Phospho-NFKBIA (Ser32/Ser36) Antibody

Shipped with Ice Packs
In Stock

Description

Biological Context and Functional Significance

NFKBIA (IκBα) inhibits NF-κB transcription factors by sequestering them in the cytoplasm. Upon cellular stimulation (e.g., cytokines, pathogens, or stress), IκBα undergoes phosphorylation at Ser32/Ser36, triggering its ubiquitination and proteasomal degradation. This releases NF-κB dimers (e.g., RELA/p65) to translocate into the nucleus and activate target genes .

Key phosphorylation mechanisms:

  • Kinases involved: Phosphorylation at Ser32/Ser36 is mediated by the IKK (IκB kinase) complex, including IKKα (CHUK), IKKβ (IKBKB), and regulatory subunits like NEMO (IKBKG) . Other kinases, such as TBK1, MAP3K14, and Aurora A (AURKA), also contribute .

  • Downstream effects: Phosphorylation primes NFKBIA for recognition by E3 ubiquitin ligases (e.g., SCFβ-TrCP), leading to polyubiquitination and degradation .

Western Blot (WB)

  • Detects phosphorylated IκBα (~39 kDa) in lysates from stimulated cells (e.g., TNFα-treated HeLa cells) .

  • Recommended dilutions: 1:300–1:5000 .

Immunohistochemistry (IHC)/Immunofluorescence (IF)

  • Localizes phospho-IκBα in tissue sections or cultured cells. Optimized for paraffin-embedded (IHC-P) or frozen (IHC-F) samples .

Flow Cytometry

  • Thermo Fisher’s monoclonal antibody (RILYB3R) enables intracellular staining for phospho-IκBα in immune cells .

Phosphorylation Sites

  • Ser32: Primarily targeted by IKKβ and TBK1 .

  • Ser36: Phosphorylated by IKKα and Aurora A .

Regulatory Interactions

  • Ubiquitination: Phosphorylated IκBα is recognized by β-TrCP, leading to K48-linked polyubiquitination .

  • Negative regulators: DDRGK1 stabilizes phosphorylated IκBα, delaying NF-κB activation .

Validation and Quality Control

  • Specificity tests: Blocking assays with phosphorylated peptides confirm target selectivity .

  • Batch consistency: Antibodies are affinity-purified using phosphopeptide-conjugated columns .

Product Specs

Form
Supplied at a concentration of 1.0 mg/mL in phosphate buffered saline (without Mg2+ and Ca2+), pH 7.4, containing 150 mM NaCl, 0.02% sodium azide, and 50% glycerol.
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. The delivery time may vary depending on the purchase method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
I kappa B alpha antibody; I-kappa-B-alpha antibody; IkappaBalpha antibody; IkB-alpha antibody; IKBA antibody; IKBA_HUMAN antibody; IKBalpha antibody; MAD 3 antibody; MAD3 antibody; Major histocompatibility complex enhancer-binding protein MAD3 antibody; NF kappa B inhibitor alpha antibody; NF-kappa-B inhibitor alpha antibody; NFKBI antibody; NFKBIA antibody; Nuclear factor of kappa light chain gene enhancer in B cells antibody; Nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor alpha antibody
Target Names
Uniprot No.

