Phospho-RELA (Thr435) Antibody

Shipped with Ice Packs
In Stock

Product Specs

Form
Supplied at 1.0 mg/mL in phosphate buffered saline (without Mg2+ and Ca2+), pH 7.4, 150 mM NaCl, 0.02% sodium azide and 50% glycerol.
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery time may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery details.
Synonyms
Avian reticuloendotheliosis viral (v rel) oncogene homolog A antibody; MGC131774 antibody; NF kappa B p65delta3 antibody; nfkappabp65 antibody; NFkB p65 antibody; NFKB3 antibody; Nuclear factor kappaB antibody; Nuclear Factor NF Kappa B p65 Subunit antibody; Nuclear factor NF-kappa-B p65 subunit antibody; Nuclear factor of kappa light polypeptide gene enhancer in B cells 3 antibody; Nuclear factor of kappa light polypeptide gene enhancer in B-cells 3 antibody; OTTHUMP00000233473 antibody; OTTHUMP00000233474 antibody; OTTHUMP00000233475 antibody; OTTHUMP00000233476 antibody; OTTHUMP00000233900 antibody; p65 antibody; p65 NF kappaB antibody; p65 NFkB antibody; relA antibody; TF65_HUMAN antibody; Transcription factor NFKB3 antibody; Transcription factor p65 antibody; v rel avian reticuloendotheliosis viral oncogene homolog A (nuclear factor of kappa light polypeptide gene enhancer in B cells 3 (p65)) antibody; V rel avian reticuloendotheliosis viral oncogene homolog A antibody; v rel reticuloendotheliosis viral oncogene homolog A (avian) antibody; V rel reticuloendotheliosis viral oncogene homolog A, nuclear factor of kappa light polypeptide gene enhancer in B cells 3, p65 antibody
Target Names
Uniprot No.

