Phospho-RELA (T435) Antibody

Shipped with Ice Packs
In Stock

Description

Introduction and Overview

Phospho-RELA (T435) Antibody is a highly specific immunological reagent designed to detect and quantify the phosphorylated form of RELA (also known as p65 or NF-κB p65 subunit) at the threonine 435 residue. The RELA protein serves as a key component of the Nuclear Factor kappa B (NF-κB) signaling pathway, which plays crucial roles in inflammation, immune response, and cell survival mechanisms . The phosphorylation status at T435 specifically modulates NF-κB activity, making this antibody particularly valuable for researchers investigating disease mechanisms and developing targeted therapies .

The NF-κB complex typically exists as a heterodimer of p50 (NFKB1) and p65 (RELA), with the p50/p65 heterodimer representing the most abundant form of NF-κB in cells . Under normal conditions, this complex is sequestered in the cytoplasm through interaction with inhibitory proteins known as I-kappa-B proteins. Upon cellular stimulation, a series of signaling events leads to the phosphorylation and subsequent degradation of these inhibitory proteins, allowing the NF-κB complex to translocate to the nucleus and regulate gene expression .

Species Reactivity and Applications

The utility of Phospho-RELA (T435) Antibody extends across multiple experimental approaches and species models. Understanding its reactivity profile and recommended dilutions for various applications is essential for researchers planning their experiments.

Species Reactivity

Various commercial sources of Phospho-RELA (T435) Antibody have demonstrated reactivity against several species, making this antibody versatile for comparative studies across different model organisms .

SpeciesReactivity
HumanYes
MouseYes
RatYes
MonkeyYes (in some products)

Validated Applications and Recommended Dilutions

The antibody has been validated for multiple applications in molecular and cellular biology research :

ApplicationRecommended Dilution/Amount
Western Blot (WB)1:500 - 1:2000
Immunohistochemistry (IHC)1:50 - 1:300
Immunoprecipitation (IP)2-5 μg
ELISA1:20000
Immunofluorescence (IF)1:100 - 1:200

These applications enable researchers to detect and quantify Phospho-RELA (T435) in various experimental contexts, from protein expression analysis to spatial localization studies within tissues and cells .

Functional Significance of RELA T435 Phosphorylation

The phosphorylation of RELA at threonine 435 represents a critical regulatory mechanism within the NF-κB signaling pathway. Research has revealed specific functional consequences of this post-translational modification that impact cellular responses to various stimuli.

Regulation of Transcriptional Activity

The phosphorylation status of RELA at T435 has significant implications for NF-κB-mediated transcriptional regulation. According to research findings, T435 is located within the TA2 region of the Transactivation Domain (TAD) of RELA . Phosphorylation at this site is notably downregulated upon TNFα stimulation, which results in decreased interaction between RELA and histone deacetylase 1 (HDAC1) . This reduced interaction selectively enhances NF-κB-dependent gene expression, highlighting the importance of T435 phosphorylation in modulating specific transcriptional outcomes.

Role in Cisplatin Sensitivity

Research has demonstrated that protein phosphatase 4 (PP4) specifically dephosphorylates RELA at T435, which enhances cisplatin-induced NF-κB transcriptional activity . This enhancement reduces cellular resistance to cisplatin and increases its anticancer effects. Conversely, the maintenance of phosphorylation at T435 inhibits RELA transcriptional activation in response to cisplatin stimulation, potentially contributing to chemotherapy resistance mechanisms .

Implications in Neurological Conditions

Kim et al. observed increased RELA-T435 phosphorylation in endothelial cells expressing SMI-71 during severe vasogenic edema triggered by status epilepticus in the piriform cortex . The study demonstrated that neutralizing TNF-α through soluble TNF p55 receptor (sTNFp55R) infusion inhibited RELA T435 phosphorylation in endothelial cells, alleviating vasogenic edema and neuronal damage induced by status epilepticus . This finding suggests potential therapeutic strategies targeting T435 phosphorylation for certain neurological conditions.