Target Background

Function
This antibody targets Phospho-NFKBIA (Ser32/Ser36), which plays a crucial role in regulating the activity of NF-kappa-B/REL complexes. It functions by trapping REL dimers in the cytoplasm, effectively masking their nuclear localization signals and preventing their translocation to the nucleus. Upon cellular stimulation by immune or proinflammatory responses, Phospho-NFKBIA (Ser32/Ser36) becomes phosphorylated, promoting ubiquitination and degradation. This process enables the dimeric RELA to translocate to the nucleus, where it activates transcription.
Gene References Into Functions
  1. This research demonstrates changes in NF-kB expression levels induced by cell-free DNA in various cell types. PMID: 29743966
  2. The results of real-time PCR and western blotting indicated that Huaier extract reduced p65 and c-Met expression while increasing IkappaBalpha expression. Conversely, paclitaxel increased p65 expression and decreased IkappaBalpha and c-Met expression. These findings suggest that the molecular mechanisms may involve the inhibition of the NF-kappaB pathway and c-Met expression. PMID: 29039556
  3. Colorectal cancer cases exhibited an increase in the frequency of the NFkappaBIA-881G allele among Egyptian subjects. PMID: 28389768
  4. This study investigates the association between polymorphisms and the progression of chronic hepatitis B Virus infection in the Chinese Han population. PMID: 29093318
  5. miR-668 was upregulated in radioresistant human breast cancer cell lines MCF-7R and T-47DR. This upregulation targeted IkappaBalpha, activating the NF-kappaB pathway and subsequently increasing the radioresistance of breast cancer cells. PMID: 28138801
  6. Pristimerin effectively suppressed tumor necrosis factor a (TNFalpha)-induced IkappaBa phosphorylation, translocation of p65, and expression of NFkappaB-dependent genes. Additionally, pristimerin reduced cell viability and clonogenic ability in Uveal melanoma (UM) cells. A synergistic effect was observed when pristimerin was combined with vinblastine, a frontline therapeutic agent for UM. PMID: 28766683
  7. These data collectively demonstrate the functional significance of IkappaBalpha-mediated stripping of NFkappaB from DNA in the kinetic control of NFkappaB signaling. PMID: 28167786
  8. The findings suggest that genetic polymorphisms of NFKB1A rs696, pre-miR-146a rs2910164, and pre-miR-499 rs3746444 may serve as novel markers of AT susceptibility. PMID: 28674224
  9. Combining XPO1 inhibitor therapy with bortezomib or carfilzomib induces nuclear localization of IkappaBalpha and overcomes acquired proteasome inhibitor resistance in human multiple myeloma. PMID: 27806331
  10. Molecular docking analysis indicated that transcription factor NF-kappaB is a potential molecular target modulated by DTTF. The drug effectively blocked the TNFalpha-induced phosphorylation of upstream IkappaBalpha kinase in a time-dependent manner, leading to the suppression of NF-kappaB activation and nuclear translocation. PMID: 27882436
  11. This research demonstrates that HOTAIR regulates NF-kB activation by decreasing Ik-Ba (NF-kB inhibitor). By inducing prolonged NF-kB activation and expression of NF-kB target genes during DNA damage, HOTAIR plays a critical role in cellular senescence and platinum sensitivity. PMID: 27041570
  12. The authors present amide hydrogen/deuterium exchange data revealing long-range allosteric changes in the NFkappaB (RelA-p50) heterodimer induced by DNA or IkappaBalpha binding. PMID: 28249778
  13. Sam68 is essential for DNA damage-induced NF-kappaB activation and colon tumorigenesis. PMID: 27458801
  14. Specifically, BCA2 functions as an E3 SUMO ligase in the SUMOylation of IkappaBalpha, which in turn enhances the sequestration of NF-kappaB components in the cytoplasm. Given that HIV-1 utilizes NF-kappaB to promote proviral transcription, the BCA2-mediated inhibition of NF-kappaB significantly decreases the transcriptional activity of HIV-1. PMID: 28122985
  15. This study found that NFKBIA mRNAs were significantly expressed in normal tissues compared to glioma specimens. PMID: 27538656
  16. The findings highlight the prognostic value of NFKB inhibitor alpha (NFKBIA) in lower-grade gliomas (LGGs). PMID: 27052952
  17. W346 effectively inhibited tumor necrosis factor (TNF-a)-induced NF-kappaB activation by suppressing IKK phosphorylation, inhibiting IkB-a degradation, and restraining the accumulation of NF-kappaB subunit p65 nuclear translocation. W346 also impacted NF-kappaB-regulated downstream products involved in cycle arrest and apoptosis. PMID: 26520440
  18. Treating cells with SZC014 resulted in a decrease in phosphorylation of IkBa and NF-kappaB/p65 and NF-kappaB/p65 nuclear translocation. The cytotoxic activities of seven OA derivatives were generally stronger than that of OA, with SZC014 exhibiting the most potent anticancer activity in SGC7901 cells, suggesting its potential as a promising chemotherapeutic agent for the treatment of gastric cancer. PMID: 26547583
  19. Network analysis identified NFKBIA as a pathogenic gene in childhood asthma. PMID: 27420950
  20. HMBA effectively increased prostratin-induced phosphorylation and degradation of NF-kappaB inhibitor IkappaBalpha, thereby enhancing and prolonging prostratin-induced nuclear translocation of NF-kappaB, a prerequisite for stimulating transcription initiation. PMID: 27529070