Target Background

Function
NF-κB is a versatile transcription factor found in nearly all cell types. It acts as the endpoint of numerous signal transduction pathways, initiated by a wide array of stimuli associated with diverse biological processes. These include inflammation, immunity, differentiation, cell growth, tumorigenesis, and apoptosis. NF-κB is a homo- or heterodimeric complex, composed of proteins containing the Rel-like domain, such as RELA/p65, RELB, NFKB1/p105, NFKB1/p50, REL, and NFKB2/p52. The RELA-NFKB1 heterodimer is generally the most abundant complex. These dimers bind to κB sites in the DNA of their target genes. Each dimer exhibits distinct preferences for specific κB sites, displaying varying affinities and binding specificities. Different dimer combinations function as either transcriptional activators or repressors. For instance, the NF-κB heterodimers RELA-NFKB1 and RELA-REL act as transcriptional activators. NF-κB is regulated by various mechanisms involving post-translational modifications, subcellular compartmentalization, and interactions with cofactors or corepressors. NF-κB complexes remain inactive in the cytoplasm, bound to members of the NF-κB inhibitor (IκB) family. Upon activation, IκB is phosphorylated by IκB kinases (IKKs) in response to various stimuli. Subsequent degradation of IκB liberates the active NF-κB complex, which translocates to the nucleus. The inhibitory effect of IκB on NF-κB, through cytoplasmic retention, primarily occurs through interaction with RELA. RELA possesses a weak DNA-binding site, directly contributing to DNA binding within the NF-κB complex. Beyond its direct role as a transcriptional activator, RELA also modulates promoter accessibility to transcription factors, indirectly regulating gene expression. RELA associates with chromatin at the NF-κB promoter region through interaction with DDX1. RELA is essential for cytokine gene expression in T cells. The NF-κB homodimer RELA-RELA is believed to play a role in invasin-mediated activation of IL-8 expression. RELA is a crucial transcription factor regulating the interferon response during SARS-CoV-2 infection.
Gene References Into Functions
  1. These findings suggest that resveratrol induces chondrosarcoma cell apoptosis via a SIRT1-activated NF-κB (p65 subunit of NF-κB complex) deacetylation, exhibiting anti-chondrosarcoma activity in vivo. PMID: 28600541
  2. Enhanced IL-1β production by the v65Stop mutant is partially attributed to the induction of DNA binding and transcriptional activity of NF-κB. PMID: 30332797
  3. This study, utilizing integrative analysis of transcriptomic, metabolomic, and clinical data, proposes a model of GOT2 transcriptional regulation. In this model, cooperative phosphorylation of STAT3 and direct joint binding of STAT3 and p65/NF-κB to the proximal GOT2 promoter are crucial. PMID: 29666362
  4. These results delineate a novel role of MKRN2 in negatively regulating NF-κB-mediated inflammatory responses, in cooperation with PDLIM2. PMID: 28378844
  5. Compared to patients with NF-κB-94 ins/del ATTG ins/ins and ins/del, multiple myeloma patients with del/del exhibited the highest myeloma cell ratio. PMID: 30211233
  6. The riboflavin transporter-3 (SLC52A3) 5'-flanking regions contain NF-κB p65/Rel-B-binding sites, crucial for mediating SLC52A3 transcriptional activity in esophageal squamous cell carcinoma (ESCC) cells. PMID: 29428966
  7. Akirin-2 emerges as a novel biomarker for imatinib resistance. Targeting Akirin-2, NFκB, and β-catenin genes may offer a strategy to overcome imatinib resistance in chronic myeloid leukemia (CML). PMID: 29945498
  8. The NF-κB-94ins/del ATTG genotype might serve as a novel biomarker and potential therapeutic target for immune thrombocytopenia. PMID: 30140708
  9. Our findings suggest that melatonin may exert anti-tumor activities against thyroid carcinoma by inhibiting p65 phosphorylation and inducing reactive oxygen species. Melatonin's radiosensitizing properties may hold clinical benefits in thyroid cancer. PMID: 29525603
  10. The antiproliferative effect of lutein was mediated by activation of the NrF2/ARE pathway and blocking of the NF-κB signaling pathway. Lutein treatment reduced NF-κB signaling pathway-related NF-κB p65 protein expression. PMID: 29336610
  11. Furthermore, this study suggests that SNHG15 might be involved in the nuclear factor-κB signaling pathway, inducing the epithelial-mesenchymal transition process and promoting renal cell carcinoma invasion and migration. PMID: 29750422