When searching for this antibody in scientific literature or commercial catalogs, researchers may encounter various alternative names and synonyms :

  • Anti-Phospho-NF-kB p65/RelA-T435 Rabbit Polyclonal Antibody

  • Anti-Phospho-RELA-T435 antibody produced in rabbit

  • Phospho-NF-kB p65-T435 antibody

  • Anti-Phospho-N kappa-p65 (T435) RELA Antibody

Broader Context: Post-Translational Modifications of RELA

The phosphorylation of RELA at T435 represents just one of multiple post-translational modifications that regulate this protein's activity. Understanding the broader landscape of RELA modifications provides important context for interpreting research findings related to T435 phosphorylation.

Anrather et al. identified multiple phosphorylation sites on RELA, with varying degrees of phosphorylation at various serine residues . Their research suggested that differential phosphorylation at multiple sites could explain why RELA targets specific gene subsets in response to different cellular signals . Additional work by Lanucara et al. employed mass spectrometry to identify seven novel phosphorylation sites on RELA in neuroblastoma cells responding to TNF-α stimulation .

Beyond T435, other functionally significant phosphorylation sites on RELA include T464, which is crucial for mitochondrial regulation. Zhou et al. demonstrated that resveratrol activates RELA by phosphorylating T464 via protein kinase C activation of protein kinase G, promoting interaction between RELA and PGC-1a that affects mitochondrial content and hepatic steatosis .