  21. Enhanced miR-381a-3p expression contributed to the injury of osteoarthritis primarily by inhibiting the expression of IkappaBalpha. PMID: 27312547
  22. The timely and efficient degradation of ubiquitinated IkappaB[alpha], along with the timely and efficient liberation of RelA from ubiquitinated IkappaB[alpha] and RelA nuclear translocation, fundamentally depends on the presence of functional p97/VCP. PMID: 26463447
  23. Activated Rac1 regulates the degradation of IkappaBalpha and the nuclear translocation of STAT3-NFkappaB complexes in starved cancer cells. PMID: 27151455
  24. A mutation in a Chinese patient resulted in mycobacterial infections without ectodermal dysplasia. PMID: 26691317
  25. DAT stabilized IkBa by inhibiting the phosphorylation of Ika by the IkB kinase (IKK) complex. DAT induced proteasomal degradation of TRAF6, and DAT suppressed IKKb-phosphorylation through downregulation of TRAF6. PMID: 26647777
  26. The rs3138053 polymorphism of the NFKBIA gene is a candidate for susceptibility to overall cancers, while rs696 plays a protective role [meta-analysis]. PMID: 26488500
  27. This study identifies a novel BCR-ABL/IkappaBalpha/p53 network, whereby BCR-ABL functionally inactivates a key tumor suppressor in chronic myeloid leukemia. PMID: 26295305
  28. The study demonstrates an association between functional polymorphisms of IkappaBalpha rs696 and smoking with the risk of defective spermatogenesis, suggesting an interaction between the NF-kappaB signaling pathway and smoking-related ROS in human spermatogenesis. PMID: 25352423
  29. This genetic variation is associated with susceptibility to acute kidney injury. PMID: 26477820
  30. MicroRNA-19a mediates gastric carcinoma cell proliferation through the activation of IkappaBalpha. PMID: 26239140
  31. No association was observed between NFKBIA variants and the risk of liver cancer. PMID: 24578542
  32. SM22alpha is a phosphorylation-regulated suppressor of IKK-IkappaBalpha-NF-kappaB signaling cascades. PMID: 25937534
  33. The data suggest that the activity of IKBalpha can be regulated by dietary factors. Dietary supplementation with luteolin inhibits vascular endothelial inflammation by suppressing IKBalpha/NFkappaB signaling. PMID: 25577468
  34. This review and meta-analysis of the association of NFKBIA -881 A>G polymorphism with cancer susceptibility reveals that the -881 A>G polymorphism may increase the risk of cancer development in Asian populations. PMID: 26252270
  35. miR-126 may play a significant role in hepatic fibrosis by downregulating the expression of IkappaBalpha, partly through the NF-kappaB signaling pathway. PMID: 25974152
  36. IkappaBetaalpha inhibits apoptosis at the outer mitochondrial membrane independently of NF-kappaB retention. PMID: 25361605
  37. The single nucleotide polymorphism rs1957106 CT and TT genotypes were found to be associated with lower NFKBIA protein levels and a poor prognosis in patients with glioblastoma. PMID: 25215581
  38. The data suggest that the NFKBIA 126 G/A polymorphism may be helpful in identifying liver transplant recipients with an increased susceptibility to develop recurrent acute rejections. PMID: 25112903
  39. Expression of IkappaBalpha in human bladder cancer cells is negatively correlated with epithelial-mesenchymal transition and tumor invasion in vitro. PMID: 25374080
  40. NFKBIA-rs2233419AG was associated with a significantly increased risk of developing recurrent wheezing. PMID: 25326706
  41. miR-196a can directly interact with IkappaBalpha 3'-UTR to suppress IkappaBalpha expression, subsequently promoting the activation of NF-kappaB. PMID: 24463357
  42. MiR-196a promotes pancreatic cancer progression by targeting nuclear factor kappa-B-inhibitor alpha. PMID: 24504166
  43. Following bortezomib treatment, there was accumulation of IkappaB-alpha (IkappaBalpha) without affecting its phosphorylation status at an early time point. PMID: 23697845
  44. This study reveals that polymorphisms in the IkB-alpha promoter (-881 A/G, -826 C/T) are strongly associated with the susceptibility of Iranian Multiple Sclerosis patients. PMID: 24368589
  45. The results of this study suggest that oligodendroglial IkappaBalpha expression and NF-kappaB are activated early in the course of MSA, and their balance contributes to the decision of cellular demise. PMID: 24361600
  46. No statistically significant CRC risk association was found for the NFKBIA polymorphisms. PMID: 23996241
  47. The analysis indicates that NFKBIA deletions are present, though not frequent, in Glioblastoma multiforme (GBM). These deletions become more frequent in GBM neurospheres (NS), an event that appears to be influenced by the presence of EGF in the culture medium. PMID: 24330732
  48. The analysis of IkappaBalpha expression at the salivary gland epithelial cell level could potentially serve as a new hallmark of Sjogren's syndrome progression. PMID: 23377923
  49. IkappaBalpha sequesters not only p65 but also RPS3 in the cytoplasm. PMID: 24457201
  50. NF-kB expression was downregulated, and its cytoplasmic inhibitor IKBalpha was increased in CTLA4-Ig treated macrophages. PMID: 24295474