  12. This revealed that the overexpression of p65 partially reversed SOX4 downregulation-induced apoptosis. In conclusion, our results demonstrate that inhibition of SOX4 significantly induced melanoma cell apoptosis via downregulation of the NF-κB signaling pathway, potentially offering a novel therapeutic approach for melanoma. PMID: 29767266
  13. Downregulation of HAGLROS may alleviate lipopolysaccharide-induced inflammatory injury in WI-38 cells by modulating the miR-100/NF-κB axis. PMID: 29673591
  14. Our observations suggest that the RelA-activation domain and multiple cofactor proteins function cooperatively to prime the RelA-DNA binding domain and stabilize the RelA:DNA complex in cells. PMID: 29708732
  15. Results indicate that MKL1 influences the chromatin structure of pro-inflammatory genes. Specifically, MKL1 defines histone H3K4 trimethylation landscape for NF-κB dependent transcription. PMID: 28298643
  16. This study examined the association of SIRT2 and the p53/NF-κB p65 signaling pathways in preventing high glucose-induced vascular endothelial cell injury. Results demonstrated that SIRT2 overexpression is linked to deacetylation of p53 and NF-κB p65, which inhibits high glucose-induced apoptosis and vascular endothelial cell inflammation response. PMID: 29189925
  17. In conclusion, the spindle cell morphology is induced by RelA activation (p-RelA S468) through IKKε upregulation in human herpesvirus 8 vFLIP-expressing EA hy926 cells. PMID: 30029010
  18. High P65 expression is associated with doxorubicin resistance in breast cancer. PMID: 29181822
  19. Reduced miR-138 expression enhanced the destruction of cartilage tissues among osteoarthritis patients, primarily through targeting p65. PMID: 28537665
  20. The present results indicate that vascular smooth proliferation is regulated by activation of the NF-κB p65/miR17/RB pathway. As NF-κB p65 signaling is activated during and is a master regulator of the inflammatory response, these findings may provide a mechanism for the excessive proliferation of vascular smooth muscle cells (VSMCs) under inflammation during vascular disorders. This research could identify novel targets for the treatment of vascular... PMID: 29115381
  21. Real-time PCR and western blotting revealed that Huaier extract decreased p65 and c-Met expression, while increasing IκBα expression. Conversely, paclitaxel increased p65 expression while reducing IκBα and c-Met expression. The molecular mechanisms may involve inhibition of the NF-κB pathway and c-Met expression. PMID: 29039556
  22. Ghrelin effectively suppressed TNF-α-induced inflammatory factors' (including ICAM-1, VCAM-1, MCP-1, and IL-1β) expression by inhibiting AMPK phosphorylation and p65 expression in both HUVEC and THP-1 cells. PMID: 28653238
  23. These data indicate that the MALAT1/miR146a/NF-κB pathway plays a key role in LPS-induced acute kidney injury (AKI), providing novel insights into the mechanisms of this therapeutic candidate for the treatment of the disease. PMID: 29115409
  24. Cytosolic AGR2 contributed to cell metastasis due to its stabilizing effect on p65 protein, which subsequently activated NF-κB and facilitated epithelial to mesenchymal transition (EMT). PMID: 29410027
  25. We provide evidence that S100A7 also inhibits YAP expression and activity through p65/NFκB-mediated repression of ΔNp63, and S100A7 represses drug-induced apoptosis via inhibition of YAP. PMID: 28923839
  26. This study demonstrates age-related reductions in serum IL-12 in healthy non-obese subjects. PMID: 28762199
  27. NF-κB p65 potentiated tumor growth by suppressing a novel target, LPTS. PMID: 29017500
  28. p65 siRNA retroviruses effectively suppressed the activation of the NFκB signal pathway. PMID: 28990087
  29. miR-215 facilitated HCV replication through inactivation of the NF-κB pathway by inhibiting TRIM22, providing a potential novel target for HCV infection. PMID: 29749134
  30. Acute inflammation after injury initiates essential regenerative signals, partially through NF-κB-mediated signaling that activates neural stem cells to reconstitute the olfactory epithelium. Loss of RelA in the regenerating neuroepithelium disrupts the homeostasis between proliferation and apoptosis. PMID: 28696292
  31. PAK5-mediated phosphorylation and nuclear translocation of NF-κB-p65 promote breast cancer cell proliferation in vitro and in vivo. PMID: 29041983
