocktails in lysis buffers to prevent dephosphorylation during sample preparation. This is critical as phosphorylation at S32 can be rapidly reversed by cellular phosphatases.
Enrichment strategies:
Immunoprecipitation prior to Western blotting
Subcellular fractionation to concentrate cytoplasmic fractions where phosphorylated NFKBIA predominantly localizes
Stimulus-based enrichment using NF-κB pathway activators (TNF-α, IL-1β) to increase phosphorylation events
Signal amplification:
For Western blotting: Use high-sensitivity chemiluminescent substrates and longer exposure times
For immunofluorescence: Implement tyramide signal amplification (TSA) systems that can enhance detection sensitivity by 10-100 fold
Specialized detection systems:
These approaches should be systematically tested and optimized for specific experimental conditions and cell types being investigated.
Distinguishing specific from non-specific signals requires implementation of rigorous controls:
Phosphatase treatment control: Treating a duplicate sample with lambda phosphatase before antibody incubation should eliminate specific phospho-S32 signal while leaving non-specific binding intact.
Peptide competition assay: Pre-incubation of the antibody with excess phospho-S32 peptide should block specific binding sites and eliminate specific signals.
Genetic validation approaches:
Using NFKBIA knockout cell lines as negative controls
Employing NFKBIA S32A mutant cells (where serine is replaced with non-phosphorylatable alanine)
Implementing siRNA knockdown of NFKBIA with appropriate controls
Stimulus-response validation: Treatment with known NF-κB pathway activators should increase phospho-S32 signal intensity in a time-dependent manner, while pathway inhibitors should reduce signal .
Molecular weight verification: Genuine phospho-NFKBIA should appear at approximately 39 kDa on Western blots, with potential shifts due to additional post-translational modifications.
Implementing these controls systematically helps delineate true phospho-NFKBIA detection from artifactual signals.
Accurate quantification of NFKBIA phosphorylation requires careful experimental design and analysis:
Normalization strategy:
Dual probing for total NFKBIA and phospho-NFKBIA to calculate phosphorylation ratio
Use of stable housekeeping proteins (β-actin, GAPDH) as loading controls
Consider normalizing to total protein load using reversible protein stains
Temporal dynamics:
Implement detailed time-course experiments as S32 phosphorylation exhibits rapid and often transient kinetics
Standard time points should include 0, 5, 15, 30, 60, and 120 minutes post-stimulation
Quantification methodology:
Use linear range detection systems rather than oversaturated exposures
Employ digital image analysis software with background subtraction
Perform replicate experiments (minimum n=3) for statistical analysis
Consideration of feedback mechanisms:
These considerations ensure that observed differences in phosphorylation status accurately reflect biological regulation rather than technical artifacts.
NFKBIA phosphorylation at S32 is embedded within a complex signaling network with multiple regulatory inputs and outputs:
Upstream regulators:
IKK complex (composed of IKKα, IKKβ, and IKKγ/NEMO) directly phosphorylates NFKBIA at S32 and S36
Multiple signaling cascades converge on IKK activation, including TNF receptor family, IL-1 receptor, and pattern recognition receptors
DNA damage response pathways can trigger IKK activation via NEMO SUMOylation and phosphorylation
Downstream consequences:
Phosphorylation at S32/S36 triggers K48-linked polyubiquitination and proteasomal degradation
Released NF-κB dimers (predominantly p65/p50) translocate to the nucleus
Transcriptional activation of target genes including NFKBIA itself creates a negative feedback loop
This feedback mechanism ensures that NF-κB activation is typically transient under normal conditions
Intersection with other pathways:
Cross-regulation with MAPK pathways through shared adaptor proteins
Integration with cell death/survival pathways through regulation of anti-apoptotic genes
Connection to cell cycle regulation via p21 and cyclin D expression
Understanding these network interactions is essential for interpreting experimental results in a broader biological context.