Show More

Hide All

Database Links

HGNC: 7797

OMIM: 164008

KEGG: hsa:4792

STRING: 9606.ENSP00000216797

UniGene: Hs.81328

Involvement In Disease
Ectodermal dysplasia, anhidrotic, with T-cell immunodeficiency autosomal dominant (ADEDAID)
Protein Families
NF-kappa-B inhibitor family
Subcellular Location
Cytoplasm. Nucleus.

Q&A

What is NFKBIA and its role in the NF-κB signaling pathway?

NFKBIA (also known as IκBα) is a critical inhibitory protein in the NF-κB signaling pathway. It functions by sequestering NF-κB dimers in the cytoplasm through masking their nuclear localization signals (NLS), thereby keeping NF-κB in an inactive state. The protein is part of a regulatory feedback mechanism that ensures NF-κB activation is properly controlled and typically transient. When cells receive appropriate stimuli (inflammatory signals, growth factors, or T/B cell receptor engagement), the IκB kinase (IKK) complex phosphorylates IκBα at serines 32 and 36, leading to its ubiquitination and subsequent degradation by the 26S proteasome. This liberates NF-κB, allowing its translocation to the nucleus where it activates transcription of target genes, including NFKBIA itself, thereby creating a negative feedback loop .

Why is phosphorylation of NFKBIA at Ser32 and Ser36 specifically important?

The phosphorylation of NFKBIA (IκBα) at serine residues 32 and 36 represents a critical regulatory checkpoint in the canonical NF-κB signaling pathway. This specific dual-site phosphorylation creates a recognition signal for the β-TrCP ubiquitin ligase complex, which subsequently polyubiquitinates IκBα, marking it for proteasomal degradation. This process is specifically mediated by the IKK complex composed of IKKα, IKKβ, and NEMO (IKKγ). The highly conserved nature of these phosphorylation sites across species underscores their evolutionary significance in regulating NF-κB activation. Without phosphorylation at both Ser32 and Ser36, IκBα remains stable and continues to sequester NF-κB in the cytoplasm, preventing its transcriptional activity .

How does the NFKBIA-NF-κB relationship function as a regulatory circuit?

The NFKBIA-NF-κB relationship forms a sophisticated regulatory circuit based on negative feedback. When NF-κB is activated and translocates to the nucleus following IκBα degradation, it induces the transcription of numerous target genes, prominently including the NFKBIA gene itself. This newly synthesized IκBα protein enters the nucleus, binds to NF-κB, and shuttles it back to the cytoplasm, effectively terminating the transcriptional response. This self-regulatory circuit ensures that NF-κB activation is typically transient rather than sustained, unless there is continued stimulation or pathway dysregulation. Interestingly, in certain contexts such as cellular senescence, this regulatory circuit can be disrupted through mechanisms including altered phosphorylation of NF-κB family members (particularly phosphorylation of p65/RelA at Ser468), which can lead to transcriptional silencing of NFKBIA and subsequent constitutive activation of NF-κB independent of the classical IKK-mediated pathway .