  32. While 3-methyladenine rescues cell damage, our data suggest that ischemia/reperfusion (I/R) promotes NF-κB p65 activity mediated by Beclin 1-mediated autophagic flux, thereby exacerbating myocardial injury. PMID: 27857190
  33. Taken together, these data indicate that up-regulation of ANXA4 leads to activation of the NF-κB pathway and its target genes in a feedback regulatory mechanism via the p65 subunit, resulting in tumor growth in gallbladder cancer (GBC). PMID: 27491820
  34. p65 is significantly upregulated in BBN-induced high invasive bladder cancers and human bladder cancer cell lines. Our studies have also uncovered a new PTEN/FBW7/RhoGDIα axis, responsible for the oncogenic role of RelA p65 in promoting human bladder cancer cell migration. PMID: 28772241
  35. p65 O-GlcNAcylation promotes lung metastasis of cervical cancer cells by activating CXCR4 expression. PMID: 28681591
  36. We demonstrated that pristimerin suppressed tumor necrosis factor α (TNFα)-induced IκBα phosphorylation, translocation of p65, and expression of NFκB-dependent genes. Furthermore, pristimerin decreased cell viability and clonogenic ability of uveal melanoma (UM) cells. A synergistic effect was observed in the treatment with pristimerin combined with vinblastine, a frontline therapeutic agent for UM. PMID: 28766683
  37. This study establishes p65 as a novel target of IMP3 in increasing glioma cell migration and highlights the significance of the IMP3-p65 feedback loop for therapeutic targeting in glioblastoma (GBM). PMID: 28465487
  38. High NF-κB p65 expression is associated with resistance to doxorubicin in breast cancer. PMID: 27878697
  39. In colon cancer cell migration, activin utilizes NFκB to induce MDM2 activity leading to the degradation of p21 in a PI3K-dependent mechanism. PMID: 28418896
  40. This research investigated melatonin's role in cell senescence, autophagy, sirtuin 1 expression, and acetylation of RelA in hydrogen peroxide-treated SH-SY5Y cells. PMID: 28295567
  41. The data demonstrate that miR-125b regulates nasopharyngeal carcinoma cell proliferation and apoptosis by targeting the A20/NF-κB signaling pathway. miR-125b acts as an oncogene, whereas A20 functions as a tumor suppressor. PMID: 28569771
  42. NF-κB physically interacts with FOXM1 and promotes transcription of the FOXM1 gene. NF-κB directly binds to the FOXM1 gene promoter. Silencing p65 attenuates FOXM1 and β-catenin expression. NF-κB activation is required for nuclear translocation of FOXM1 and β-catenin. FOXM1 and β-catenin positively regulate NF-κB. Knockdown of β-catenin and FOXM1 downregulates p65 protein and NF-κB-dependent reporter... PMID: 27492973
  43. Paclitaxel (PTX) treatment of THP-1 macrophages for 1 hour induced marked intranuclear translocation of NF-κB p65. Low-dose PTX inhibited the M2 phenotype and induced the M1 phenotype via TLR4 signaling, suggesting that low-dose PTX can alter the macrophage phenotype, while clinical doses can kill cancer cells. These results suggest that the anticancer effects of PTX are due to both its cytotoxic and immunomodulatory activities. PMID: 28440494
  44. Sphk1 induced NF-κB-p65 activation, increased expression of cyclin D1, shortened the cell division cycle, and thus promoted proliferation of breast epithelial cells. PMID: 27811358
  45. Expression of NF-κB/p65 has prognostic value in high-risk non-germinal center B-cell-like subtype diffuse large B-cell lymphoma. PMID: 28039454
  46. The NFKB1 -94 insertion/deletion ATTG polymorphism is associated with decreased risks for lung cancer, nasopharyngeal carcinoma, prostate cancer, ovarian cancer, and oral squamous cell carcinoma. PMID: 28039461
  47. PU.1 supports TRAIL-induced cell death by inhibiting RelA-mediated cell survival and inducing DR5 expression. PMID: 28362429
  48. EGF and TNFα cooperatively promoted the motility of hepatocellular carcinoma (HCC) cells primarily through NF-κB/p65-mediated synergistic induction of FN in vitro. These findings highlight the crosstalk between EGF and TNFα in promoting HCC and provide potential targets for HCC prevention and treatment. PMID: 28844984
  49. The Brd4 acetyllysine-binding protein of RelA is involved in the activation of polyomavirus JC. PMID: 27007123
  50. MUC1-C activates the NF-κB p65 pathway, promotes occupancy of the MUC1-C/NF-κB complex on the DNMT1 promoter, and drives DNMT1 transcription. PMID: 27259275