Product Specs

Buffer
Liquid in PBS containing 50% glycerol, 0.5% BSA, and 0.02% sodium azide.
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for specific delivery time estimates.
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 pleiotropic transcription factor found in nearly all cell types. It serves as the endpoint of various signal transduction pathways initiated by a diverse range of stimuli associated with numerous biological processes, including inflammation, immunity, differentiation, cell growth, tumorigenesis, and apoptosis. NF-κB is a homo- or heterodimeric complex composed of Rel-like domain-containing proteins: RELA/p65, RELB, NFKB1/p105, NFKB1/p50, REL, and NFKB2/p52. The RELA-NFKB1 heterodimer is typically the most abundant complex. These dimers bind to κB sites within the DNA of their target genes. Each dimer exhibits unique preferences for different κB sites, binding with distinct affinities and specificities. Different dimer combinations can act as transcriptional activators or repressors. For instance, the NF-κB heterodimers RELA-NFKB1 and RELA-REL function as transcriptional activators. NF-κB is regulated by various mechanisms, including post-translational modification, subcellular compartmentalization, and interactions with other cofactors or corepressors. NF-κB complexes are retained in the cytoplasm in an inactive state, bound to members of the NF-κB inhibitor (IκB) family. In a conventional activation pathway, IκB is phosphorylated by IκB kinases (IKKs) in response to various activators. Subsequently, IκB is degraded, releasing the active NF-κB complex, which then translocates to the nucleus. IκB's inhibitory effect on NF-κB, through cytoplasmic retention, is primarily mediated by its interaction with RELA. RELA possesses a weak DNA-binding site that could contribute directly to DNA binding within the NF-κB complex. Beyond its role as a direct transcriptional activator, RELA can also modulate promoter accessibility to transcription factors, indirectly regulating gene expression. RELA associates with chromatin at the NF-κB promoter region through its association with DDX1. RELA is crucial for cytokine gene expression in T cells. The NF-κB homodimeric RELA-RELA complex appears to be involved in invasin-mediated activation of IL-8 expression. RELA is a key transcription factor regulating the interferon response during SARS-CoV-2 infection.
Gene References Into Functions
  1. These results suggest that resveratrol induces chondrosarcoma cell apoptosis via a SIRT1-activated NF-κB (p65 subunit of NF-κB complex) deacetylation and exhibits anti-chondrosarcoma activity in vivo. PMID: 28600541
  2. Enhanced IL-1β production by the v65Stop mutant is due in part to induction of DNA binding and the transcriptional activity of NF-κB. PMID: 30332797
  3. A study utilizing integrative analysis of transcriptomic, metabolomic, and clinical data proposes a model of GOT2 transcriptional regulation, in which the cooperative phosphorylation of STAT3 and direct joint binding of STAT3 and p65/NF-κB to the proximal GOT2 promoter are important. PMID: 29666362
  4. These results delineate a novel role of MKRN2 in negatively regulating NF-κB-mediated inflammatory responses, cooperatively with PDLIM2. PMID: 28378844
  5. Compared with patients with NF-κB-94 ins/del ATTG ins/ins and ins/del, multiple myeloma patients with del/del had the highest myeloma cell ratio. PMID: 30211233
  6. The riboflavin transporter-3 (SLC52A3) 5'-flanking regions contain NF-κB p65/Rel-B-binding sites, which are crucial for mediating SLC52A3 transcriptional activity in esophageal squamous cell carcinoma (ESCC) cells. PMID: 29428966
  7. Akirin-2 could be a novel biomarker in imatinib resistance. Targeting Akirin-2, NFkappaB and beta-catenin genes may provide an opportunity to overcome imatinib resistance in CML. PMID: 29945498
  8. NF-κB-94ins/del ATTG genotype might serve as a novel biomarker and potential target for immune thrombocytopenia. PMID: 30140708
  9. Our results suggest that melatonin may exert anti-tumor activities against thyroid carcinoma by inhibition of p65 phosphorylation and induction of reactive oxygen species. Radio-sensitization by melatonin may have clinical benefits in thyroid cancer. PMID: 29525603
  10. The effect of lutein antiproliferation was mediated by activation of the NrF2/ARE pathway, and blocking of the NF-κB signaling pathway. Lutein treatment decreased NF-κB signaling pathway related NF-κB p65 protein expression. PMID: 29336610
  11. Furthermore, the present study suggested that SNHG15 may be involved in the nuclear factorkappaB signaling pathway, induce the epithelial-mesenchymal transition process, and promote 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 demonstrated that inhibition of SOX4 markedly induced melanoma cell apoptosis via downregulation of the NF-κB signaling pathway, which thus may be a novel approach for the treatment of melanoma. PMID: 29767266
  13. Downregulation of HAGLROS may alleviate lipopolysaccharide-induced inflammatory injury in WI-38 cells via modulating 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 show that MKL1 influences the chromatin structure of pro-inflammatory genes. Specifically, MKL1 defined histone H3K4 trimethylation landscape for NF-κB dependent transcription. PMID: 28298643