What are the optimal applications for using Phospho-NFKBIA (Ser32/Ser36) antibodies in research?

Phospho-NFKBIA (Ser32/Ser36) antibodies are versatile research tools that excel in several experimental applications:

  • Western Blotting: The primary application for detecting phosphorylated IκBα, typically used at 1-2 μg/ml dilution. This method allows quantitative assessment of pathway activation by measuring phospho-IκBα levels relative to total IκBα.

  • Flow Cytometry: For analyzing phospho-IκBα levels at the single-cell level, particularly useful for heterogeneous cell populations. Pre-titrated antibodies can be used at approximately 5 μL (0.125 μg) per test for intracellular staining.

  • Immunoprecipitation: For isolating phosphorylated IκBα complexes to study associated proteins.

  • Immunofluorescence/Immunocytochemistry: For visualizing the subcellular localization of phosphorylated IκBα following stimulation.

Each application requires specific optimization of fixation, permeabilization, and detection methods to preserve the phospho-epitope and ensure specific binding .

How should positive controls be established for Phospho-NFKBIA (Ser32/Ser36) antibody experiments?

Establishing appropriate positive controls is essential for validating Phospho-NFKBIA (Ser32/Ser36) antibody experiments. A recommended protocol involves using extracts from Jurkat cells treated with a two-step stimulation:

  • Pre-treatment with 100 μg/ml ALLN (N-Acetyl-Leu-Leu-Norleucinal), a proteasome inhibitor, for 30 minutes. This prevents the rapid degradation of phosphorylated IκBα, allowing its accumulation.

  • Subsequent stimulation with 1 nM TNF-α, a potent activator of the canonical NF-κB pathway.

This treatment creates a positive control with significantly elevated levels of phosphorylated IκBα at Ser32/36. For comprehensive validation, parallel samples should be prepared:

  • Unstimulated cells (negative control)

  • ALLN-only treated cells (proteasome inhibition control)

  • TNF-α-only treated cells (stimulation control)

  • ALLN + TNF-α treated cells (positive control)

Western blot analysis should show strong phospho-IκBα signal in the positive control lane, with minimal signal in unstimulated cells .

What are the methodological considerations for detecting transient phosphorylation of NFKBIA?

Detecting the transient phosphorylation of NFKBIA presents several methodological challenges that researchers must address:

  • Timing of sample collection: The phosphorylation of IκBα at Ser32/36 is extremely rapid and transient, often peaking within 5-15 minutes of stimulation before the protein is degraded. Create a detailed time-course experiment with early time points (0, 2, 5, 10, 15, 30, 60 minutes) to capture the phosphorylation window.

  • Proteasome inhibition: To prevent rapid degradation of phosphorylated IκBα, pre-treat cells with proteasome inhibitors like ALLN (100 μg/ml) or MG132 (10 μM) for 30 minutes before stimulation.

  • Phosphatase inhibitors: Include phosphatase inhibitors (sodium orthovanadate, sodium fluoride, β-glycerophosphate) in all lysis buffers to prevent ex vivo dephosphorylation during sample preparation.

  • Rapid sample processing: Minimize the time between cell harvesting and protein denaturation to preserve phosphorylation status.

  • Quantification strategy: Always normalize phospho-IκBα signal to total IκBα and a loading control like GAPDH or β-actin. Consider using fluorescent secondary antibodies for more accurate quantification over a wider dynamic range .

How can Phospho-NFKBIA antibodies differentiate between the two phases of NF-κB activation in cellular senescence?