Show More

Hide All

Database Links

HGNC: 9955

OMIM: 164014

KEGG: hsa:5970

STRING: 9606.ENSP00000384273

UniGene: Hs.502875

Involvement In Disease
A chromosomal aberration involving C11orf95 is found in more than two-thirds of supratentorial ependymomas. Translocation with C11orf95 produces a C11orf95-RELA fusion protein. C11orf95-RELA translocations are potent oncogenes that probably transform neural stem cells by driving an aberrant NF-kappa-B transcription program (PubMed:24553141).
Subcellular Location
Nucleus. Cytoplasm.

Q&A

What is the biological significance of RelA (p65) Thr435 phosphorylation?

Thr435 phosphorylation provides a crucial regulatory mechanism for modulating NF-κB transcriptional activity in a gene-specific manner. Research has demonstrated that this post-translational modification affects the ability of RelA to interact with chromatin-modifying enzymes, particularly HDAC1 (histone deacetylase 1) . The phosphorylation state of Thr435 can significantly influence the expression of specific target genes rather than globally affecting all NF-κB-dependent genes. For example, the phosphomimetic T435D mutation has been shown to specifically enhance CXCL2 (CXC chemokine ligand 2) expression . This site-specific phosphorylation represents one of the mechanisms through which cells can fine-tune inflammatory responses by regulating a subset of NF-κB target genes.

How should I validate the specificity of a Phospho-RELA (Thr435) antibody for my research?

To validate the specificity of a Phospho-RELA (Thr435) antibody, implement the following methodological approach:

  • Phosphatase treatment control: Treat half of your sample with lambda phosphatase before immunoblotting to demonstrate phospho-specificity.

  • Blocking peptide competition: Perform parallel Western blots with antibody pre-incubated with phospho-peptide versus non-phospho-peptide .

  • Phosphomimetic mutants: Use cells expressing T435D (phosphomimetic) and T435A (phospho-null) RelA mutants as positive and negative controls .

  • Stimulus-dependent phosphorylation: Verify increased phospho-signal after TNFα treatment, as this has been shown to induce Thr435 phosphorylation .

  • Knockdown/knockout validation: Use RelA-null cells as negative controls to confirm antibody specificity .

Commercial antibodies should be validated with these approaches, as demonstrated in immunohistochemistry and Western blot analyses showing signal elimination when blocked with phospho-peptide .

What applications are most suitable for Phospho-RELA (Thr435) antibodies in research?

Based on validated applications, Phospho-RELA (Thr435) antibodies are most effectively employed in the following research applications:

ApplicationRecommended DilutionKey Considerations
Western Blot1:500-1:2000Most reliable for quantitative analysis
Immunohistochemistry1:100-1:300Effective for tissue localization studies
Immunoprecipitation1:200-500Useful for studying protein interactions
ELISA1:20000High sensitivity for quantitative detection
Chromatin Immunoprecipitation(based on optimization)Valuable for studying promoter binding

For ChIP applications, this antibody is particularly valuable for investigating how Thr435 phosphorylation affects RelA binding to specific gene promoters and the subsequent recruitment of transcriptional cofactors. When performing Western blot analysis, include both total RelA and phospho-specific antibodies to normalize phosphorylation levels to total protein expression .

How does Thr435 phosphorylation mechanistically affect RelA transcriptional activity?

The mechanistic impact of Thr435 phosphorylation on RelA transcriptional activity involves complex protein-protein interactions that affect chromatin remodeling:

  • HDAC1 Interaction: Mutation studies have demonstrated that Thr435 phosphorylation disrupts RelA interaction with HDAC1 in vitro . This disruption appears to be direct and specific to this phosphorylation site.

  • Histone Acetylation: ChIP analysis revealed that the T435D phosphomimetic mutation results in enhanced levels of histone acetylation at NF-κB target gene promoters . This is consistent with decreased HDAC1 recruitment to these regions.

  • Transcriptional Outputs: The effects are gene-specific, with the phosphomimetic T435D mutation significantly enhancing Cxcl2 mRNA levels in reconstituted Rela−/− mouse embryonic fibroblasts .

  • TAD Activity Modulation: When tested as a Gal4-TAD fusion protein, the phospho-null T435A mutation dramatically increased transcriptional activity, while the phosphomimetic T435D mutation decreased activity . This suggests that in the isolated TAD context, phosphorylation may repress transcriptional potential.

These seemingly contradictory findings between full-length RelA and the isolated TAD underscore the complex, context-dependent nature of this phosphorylation event in regulating transcriptional outcomes.

What experimental conditions optimize detection of TNFα-induced Thr435 phosphorylation?