  16. Studied association of SIRT2 and p53/NF-kB p65 signal pathways in preventing high glucose-induced vascular endothelial cell injury. Results demonstrated that SIRT2 overexpression is associated with deacetylation of p53 and NF-kB p65, which inhibits the high glucose induced apoptosis and vascular endothelial cell inflammation response. PMID: 29189925
  17. In conclusion, the spindle cell morphology should be induced by RelA activation (p-RelA S468) by IKKepsilon 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 the cartilage tissues among osteoarthritis patients, mainly through targeting p65. PMID: 28537665
  20. The present result indicated that vascular smooth proliferation is regulated by activation of the NF-κB p65/miR17/RB pathway. As NF-κB p65 signalling is activated in and is a master regulator of the inflammatory response, the present findings may provide a mechanism for the excessive proliferation of VSMCs under inflammation during vascular disorders and may identify novel targets for the treatment of vascular ... PMID: 29115381
  21. The results of real-time PCR and western blotting revealed that Huaier extract decreased p65 and c-Met expression and increased IkappaBalpha expression, while paclitaxel increased p65 expression and reduced IkappaBalpha and c-Met expression. The molecular mechanisms may be involved in the 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 through inhibiting AMPK phosphorylation and p65 expression both in HUVEC and THP-1. PMID: 28653238
  23. These data indicated that the MALAT1/miR146a/NF-κB pathway exerted key functions in LPS-induced acute kidney injury (AKI), and provided novel insights into the mechanisms of this therapeutic candidate for the treatment of the disease. PMID: 29115409
  24. Cytosolic AGR2 contributed to cell metastasis ascribed to its stabilizing effect on p65 protein, which subsequently activated the 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/NFkappaB-mediated repression of DeltaNp63, and S100A7 represses drug-induced apoptosis via inhibition of YAP. PMID: 28923839
  26. This study shows the age-related reductions in serum IL-12 in healthy nonobese subjects. PMID: 28762199
  27. NF-κB p65 potentiated tumor growth via suppressing a novel target LPTS. PMID: 29017500
  28. p65 siRNA retroviruses could suppress the activation of NFkappaB signal pathway. PMID: 28990087
  29. miR-215 facilitated HCV replication via inactivation of the NF-κB pathway by inhibiting TRIM22, providing a novel potential target for HCV infection. PMID: 29749134
  30. Acute inflammation after injury initiates important regenerative signals in part through NF-κB-mediated signaling that activates neural stem cells to reconstitute the olfactory epithelium; loss of RelA in the regenerating neuroepithelium perturbs the homeostasis between proliferation and apoptosis. PMID: 28696292
  31. PAK5-mediated phosphorylation and nuclear translocation of NF-κB-p65 promotes breast cancer cell proliferation in vitro and in vivo. PMID: 29041983
  32. While 3-methyladenine rescues cell damage. Our data thus suggest that I/R promotes NF-κB p65 activity mediated 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 GBC. PMID: 27491820
  34. p65 is significantly upregulated in BBN-induced high invasive BCs and human BC cell lines. Our studies have also uncovered a new PTEN/FBW7/RhoGDIα axis, which is responsible for the oncogenic role of RelA p65 in promotion of human BC cell migration. PMID: 28772241
  35. p65 O-GlcNAcylation promotes lung metastasis of cervical cancer cells by activating CXCR4 expression. PMID: 28681591
  36. We showed that pristimerin suppressed tumor necrosis factor α (TNFα)-induced IκBa phosphorylation, translocation of p65, and expression of NFκB-dependent genes. Moreover, pristimerin decreased cell viability and clonogenic ability of Uveal melanoma (UM) cells. A synergistic effect was observed in the treatment of pristimerin combined with vinblastine, a frontline therapeutic agent, in UM. PMID: 28766683
  37. This study establishes p65 as a novel target of IMP3 in increasing glioma cell migration and underscores the significance of IMP3-p65 feedback loop for therapeutic targeting in 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 NFkB to induce MDM2 activity leading to the degradation of p21 in a PI3K dependent mechanism. PMID: 28418896
  40. Studied 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 A20/NF-κB signaling pathway, and miR-125b acts as oncogene, whereas A20 functions as tumor suppressor. PMID: 28569771
  42. NF-κB physically interacts with FOXM1 and promotes transcription of FOXM1 gene. NF-κB directly binds FOXM1 gene promoter. Silencing p65 attenuates FOXM1 and beta-catenin expression. NF-κB activation is required for nuclear translocation of FOXM1 and beta-catenin. FOXM1 and beta-catenin positively regulate NF-κB. Knockdown of beta-catenin and FOXM1 downregulates p65 protein and NF-κB-dependent reporte... PMID: 27492973
  43. PTX treatment of THP-1 macrophages for 1 h 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, whereas clinical doses can kill cancer cells. These results suggest that the anticancer effects of PTX are due both to 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. NFKB1 -94insertion/deletion ATTG polymorphism 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 HCC cells mainly 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 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 Thr435 phosphorylation?