Phospho-NFKBIA (Ser32/Ser36) antibodies serve as critical tools for distinguishing between the mechanistically distinct phases of NF-κB activation observed during cellular senescence:

Phase 1 (Acute/Transient Activation):

  • IKK-dependent and proteasome-dependent

  • Characterized by high levels of phosphorylated IκBα (Ser32/36)

  • Associated with anti-apoptotic gene expression

  • Features prominent p65 phosphorylation at Ser536

Phase 2 (Chronic/Constitutive Activation):

  • IKK-independent and proteasome-independent

  • Features transcriptional silencing of NFKBIA

  • Associated with senescence-associated secretory phenotype (SASP)

  • Characterized by altered p65 phosphorylation status, particularly increased Ser468 phosphorylation

To effectively differentiate these phases using phospho-NFKBIA antibodies, researchers should implement a comprehensive experimental approach:

  • Conduct time-course experiments spanning both early (minutes to hours) and late (days) timepoints after senescence induction

  • Simultaneously measure:

    • Phospho-IκBα (Ser32/36) levels

    • Total IκBα protein levels

    • NFKBIA mRNA expression

    • NF-κB DNA binding activity

    • p65 phosphorylation status at multiple sites (Ser536, Ser276, Ser468)

During phase 1, researchers should observe elevated phospho-IκBα followed by decreased total IκBα, while in phase 2, both phospho-IκBα and total IκBα remain low despite continued NF-κB activity .

How does GSK3β-mediated phosphorylation affect NFKBIA regulation and how can this be experimentally validated?

GSK3β-mediated phosphorylation plays a crucial role in regulating NFKBIA through an indirect mechanism involving p65/RelA phosphorylation. To experimentally validate this regulatory pathway:

This experimental approach allows researchers to establish the causal link between GSK3β activity, p65 Ser468 phosphorylation, NFKBIA transcriptional repression, and sustained NF-κB activation in contexts like cellular senescence .

What experimental strategies can distinguish between IKK-dependent and IKK-independent mechanisms of NF-κB activation?

Distinguishing between IKK-dependent and IKK-independent mechanisms of NF-κB activation requires a multi-faceted experimental approach:

  • Pharmacological inhibition: Employ selective IKK inhibitors (e.g., BMS-345541, TPCA-1) and proteasome inhibitors (e.g., MG132, bortezomib) to determine pathway dependency. In IKK-dependent activation, both inhibitor classes should block NF-κB activation, while in IKK-independent mechanisms, only NF-κB inhibitors (not IKK/proteasome inhibitors) would be effective.

  • Genetic approaches: Utilize cells with genetic ablation or knockdown of IKK components (IKKα, IKKβ, NEMO) to validate dependency on the IKK complex. Similarly, employ cells expressing non-phosphorylatable IκBα mutants (S32A/S36A) to confirm bypass of the classical phosphorylation-dependent degradation pathway.

  • Temporal analysis of key markers: Monitor over time:

    • IKK activation (phospho-IKK)

    • IκBα phosphorylation (Ser32/36)

    • Total IκBα protein levels

    • p65 nucleocytoplasmic localization

    • NFKBIA mRNA expression

  • Nuclear extract analysis: Examine nuclear extracts for presence of IκBα-free NF-κB dimers despite the absence of IκBα phosphorylation/degradation.

  • Chromatin immunoprecipitation (ChIP): Assess NF-κB binding to target promoters, including the NFKBIA promoter, to determine whether transcriptional silencing of NFKBIA occurs during IKK-independent activation.

This comprehensive approach enables researchers to precisely delineate the mechanistic basis of NF-κB activation in different biological contexts, such as acute inflammatory responses versus cellular senescence .

What are the common technical issues when working with Phospho-NFKBIA antibodies and how can they be addressed?

IssuePossible CausesSolutions
Weak or absent signal- Sample degradation
- Insufficient phosphorylation
- Antibody deterioration
- Add phosphatase inhibitors to all buffers
- Optimize stimulation conditions
- Use fresh aliquots of antibody
- Confirm stimulation with positive control (e.g., TNF-α treated cells)
High background- Non-specific binding
- Insufficient blocking
- Secondary antibody cross-reactivity
- Increase blocking time (5% BSA in TBST recommended)
- Use more stringent washing
- Titrate antibody concentration
- Pre-adsorb secondary antibodies
Multiple bands- Cross-reactivity with related proteins
- Sample degradation
- Non-specific binding
- Use phospho-peptide competition assay
- Optimize lysis conditions (use RIPA buffer with protease/phosphatase inhibitors)
- Compare with knockout/knockdown samples
Inconsistent results- Variable phosphorylation kinetics
- Batch-to-batch antibody variation
- Sample handling differences
- Standardize stimulation protocol and timing
- Use the same lot number when possible
- Implement more rigorous standard operating procedures
Reduced antibody performance over time- Freeze-thaw cycles
- Inappropriate storage
- Aliquot antibody upon first use
- Store according to manufacturer recommendations (typically -20°C)
- Avoid repeated freeze-thaw cycles

For optimal results with phospho-specific antibodies, always validate new lots against a known positive control sample and consider including phospho-blocking peptides as specificity controls .