Optimal detection of TNFα-induced Thr435 phosphorylation requires specific experimental conditions:

  • Cell Type Selection: Mouse embryonic fibroblasts (MEFs) show more robust and rapid Thr435 phosphorylation following TNFα stimulation compared to U-2 OS cells, which exhibit weaker induction . Consider using MEFs for initial studies.

  • TNFα Concentration and Timing: Based on published protocols, use 10-20 ng/ml TNFα and monitor phosphorylation between 5-60 minutes post-stimulation .

  • Phosphatase Inhibition: Include phosphatase inhibitors (e.g., calyculin A) in cell lysis buffers to preserve phosphorylation . The study demonstrates that calyculin A treatment results in increasing levels of phosphorylation at this site.

  • Immunoprecipitation Step: Direct Western blots may show background bands that mask the phospho-specific signal. An immunoprecipitation step with anti-RelA antibody followed by immunoblotting with the phospho-specific antibody can significantly enhance detection sensitivity .

  • Nuclear Fraction Enrichment: Since active RelA translocates to the nucleus, analyzing nuclear fractions can enhance detection of the phosphorylated form.

  • Sample Preparation: For optimal results, process samples rapidly on ice to prevent dephosphorylation by cellular phosphatases.

How can I investigate the relationship between Thr435 phosphorylation and gene-specific transcriptional responses?

To comprehensively investigate how Thr435 phosphorylation affects gene-specific transcriptional responses, implement the following experimental workflow:

  • Generate Stable Cell Lines: Establish Rela−/− cells reconstituted with wild-type RelA, T435A (phospho-null), or T435D (phosphomimetic) mutants as performed in the key study .

  • Transcriptional Profiling: Perform RNA-seq or targeted gene expression analysis of NF-κB target genes across these cell lines. Key genes to examine include:

    • Cxcl2 (demonstrated to be enhanced by T435D)

    • Cxcl1 (chemokine with similar regulation)

    • Tnfaip3 (A20, important negative feedback regulator)

  • ChIP Analysis: Conduct ChIP experiments using the following antibodies:

    • Anti-RelA

    • Anti-phospho-Thr435 RelA

    • Anti-HDAC1

    • Anti-acetyl histone H3 and H4

  • Promoter Analysis: Use the ChIP primers described in the study :

    • Cxcl1 (FWD 5′-CTAATCCTTGGGAGTGGAG-3′, REV 5′-CCCTTTTATGCTCGAAAC-3′)

    • Cxcl2 (FWD 5′-CGTGCATAAAAGGAGCTCTC-3′, REV 5′-GTGCCCGAGGAAGCTTGT-3′)

    • Tnfaip3 (FWD 5′-CGCTGAGAGAGAGACAAAC-3′, REV 5′-TGGCCCTGAAGATTAACT-3′)

  • Protein-Protein Interaction Studies: Perform co-immunoprecipitation experiments to assess how Thr435 phosphorylation affects RelA interactions with transcriptional cofactors beyond HDAC1.

This integrated approach will provide mechanistic insights into how this specific phosphorylation event contributes to gene-specific transcriptional regulation.

What kinases are responsible for Thr435 phosphorylation, and how can they be experimentally manipulated?

While the specific kinases responsible for Thr435 phosphorylation aren't explicitly identified in the provided search results, a methodological approach to identify and manipulate these kinases would include:

  • Kinase Prediction Analysis: Use bioinformatic tools to analyze the amino acid sequence surrounding Thr435 (TQAGEGT*LSEALC) for potential kinase consensus motifs.

  • Kinase Inhibitor Screening: Systematically test the effects of specific kinase inhibitors on TNFα-induced Thr435 phosphorylation. Focus on kinases known to be activated in the TNFα signaling pathway, including:

    • IKK complex members

    • MAP kinases (p38, JNK, ERK)

    • PKC family members

    • GSK3β

  • Kinase Overexpression/Knockdown: Overexpress constitutively active forms or perform siRNA knockdown of candidate kinases to assess their impact on Thr435 phosphorylation.