Thr435 phosphorylation represents a key regulatory mechanism that modulates the transcriptional activity of the RelA (p65) subunit of NF-κB. This phosphorylation occurs within the C-terminal transactivation domain (TAD) of RelA and has been demonstrated to have gene-specific effects on transcription . Experimental evidence using mutational analysis reveals that phosphorylation at this site can both enhance and repress transcriptional activity depending on the target gene context. For instance, a T435D phosphomimetic mutant significantly enhances Cxcl2 (CXC chemokine ligand 2) mRNA levels in reconstituted Rela −/− mouse embryonic fibroblasts .

The phosphorylation at Thr435 provides an additional mechanism for modulating the specificity of NF-κB transcriptional activity in cells. Mechanistically, this modification appears to alter the interaction between RelA and transcriptional co-regulators, particularly histone deacetylase 1 (HDAC1), thereby affecting chromatin remodeling at target gene promoters . This phosphorylation event thus represents a sophisticated mechanism for fine-tuning inflammatory and immune responses at the transcriptional level.

How is Phospho-RELA (T435) detected in experimental settings?

Detection of Phospho-RELA (T435) relies primarily on phospho-specific antibodies that have been validated to recognize this specific modification. Several approaches can be employed:

  • Western Blotting: Phospho-RELA (T435) can be detected via western blot using specific antibodies. Due to background bands, immunoprecipitation with an anti-RelA antibody followed by western blotting with the phospho-specific antibody often yields clearer results . The observed molecular weight is typically around 58-60 kDa .

  • Immunoprecipitation: When background signal is problematic, immunoprecipitation with a general RelA antibody followed by probing with the phospho-specific antibody improves detection .

  • Chromatin Immunoprecipitation (ChIP): Phospho-RELA (T435) antibodies can be used in ChIP assays to detect the presence of phosphorylated RelA at specific promoters. Researchers have used this approach with primers for genes like Cxcl1, Cxcl2, and Tnfaip3 .

  • Immunohistochemistry: Phospho-RELA (T435) antibodies have been validated for immunohistochemistry on paraffin-embedded tissues, enabling visualization of the phosphorylated protein in tissue sections .

For optimal results, stimulation with TNFα or phosphatase inhibitors like calyculin A enhances phosphorylation levels and improves detection .

What are the practical considerations for antibody selection and validation?

When selecting and validating Phospho-RELA (T435) antibodies, researchers should consider:

  • Antibody Specificity: Ensure the antibody specifically recognizes the phosphorylated form of Thr435. ELISA analysis with phospho- and non-phospho-peptides can confirm this specificity . Commercially available antibodies are typically raised against synthetic phosphopeptides with sequences surrounding Thr435 (e.g., E-G-T(p)-L-S) .

  • Host Species and Reactivity: Available antibodies are predominantly rabbit polyclonal with reactivity to human, mouse, and rat RELA . This cross-reactivity enables comparative studies across species.

  • Validation Methods: Look for antibodies validated through multiple applications (Western blot, ELISA, IHC-P) and with positive controls such as TNFα-treated cells or phosphatase inhibitor-treated samples .

  • Working Dilutions: Typical recommended dilutions for Western blotting range from 1:500 to 1:2000 .

  • Positive Controls: NIH/3T3 cells or HeLa cells treated with TNFα are commonly used as positive controls for antibody validation .

The inclusion of appropriate controls is critical - comparisons between untreated and TNFα+CA (calyculin A) treated samples can verify the specificity and functionality of the antibody .

How does Thr435 phosphorylation affect RelA interactions with transcriptional machinery?

Thr435 phosphorylation significantly alters RelA's protein-protein interactions with transcriptional regulators, particularly those involved in chromatin remodeling. Research indicates that:

  • HDAC1 Interaction: Mutation of Thr435 disrupts RelA interaction with histone deacetylase 1 (HDAC1) in vitro . This suggests that phosphorylation at this site modulates HDAC1 recruitment to NF-κB target gene promoters.

  • Histone Acetylation Levels: Chromatin immunoprecipitation analysis reveals that the T435D phosphomimetic mutation results in enhanced levels of histone acetylation, consistent with decreased recruitment of HDAC1 . This mechanistically explains how this phosphorylation can affect chromatin accessibility and gene expression.

  • Transcriptional Consequences: The effects on transcription are gene-specific. For instance, a T435A phospho-null mutation dramatically increases transcriptional activity in reporter assays, while the T435D phosphomimetic mutation decreases activity . This suggests that phosphorylation at this site may generally repress the transcriptional potential of the RelA TAD, but with context-dependent outcomes.

  • Domain-Specific Effects: When investigating exclusively TAD-dependent effects using Gal4-DBD fusions with the RelA TAD, the T435A mutation significantly enhances transcriptional activity while T435D decreases it . This confirms that the modifications directly affect TAD functionality.

These findings collectively demonstrate that Thr435 phosphorylation serves as a molecular switch that modulates RelA's ability to interact with transcriptional co-regulators, particularly those involved in histone modifications.

What experimental approaches can resolve temporal dynamics of Thr435 phosphorylation?

The temporal dynamics of Thr435 phosphorylation present specific experimental challenges that require sophisticated approaches:

  • Time-Course Stimulation: Studies show different temporal patterns depending on cell type. In U-2 OS cells, TNFα induces weak phosphorylation, while in MEF cells, rapid Thr435 phosphorylation is detected following TNFα stimulation . Experiments should include multiple time points (e.g., 5, 10, and 20 minutes post-stimulation) to capture the transient nature of this modification .