Dysregulation of NFKBIA phosphorylation contributes to multiple pathological conditions:
Inflammatory disorders:
Excessive NFKBIA phosphorylation leads to sustained NF-κB activation and chronic inflammation
Implicated in rheumatoid arthritis, inflammatory bowel disease, and asthma pathogenesis
Cancer biology:
Cellular senescence:
Changes in p65 phosphorylation contribute to attenuated NFKBIA expression in senescent cells
This attenuation leads to constitutive IKK activity and senescence-associated secretory phenotype (SASP)
Genetic knockout studies demonstrate that loss of NFKBIA can trigger hyperproliferation and affects cell cycle progression
Neurodegenerative disorders:
Aberrant NFKBIA phosphorylation associated with neuroinflammatory processes
Implicated in Alzheimer's disease, Parkinson's disease, and multiple sclerosis pathology
These pathological connections highlight the therapeutic potential of targeting NFKBIA phosphorylation regulation.
Integration of phospho-specific antibodies into high-throughput screening requires specialized platforms and methodologies:
Antibody microarray technology:
Workflow integration:
Data analysis approaches:
Normalization against internal controls (β-actin, GAPDH)
Ratio calculation between phosphorylated and total protein signals
Statistical analysis for identifying significantly altered phosphorylation events
Pathway enrichment analysis to identify regulated signaling networks
Validation strategy:
Confirmation of key hits using orthogonal methods (Western blotting, mass spectrometry)
Functional validation through genetic or pharmacological manipulation
This systematic approach allows researchers to transition from single-target analysis to comprehensive phosphorylation profiling.
Designing experiments to capture NFKBIA phosphorylation dynamics requires careful consideration of temporal and spatial factors:
Stimulus selection and dosing:
Classical NF-κB activators: TNF-α (10 ng/ml), IL-1β (10 ng/ml), LPS (100 ng/ml)
Genotoxic agents: etoposide, doxorubicin, ionizing radiation
Dosage titration to determine threshold and saturating concentrations
Time-course design:
Rapid early sampling (0, 2, 5, 10, 15, 30 min) captures initial phosphorylation events
Extended sampling (1, 2, 4, 8, 24 h) reveals feedback regulation and adaptation
Synchronized cell populations for cell-cycle dependent analyses
Inhibitor studies:
IKK inhibitors (e.g., BMS-345541, TPCA-1) to block phosphorylation
Proteasome inhibitors (MG132, bortezomib) to prevent degradation after phosphorylation
Kinase inhibitor panels to identify non-canonical regulation
Genetic manipulation approaches:
CRISPR/Cas9 modification of phosphorylation sites (S32A, S36A, S32/36A)
Inducible expression systems for wild-type vs. mutant NFKBIA
siRNA-mediated knockdown of regulatory components
Subcellular fractionation:
Separate analysis of cytoplasmic, nuclear, and membrane fractions
Tracking phosphorylation, ubiquitination, and degradation in distinct compartments
These comprehensive experimental designs enable mechanistic insights into NFKBIA regulation under physiological and pathological conditions.
Researchers frequently encounter several technical challenges when working with phospho-specific antibodies:
Loss of phosphorylation during sample preparation:
Solution: Immediate sample processing in buffers containing phosphatase inhibitors (sodium fluoride, sodium orthovanadate, β-glycerophosphate)
Maintain samples at 4°C during all processing steps
Consider rapid heat denaturation to inactivate phosphatases
Non-specific binding in immunoassays:
Solution: Optimize blocking conditions (5% BSA often superior to milk for phospho-epitopes)
Include phospho-blocking agents in washing buffers
Test different detergent concentrations (0.05-0.3% Tween-20) to reduce background
Epitope masking by interacting proteins:
Solution: Use denaturing conditions for Western blotting
Consider alternative fixation protocols for immunofluorescence
Test mild detergent treatments to disrupt protein-protein interactions
Antibody cross-reactivity with similar phospho-motifs:
Solution: Validate specificity with phospho-deficient mutants
Perform peptide competition assays with specific and non-specific phospho-peptides
Consider additional confirmatory approaches (mass spectrometry)
Signal variability between experiments:
Solution: Standardize lysate preparation and protein quantification
Include internal control samples across experiments
Normalize to total protein and use reference standards
Addressing these challenges systematically enhances data reliability and reproducibility in phosphorylation studies.