  • In Vitro Kinase Assays: Perform in vitro kinase assays using purified candidate kinases and RelA peptides containing the Thr435 site.

  • Phosphatase Studies: The research indicates that PP4 (protein phosphatase 4)-mediated Thr435 dephosphorylation enhances RelA-mediated activation following cisplatin treatment . This suggests that targeting PP4 could be an alternative approach to modulate Thr435 phosphorylation levels.

This systematic approach would help identify the relevant kinases and provide tools for experimental manipulation of Thr435 phosphorylation in various research contexts.

How does Thr435 phosphorylation compare to other phosphorylation sites on RelA in terms of functional outcomes?

RelA contains multiple phosphorylation sites that collectively create a complex regulatory network. A comparative analysis of Thr435 phosphorylation with other sites reveals:

Phosphorylation SiteKinase(s)Functional ImpactComparison to Thr435
Thr435Not specified in sourcesDisrupts HDAC1 interaction; Enhances specific gene expression (e.g., Cxcl2) -
Ser536IKKα/β, RSK1, TBK1Enhances transcriptional activity; Affects DNA bindingMore broadly activating than Thr435, which shows gene selectivity
Ser276PKA, MSK1/2Promotes CBP/p300 recruitment; Enhances activityOpposite effect to Thr435 regarding histone-modifying enzyme recruitment
Ser468GSK3β, IKKεPromotes both activation and termination of NF-κB responsesMore complex temporal regulation than Thr435
Ser529CKIIEnhances transcriptional activityLess gene-specific than Thr435

The research on Thr435 phosphorylation reveals a unique aspect of RelA regulation where phosphorylation at this site appears to have gene-specific effects rather than globally affecting RelA activity . This contrasts with some other sites (particularly Ser536 and Ser276) that tend to have broader effects on RelA transcriptional capacity.

The context-dependent nature of Thr435 phosphorylation is evident in the observation that a T435A mutation dramatically increases transcriptional activity when tested in the isolated TAD, yet the T435D phosphomimetic enhances specific gene expression in the context of full-length RelA . This suggests that Thr435 phosphorylation operates within a complex network of modifications that collectively determine RelA function in a gene-specific manner.

What are common pitfalls when working with Phospho-RELA (Thr435) antibodies and how can they be avoided?

When working with Phospho-RELA (Thr435) antibodies, researchers frequently encounter several challenges. Here are methodological solutions to common pitfalls:

  • Background Signal Issues:

    • Problem: Direct Western blots may show background bands that mask the phospho-specific signal .

    • Solution: Implement an immunoprecipitation step with anti-RelA antibody before immunoblotting with the phospho-specific antibody .

  • Phosphorylation Loss During Sample Processing:

    • Problem: Rapid dephosphorylation by cellular phosphatases.

    • Solution: Include phosphatase inhibitors (e.g., calyculin A) in all buffers and process samples rapidly on ice .

  • Cell Type Variability:

    • Problem: Variable detection of Thr435 phosphorylation across cell types.

    • Solution: The research indicates that MEFs show more robust phosphorylation than U-2 OS cells following TNFα stimulation . Select appropriate cell types for initial studies.

  • Antibody Cross-Reactivity:

    • Problem: Non-specific binding to other phosphorylated proteins.

    • Solution: Always validate specificity using blocking peptides and phospho-null mutants (T435A) .

  • Fixation-Induced Epitope Masking in IHC:

    • Problem: Formalin fixation can mask phospho-epitopes.

    • Solution: Optimize antigen retrieval methods; validated IHC protocols indicate successful detection in paraffin-embedded tissues .

  • Quantification Challenges:

    • Problem: Difficulty normalizing phospho-signal to total RelA.

    • Solution: Always run parallel blots or sequential probing for total RelA and calculate the phospho/total ratio.

What sample preparation methods best preserve Thr435 phosphorylation for different analytical techniques?