  • Phosphatase Inhibition: Treatment with serine/threonine phosphatase inhibitors like calyculin A results in increasing levels of phosphorylation at Thr435 . This approach can help stabilize the phosphorylation for detection purposes.

  • Combined Immunoprecipitation and Western Blotting: Due to background bands in direct Western blots, immunoprecipitation with an anti-RelA antibody followed by probing with the phospho-specific antibody provides clearer visualization of temporal dynamics .

  • Promoter-Specific ChIP Analysis: Thr435 phosphorylation of promoter-bound RelA can be detected at specific NF-κB target genes following TNFα treatment. Using primers for genes like Cxcl1, Cxcl2, and Tnfaip3 allows for monitoring of gene-specific temporal patterns .

  • Quantitative Phosphoproteomics: Mass spectrometry-based approaches can provide unbiased assessment of phosphorylation dynamics, although this requires appropriate enrichment strategies for phosphopeptides.

When designing temporal studies, it's important to consider that different cell types may exhibit distinct phosphorylation kinetics in response to the same stimulus .

How does Thr435 phosphorylation integrate with other post-translational modifications of RelA?

RelA undergoes numerous post-translational modifications that collectively form a complex regulatory code. Understanding how Thr435 phosphorylation integrates with other modifications requires consideration of:

  • Modification Crosstalk: Phosphorylation of sites within the TAD of RelA leads to both increased and decreased levels of transcriptional activity, with the precise effect dependent on context and gene target . The interplay between Thr435 phosphorylation and other modifications (phosphorylation, acetylation, methylation, ubiquitination) remains an area requiring further investigation.

  • Phosphorylation-Dephosphorylation Cycles: PP4 (protein phosphatase 4)-mediated Thr435 dephosphorylation was previously proposed to enhance RelA-mediated activation following cisplatin treatment, suggesting that phosphorylation at this site can negatively affect RelA activity in specific contexts . This highlights the importance of considering both kinases and phosphatases in the regulatory cycle.

  • Functional Consequences: While some modifications enhance transcriptional activity, others may repress it or affect protein stability and degradation . Creating a comprehensive map of how these modifications interact requires systematic mutation studies and mass spectrometry analysis.

  • Spatial Considerations: RelA localizes to both cytoplasm and nucleus , and different modifications may occur preferentially in different cellular compartments. Fractionation studies combined with phospho-specific detection can help resolve these spatial dynamics.

  • Stimulus-Specific Responses: Different stimuli (TNFα, cisplatin, etc.) may induce distinct patterns of post-translational modifications . Comparative studies across multiple stimuli can help elucidate these patterns.

Understanding this modification "code" remains challenging but is essential for developing targeted interventions in NF-κB-related diseases.

What are the technical considerations for using Phospho-RELA (T435) antibodies in chromatin immunoprecipitation?

Chromatin immunoprecipitation (ChIP) with Phospho-RELA (T435) antibodies requires specific technical considerations:

  • Primer Design: For successful ChIP experiments, researchers have used specific primers for NF-κB target genes:

    • 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′

  • Antibody Specificity and Validation: Due to the transient nature of this phosphorylation, antibody specificity is critical. Control experiments should include IgG controls, input controls, and ideally, comparisons with ChIP using general RelA antibodies .

  • Stimulus Conditions: TNFα treatment induces Thr435 phosphorylation of promoter-bound RelA at NF-κB target genes . Optimizing stimulus conditions (concentration, timing) is essential for successful ChIP experiments.

  • Sequential ChIP: To determine if RelA phosphorylated at Thr435 is associated with specific co-factors or histone modifications, sequential ChIP (re-ChIP) approaches may be employed, immunoprecipitating first with anti-RelA and then with anti-phospho-Thr435 antibodies.

  • Quantification Methods: qPCR is typically used to quantify ChIP results, requiring careful normalization against input DNA and selection of appropriate reference genes or regions .

These technical considerations are essential for generating reliable and reproducible data when investigating the genomic binding patterns of Phospho-RELA (T435).