How can researchers validate the specificity of Phospho-NFKBIA (Ser32/Ser36) antibodies?

Validating the specificity of Phospho-NFKBIA (Ser32/Ser36) antibodies is critical for reliable research outcomes. A comprehensive validation approach includes:

  • Phospho-peptide competition assay: Pre-incubate the antibody with increasing concentrations of the phosphorylated peptide immunogen (containing phospho-Ser32/36) before application to samples. A genuine phospho-specific antibody will show dose-dependent signal reduction. Include non-phosphorylated peptide controls to confirm phospho-specificity.

  • Genetic validation: Compare antibody reactivity between:

    • Wild-type cells

    • Cells expressing non-phosphorylatable IκBα mutant (S32A/S36A)

    • NFKBIA knockout cells

  • Kinase inhibition: Treat cells with specific IKK inhibitors prior to stimulation. The phospho-signal should be significantly reduced or eliminated in inhibitor-treated samples.

  • Phosphatase treatment: Process duplicate samples with and without lambda phosphatase treatment. Phosphatase-treated samples should show complete loss of phospho-specific signal while retaining total IκBα signal.

  • Stimulus-response correlation: Demonstrate appropriate temporal dynamics following canonical NF-κB pathway activation (e.g., TNF-α treatment), showing rapid phosphorylation followed by protein degradation.

  • Mass spectrometry validation: For definitive confirmation, immunoprecipitate IκBα and perform mass spectrometry to identify phosphorylation specifically at Ser32/36 positions, correlating with antibody detection .

How does NFKBIA phosphorylation integrate with other post-translational modifications in NF-κB signaling?

NFKBIA (IκBα) phosphorylation at Ser32/36 operates within a complex network of post-translational modifications (PTMs) that collectively regulate NF-κB signaling:

  • Hierarchical modification sequence: Phosphorylation at Ser32/36 by IKK precedes and is required for lysine 21/22 ubiquitination by the SCF-βTrCP ubiquitin ligase complex, creating a sequential PTM cascade.

  • Regulatory phosphorylation sites: Beyond Ser32/36, IκBα contains additional regulatory phosphorylation sites:

    • Tyr42 phosphorylation (by Src family kinases) provides an alternative degradation pathway

    • C-terminal phosphorylation affects protein stability and function

    • Ser283/289 phosphorylation modulates IκBα-NF-κB binding

  • Cross-regulation with p65 modifications: The phosphorylation status of p65/RelA directly impacts NFKBIA regulation:

    • p65 Ser536 phosphorylation (mediated by IKK) enhances transcriptional activity

    • p65 Ser468 phosphorylation (partially mediated by GSK3β) can repress NFKBIA transcription

    • p65 Ser276 phosphorylation (by PKA and MSK1) enhances coactivator recruitment

  • Feedback and cross-talk mechanisms: The pathway integrates with multiple signaling networks:

    • ATM-PARP1-TRAF6-IKK cascade links DNA damage to NF-κB activation

    • GSK3β functions as a node connecting NF-κB signaling with Wnt pathway and senescence processes

This integrated network of PTMs creates multiple regulatory nodes that can be differentially targeted to modulate NF-κB signaling in different cellular contexts, particularly during transitions between acute and chronic activation states .

What is the relationship between NFKBIA phosphorylation and cellular senescence mechanisms?