Optimal preservation of Thr435 phosphorylation requires technique-specific sample preparation methods:

TechniqueSample Preparation MethodCritical Considerations
Western BlotDirect lysis in buffer containing phosphatase inhibitors (sodium fluoride, sodium orthovanadate, calyculin A) Rapid processing on ice; avoid repeated freeze-thaw cycles
ImmunoprecipitationGentle cell lysis followed by RelA pulldown before phospho-detection Pre-clear lysates thoroughly to reduce background
IHCRapid fixation; optimal antigen retrieval Consider using phospho-blocking peptides as controls
ChIPProtein-DNA crosslinking with formaldehyde; sonication optimization Include phosphatase inhibitors in all buffers
Cell FractionationSeparate nuclear and cytoplasmic fractions with NP-40 bufferMaintain phosphatase inhibitors throughout fractionation

The research specifically demonstrates that immunoprecipitation with an anti-RelA antibody followed by immunoblotting with the phospho-specific antibody significantly enhances detection sensitivity compared to direct Western blots . This approach should be considered when working with challenging samples or when signal-to-noise ratio is problematic.

How might single-cell analysis techniques advance our understanding of RelA Thr435 phosphorylation dynamics?

Single-cell analysis presents transformative opportunities for studying the heterogeneity and dynamics of RelA Thr435 phosphorylation:

  • Single-Cell Phospho-Flow Cytometry:

    • Implementation of phospho-specific antibodies in flow cytometry would allow quantification of phosphorylation levels across thousands of individual cells.

    • This approach could reveal cell-to-cell variability in TNFα responses that may be masked in population-based analyses.

  • Single-Cell RNA-Seq Integration:

    • Combining phospho-flow with single-cell RNA-seq would allow correlation between Thr435 phosphorylation states and gene expression profiles at the single-cell level.

    • This could reveal subpopulations of cells with distinct phosphorylation states and corresponding transcriptional outputs.

  • Live-Cell Phosphorylation Sensors:

    • Development of FRET-based biosensors incorporating the RelA region around Thr435 would enable real-time visualization of phosphorylation dynamics in living cells.

    • This approach could reveal the temporal dynamics of Thr435 phosphorylation and dephosphorylation following TNFα stimulation with unprecedented resolution.

  • Spatial Proteomics:

    • Mass spectrometry imaging or proximity ligation assays could map the subcellular localization of phosphorylated RelA at Thr435.

    • This would provide insights into whether Thr435 phosphorylation occurs predominantly in specific nuclear regions or protein complexes.

These single-cell approaches would address limitations of current bulk analyses and potentially reveal new regulatory mechanisms governing the specificity of NF-κB transcriptional responses.

What therapeutic implications does targeting RelA Thr435 phosphorylation have for inflammatory diseases?

The gene-specific effects of Thr435 phosphorylation suggest potential therapeutic applications:

  • Selective Inflammatory Gene Modulation:

    • Since Thr435 phosphorylation specifically enhances expression of certain genes (e.g., Cxcl2) while not globally affecting NF-κB activity, targeting this modification could provide more selective anti-inflammatory effects compared to broad NF-κB inhibitors.

  • Identification of Pathway-Specific Inhibitors:

    • Targeting the kinases responsible for Thr435 phosphorylation could provide more selective therapeutic options than targeting upstream NF-κB pathway components.

    • Alternatively, inhibitors that disrupt the structural changes induced by Thr435 phosphorylation could be developed.

  • Biomarker Potential:

    • Phospho-Thr435 levels could serve as biomarkers for specific inflammatory states or disease progression.

    • The available antibodies could be adapted for clinical diagnostic applications.

  • Combined Epigenetic Therapy Approaches:

    • Since Thr435 phosphorylation affects HDAC1 recruitment and histone acetylation , combination therapies targeting both this phosphorylation site and histone-modifying enzymes might provide synergistic anti-inflammatory effects.

Future research should focus on identifying the specific inflammatory conditions where Thr435 phosphorylation plays a critical role, as this would determine the most promising therapeutic applications.

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.