How can Phospho-RELA (T435) antibodies be utilized in studying disease mechanisms?

Phospho-RELA (T435) antibodies offer valuable tools for investigating disease mechanisms, particularly in conditions involving dysregulated inflammation and immune responses:

  • Cancer Research: These antibodies have been successfully used in cancer studies, including breast carcinoma tissue analysis via immunohistochemistry . The NF-κB pathway plays crucial roles in cancer-related inflammation and cell survival, making this phosphorylation potentially relevant to cancer progression.

  • Inflammatory Diseases: Given the role of NF-κB in inflammatory processes, studying Thr435 phosphorylation may provide insights into conditions like rheumatoid arthritis, inflammatory bowel disease, and asthma. The gene-specific effects of this modification on inflammatory mediators like Cxcl2 are particularly relevant .

  • Mechanistic Studies: In disease models, these antibodies can help elucidate how signaling pathways converge on NF-κB regulation. For example, researchers have used these antibodies in studies of oxidative stress, as demonstrated in work by Tian Li et al. using MLE-12 cells .

  • Therapeutic Response Monitoring: Since NF-κB is a target for various therapeutic interventions, monitoring Thr435 phosphorylation could potentially serve as a biomarker for treatment efficacy.

  • Tissue-Specific Analyses: The antibodies' validated use in immunohistochemistry enables analysis of tissue-specific patterns of RelA phosphorylation in disease states .

When designing disease-focused studies, researchers should consider the cell type-specific and stimulus-specific nature of Thr435 phosphorylation, as these patterns may vary considerably across different pathological contexts .

What are the optimal conditions for inducing and detecting Thr435 phosphorylation?

Optimizing experimental conditions for reliable detection of Thr435 phosphorylation requires attention to several factors:

  • Stimulation Protocols:

    • TNFα treatment: Effective concentrations range from 5 ng/ml to 20 ng/ml, with phosphorylation detectable as early as 5 minutes post-stimulation in some cell types .

    • Phosphatase inhibition: Treatment with calyculin A (a serine/threonine phosphatase inhibitor) results in increasing levels of phosphorylation at Thr435 and can be used to enhance detection .

  • Cell Type Considerations:

    • MEF cells show rapid Thr435 phosphorylation following TNFα stimulation .

    • U-2 OS cells show weak induction of phosphorylation at this site following TNFα stimulation .

    • HeLa cells treated with TNFα show detectable phosphorylation and serve as good positive controls .

    • NIH/3T3 cells are also used as positive controls for antibody validation .

  • Detection Methods:

    • Direct Western blotting may show background bands that can mask the specific signal .

    • Immunoprecipitation with anti-RelA antibody followed by Western blotting with the phospho-specific antibody yields clearer results .

    • For Western blotting, recommended antibody dilutions range from 1:500 to 1:2000 .

  • Sample Preparation:

    • Protein loading amount of approximately 30 μg is recommended for Western blot detection .

    • Primary antibody incubation conditions: overnight at 4°C for optimal results .

  • Controls:

    • Untreated versus TNFα+CA (calyculin A) treated samples provide important controls .

    • Time-course experiments (5, 10, 20 minutes post-stimulation) help capture the transient nature of the phosphorylation .

These optimized conditions ensure reliable detection of Thr435 phosphorylation across different experimental settings.

How can mutational analysis be used to study the functional significance of Thr435 phosphorylation?

Mutational analysis provides critical insights into the functional significance of Thr435 phosphorylation:

  • Phospho-Null and Phospho-Mimetic Mutations:

    • T435A (phospho-null) mutation: Replacing threonine with alanine prevents phosphorylation at this site.

    • T435D (phospho-mimetic) mutation: Replacing threonine with aspartic acid mimics constitutive phosphorylation .

  • Expression Systems:

    • Full-length RelA mutants: Allow investigation of effects in the context of the complete protein.

    • TAD-fusion proteins with the DNA-binding domain (DBD) of Gal4: Enable isolation of TAD-dependent effects .

    • Reconstitution in Rela −/− mouse embryonic fibroblasts: Provides a clean genetic background for functional studies .

  • Functional Readouts:

    • Reporter assays: T435A mutation dramatically increases transcriptional activity, while T435D mutation decreases activity in U-2 OS cells .