The relationship between NFKBIA phosphorylation and cellular senescence involves a biphasic NF-κB activation pattern with distinct regulatory mechanisms:

Early Phase (Acute Response):

  • Triggered by DNA damage through the ATM-PARP1-TRAF6-IKK cascade

  • Characterized by canonical IKK-dependent phosphorylation of IκBα at Ser32/36

  • Results in proteasomal degradation of IκBα

  • Drives expression of anti-apoptotic genes

  • Self-limiting due to NF-κB-induced NFKBIA re-expression

Late Phase (Senescence-Associated):

  • Emerges days after senescence induction

  • Operates independently of IKK and proteasome activity

  • Features altered p65 phosphorylation, particularly increased Ser468 phosphorylation mediated partly by GSK3β

  • Results in transcriptional silencing of the NFKBIA gene

  • Leads to constitutive NF-κB activation

  • Drives expression of senescence-associated secretory phenotype (SASP) genes

During senescence, GSK3β exhibits increased kinase activity (related to downregulation of Wnt signaling and SAHF formation), contributing to p65 Ser468 phosphorylation. This modification represses NFKBIA transcription, thereby preventing IκBα synthesis and allowing sustained NF-κB activation despite the absence of ongoing IKK activity.

This mechanism represents a novel physiological mode of NF-κB activation with significant implications for understanding chronic inflammation, aging, and responses to genotoxic cancer treatments .

How can single-cell analysis of phospho-NFKBIA improve our understanding of NF-κB signaling heterogeneity?

Single-cell analysis of phospho-NFKBIA offers transformative potential for understanding the heterogeneity and dynamics of NF-κB signaling across cell populations:

  • Resolution of signaling heterogeneity: Flow cytometry-based detection of phospho-IκBα (Ser32/36) enables quantification of cell-to-cell variability in pathway activation, revealing subpopulations with distinct signaling states that would be masked in bulk analyses.

  • Correlation with cellular phenotypes: By combining phospho-IκBα detection with markers of cell state (proliferation, differentiation, senescence), researchers can map the relationship between NF-κB pathway activation status and specific cellular phenotypes.

  • Temporal dynamics at single-cell resolution: Time-course experiments can capture the asynchronous nature of NF-κB activation and reveal whether cells respond to stimuli in a digital (all-or-none) or analog (graded) manner.

  • Integration with single-cell transcriptomics: Emerging technologies like CITE-seq could potentially combine phospho-protein detection with single-cell RNA-seq, allowing direct correlation between IκBα phosphorylation status and transcriptional outputs.

  • Microfluidic approaches: Live-cell imaging of fluorescent reporters combined with microfluidic delivery of stimuli can track individual cell responses over time, revealing oscillatory behaviors and response thresholds.

For optimal implementation, researchers should consider:

  • Fixation methods that preserve phospho-epitopes while maintaining cellular integrity

  • Careful antibody validation for flow cytometry applications

  • Development of appropriate compensation controls when multiplexing

  • Statistical approaches for identifying and characterizing cell subpopulations .

What are the implications of NFKBIA regulation for developing targeted therapeutics for inflammatory and age-related diseases?

Understanding NFKBIA regulation offers several promising avenues for therapeutic development targeting inflammatory and age-related diseases:

  • Dual-phase targeting strategies: Based on the distinct phases of NF-κB activation, therapeutics could selectively target:

    • Acute phase: IKK inhibitors or proteasome modulators to block canonical pathway activation

    • Chronic/senescence phase: Compounds targeting the GSK3β-p65(Ser468)-NFKBIA transcriptional silencing axis

  • Senolytic approach: Therapeutics could specifically eliminate senescent cells with constitutive NF-κB activation driven by NFKBIA silencing, potentially reversing age-related tissue dysfunction.

  • SASP modulation: Rather than blocking all NF-κB activity, selectively inhibiting the transcriptional program driving the senescence-associated secretory phenotype while preserving essential NF-κB functions could reduce inflammation without compromising immunity.

  • Restoration of feedback mechanisms: Compounds that restore NFKBIA expression in contexts where it's transcriptionally silenced could reestablish normal regulatory feedback.

  • Phosphorylation-specific interventions: Development of compounds that specifically block IκBα phosphorylation at Ser32/36 without affecting other IKK substrates could provide more selective NF-κB inhibition.

These approaches offer the potential for more precise modulation of NF-κB signaling compared to current broad-spectrum inhibitors, potentially addressing conditions including chronic inflammation, neurodegenerative diseases, and age-related pathologies where dysregulated NF-κB activity contributes to disease progression .

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.