Differentiating phosphorylation changes from expression changes requires specialized experimental approaches:
Dual detection strategies:
Sequential probing of the same membrane with phospho-specific and total NFKBIA antibodies
Use different host species antibodies to allow simultaneous detection
Calculate phospho/total ratio to normalize for expression differences
Protein synthesis inhibition:
Pretreatment with cycloheximide to block new protein synthesis
Isolates phosphorylation changes from transcriptional/translational regulation
Time-course analysis to account for NFKBIA protein half-life (~30 minutes)
Transcriptional analysis:
Proteasomal inhibition studies:
Compare phosphorylation in presence/absence of proteasome inhibitors
Reveals whether apparent decreases in phospho-signal result from degradation
Can unmask phosphorylation events that normally lead to rapid protein turnover
Pulse-chase experimentation:
Metabolic labeling to track protein synthesis and degradation rates
Enables discrimination between regulated synthesis and targeted degradation
Particularly valuable for dynamic pathway components like NFKBIA
These approaches provide a comprehensive assessment of the complex interplay between NFKBIA phosphorylation, expression, and degradation.
Proximity Ligation Assay (PLA) offers significant advantages for studying NFKBIA phosphorylation:
Mechanism and advantages:
Utilizes antibody pairs that recognize different epitopes on the same protein
Employs oligonucleotide-conjugated secondary antibodies that, when in close proximity, enable rolling circle amplification
Each phosphorylated protein appears as a distinct fluorescent dot
Provides single-molecule resolution with exceptional sensitivity
Implementation for NFKBIA research:
Requires paired antibodies: one against phospho-S32 and another against a distant NFKBIA epitope
Enables visualization of rare phosphorylation events even in fixed tissue samples
Allows subcellular localization analysis of phosphorylation events
Facilitates quantification of phosphorylation levels at single-cell resolution
Quantification approaches:
Digital image analysis using specialized software (e.g., BlobFinder)
Automated counting of discrete fluorescent puncta per cell
Statistical comparison across experimental conditions
Advantages over conventional methods:
100-1000× higher sensitivity than traditional immunofluorescence
Eliminates false positives from non-specific antibody binding
Enables detection of endogenous protein modifications without overexpression
PLA technology represents a powerful approach for detailed spatial analysis of NFKBIA phosphorylation in complex biological samples.
NFKBIA phosphorylation plays a complex role in cellular senescence mechanisms:
Biphasic regulation in senescence:
Senescence-associated secretory phenotype (SASP):
Cell cycle regulation:
Genetic studies demonstrate that loss of NFKBIA promotes hyperproliferation and crypt hyperplasia in intestinal models
Intestinal epithelium-restricted knockout of NFKBIA leads to enrichment of genes responsible for cell cycle progression
This indicates that constitutive NF-κB activation in vivo regulates proliferation in a positive manner
Therapeutic implications:
Targeting NFKBIA phosphorylation could potentially modulate senescence phenotypes
Selective interference with SASP without affecting proliferative arrest might provide new approaches to age-related diseases
Understanding these connections provides insights into fundamental aging mechanisms and potential interventional strategies.
Mass spectrometry (MS) offers complementary advantages to antibody-based detection of NFKBIA phosphorylation:
Comprehensive phosphorylation profiling:
MS can simultaneously detect multiple phosphorylation sites on NFKBIA beyond S32
Enables discovery of novel, previously uncharacterized phosphorylation events
Provides stoichiometric information about phosphorylation levels
Experimental workflow integration:
Immunoprecipitation of NFKBIA followed by MS analysis
IMAC (Immobilized Metal Affinity Chromatography) enrichment of phosphopeptides
Parallel Reaction Monitoring (PRM) for targeted quantification of specific phosphosites
Advantages for complex studies:
No requirement for site-specific antibodies for novel phosphorylation sites
Ability to detect multiple modifications simultaneously (phosphorylation, ubiquitination, acetylation)
Identification of modification-dependent protein interactions
Limitations and mitigation strategies:
Lower sensitivity compared to antibody-based methods
Solution: Implement phosphopeptide enrichment strategies
Challenging quantification in complex samples
Solution: Use isotope-labeled standards for absolute quantification
Validation approach:
MS findings can guide development of new phospho-specific antibodies
Antibody-based methods can provide spatial information lacking in MS analysis
Combination provides both discovery and targeted validation capabilities
This integrative approach leverages the strengths of both technologies for comprehensive characterization of NFKBIA modifications and their functional consequences.