    • Gene expression analysis: T435D phosphomimetic mutant significantly enhances Cxcl2 mRNA levels in reconstituted Rela −/− MEFs .

    • Chromatin immunoprecipitation: T435D mutation results in enhanced levels of histone acetylation associated with decreased recruitment of HDAC1 .

  • Protein Interaction Studies:

    • In vitro binding assays: Mutation of Thr435 disrupts RelA interaction with HDAC1 .

    • Co-immunoprecipitation: Can be used to validate altered interactions in cellular contexts.

  • Experimental Design Considerations:

    • Gene-specific effects: Mutations may have different effects depending on the target gene context .

    • Cell type-specific effects: Responses may vary across different cell types .

This systematic mutational approach has revealed that Thr435 phosphorylation modulates RelA function in a context-dependent manner, affecting both its transcriptional activity and protein interactions.

What are the emerging areas of research involving Phospho-RELA (T435)?

Several promising research directions are emerging in the study of Phospho-RELA (T435):

  • Integration with Single-Cell Technologies: Applying single-cell approaches to study cell-to-cell variation in Thr435 phosphorylation patterns could reveal heterogeneity in NF-κB signaling responses that bulk analyses miss.

  • Structural Biology Insights: Resolving how Thr435 phosphorylation alters the three-dimensional structure of RelA could provide mechanistic insights into its effects on protein-protein interactions and DNA binding.

  • Kinase and Phosphatase Networks: While PP4 has been implicated in Thr435 dephosphorylation , the specific kinases responsible for this phosphorylation remain to be fully characterized. Mapping these regulatory networks would provide new intervention points.

  • Therapeutic Targeting: Given the gene-specific effects of Thr435 phosphorylation, developing strategies to selectively modulate this modification could offer more precise approaches to targeting NF-κB in disease.

  • Cross-Talk with Non-Canonical NF-κB Pathways: Investigating how Thr435 phosphorylation interfaces with non-canonical NF-κB signaling could reveal additional regulatory mechanisms.

  • Tissue-Specific Regulation: Since differential phosphorylation patterns have been observed across cell types , exploring tissue-specific regulation of Thr435 phosphorylation could help explain context-dependent NF-κB functions.

  • Roles in Chronic Inflammation: Given the importance of NF-κB in inflammatory processes, understanding how sustained alterations in Thr435 phosphorylation contribute to chronic inflammatory conditions represents an important research direction.

These emerging areas highlight the continuing importance of Phospho-RELA (T435) antibodies as tools for advancing our understanding of NF-κB signaling complexity.

What are the limitations of current Phospho-RELA (T435) antibodies and potential solutions?

Current Phospho-RELA (T435) antibodies face several limitations that researchers should consider:

  • Background Signal: Direct Western blotting often shows background bands that can mask the specific signal .

    • Solution: Immunoprecipitation with anti-RelA antibody before Western blotting with phospho-specific antibody improves signal clarity .

  • Transient Nature of Phosphorylation: The transient nature of Thr435 phosphorylation can make detection challenging.

    • Solution: Use of phosphatase inhibitors like calyculin A can stabilize the phosphorylated state .

  • Cell Type Variability: Different cell types show variable patterns of Thr435 phosphorylation in response to the same stimulus .

    • Solution: Thorough validation in each cell type of interest and optimization of stimulation protocols.

  • Specificity Concerns: Ensuring absolute specificity for the phosphorylated form remains challenging.

    • Solution: Two-step peptide affinity chromatography with phospho- and non-phospho-peptides during antibody purification improves specificity .

  • Limited Applications: Not all commercially available antibodies are validated for all potential applications.

    • Solution: Comprehensive validation across multiple applications (WB, ELISA, IHC-P, ChIP) would expand utility .

  • Reproducibility Issues: Batch-to-batch variation in polyclonal antibodies can affect reproducibility.

    • Solution: Development of monoclonal antibodies or recombinant antibodies could improve consistency.

  • Cross-Reactivity: Some antibodies show cross-reactivity with related phosphorylation sites.

    • Solution: Careful validation using phospho-null mutants (T435A) as negative controls.

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.