Phospho-RELA (T505) Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
The antibody is provided as a liquid solution in PBS containing 50% glycerol, 0.5% BSA and 0.02% sodium azide.
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchase method or location. Please consult your local distributor for specific delivery times.
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 present in nearly all cell types. It serves as the endpoint of a series of signal transduction events initiated by a wide array of stimuli related to 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 heterodimeric RELA-NFKB1 complex appears to be the most abundant. These dimers bind to κB sites in the DNA of their target genes. Individual dimers demonstrate distinct preferences for various κB sites, exhibiting differing binding affinities and specificities. Different dimer combinations function as either transcriptional activators or repressors. For instance, the NF-κB heterodimeric RELA-NFKB1 and RELA-REL complexes act as transcriptional activators. NF-κB is subject to regulation by various mechanisms of post-translational modification and subcellular compartmentalization, as well as by interactions with other cofactors or corepressors. NF-κB complexes remain in an inactive state within the cytoplasm, bound to members of the NF-κB inhibitor (I-κB) family. In a typical activation pathway, I-κB undergoes phosphorylation by I-κB kinases (IKKs) in response to various activators. This phosphorylation leads to I-κB degradation, liberating the active NF-κB complex, which then translocates to the nucleus. The inhibitory effect of I-κB on NF-κB, through its retention in the cytoplasm, is primarily exerted through its interaction with RELA. RELA exhibits a weak DNA-binding site, which can contribute directly to DNA binding within the NF-κB complex. Beyond its function as a direct transcriptional activator, RELA can also modulate promoter accessibility to transcription factors, indirectly regulating gene expression. It associates with chromatin at the NF-κB promoter region through an association with DDX1. RELA is crucial for cytokine gene expression in T-cells. The NF-κB homodimeric RELA-RELA complex seems to be involved in invasin-mediated activation of IL-8 expression. RELA serves as a key transcription factor regulating the IFN response during SARS-CoV-2 infection.
Gene References Into Functions
  1. These findings suggest that resveratrol induces chondrosarcoma cell apoptosis through 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 partially attributed to the 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, where the cooperative phosphorylation of STAT3 and the 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 collaboration 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 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, which are essential 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, NFκB, and β-catenin genes may provide an opportunity to overcome imatinib resistance in CML. PMID: 29945498
  8. The NF-κB-94ins/del ATTG genotype may serve as a novel biomarker and potential 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. Radio-sensitization by melatonin might have clinical benefits in thyroid cancer. PMID: 29525603
  10. The antiproliferative effect of lutein was mediated by activation of the NrF2/ARE pathway and the blocking of the NF-κB signaling pathway. Lutein treatment decreased NF-κB signaling pathway-related NF-κB p65 protein expression. PMID: 29336610
  11. Furthermore, this study suggested that SNHG15 might be involved in the nuclear factor-κB 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 significantly induced melanoma cell apoptosis via downregulation of the NF-κB signaling pathway, which could potentially serve as a novel approach for the treatment of 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 defined histone H3K4 trimethylation landscape for NF-κB dependent transcription. PMID: 28298643
  16. This study investigated the 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 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 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 indicated that vascular smooth proliferation is regulated by activation of the NF-κB p65/miR17/RB pathway. As NF-κB p65 signaling is activated in and is a master regulator of the inflammatory response, these 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 IκBα expression, while paclitaxel increased p65 expression and reduced IκBα and c-Met expression. The molecular mechanisms may involve 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 by inhibiting AMPK phosphorylation and p65 expression in both 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 attributed 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 highlights the age-related reductions in serum IL-12 in healthy nonobese subjects. PMID: 28762199
  27. NF-κB p65 potentiated tumor growth by suppressing a novel target LPTS. PMID: 29017500
  28. p65 siRNA retroviruses could suppress the activation of the NFκB signaling 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 following injury initiates important regenerative signals, in part through NF-κB-mediated signaling, which 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 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 promoting 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 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 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 the 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. This study 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, where miR-125b acts as an oncogene, while 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. 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 -94insertion/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 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 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 T505 phosphorylation?

RelA (p65) phosphorylation at threonine 505 acts as a negative regulator of NF-κB function. Unlike other phosphorylation events that positively regulate NF-κB activity, T505 phosphorylation suppresses its ability to induce diverse cellular processes including apoptosis, autophagy, proliferation, and migration . This phosphorylation occurs during S-phase of the cell cycle and in response to specific DNA-damaging agents like cisplatin, providing a mechanism of crosstalk between NF-κB signaling and DNA replication stress . In vivo studies have confirmed that RelA T505 phosphorylation provides an important physiological regulatory mechanism that antagonizes and limits aspects of RelA function associated with tumor-promoting activities .

How does RelA T505 phosphorylation differ from other RelA phosphorylation sites?

RelA T505 phosphorylation stands apart from other phosphorylation sites in several key aspects:

  • It functions as a negative regulator of NF-κB activity, whereas most other sites (like S536) positively regulate NF-κB function

  • It is specifically induced by cisplatin and other DNA replication stress inducers, but not by TNFα or other common NF-κB activators

  • It is mediated by Chk1 kinase rather than IKK family kinases that phosphorylate many other RelA sites

  • T505 phosphorylated RelA associates with HDAC1 corepressor complexes rather than CBP coactivator complexes found with S468 phosphorylated RelA

  • Its phosphorylation peaks during S-phase of the cell cycle, whereas S536 phosphorylation is highest in G2 phase

What phenotypic changes are associated with disruption of RelA T505 phosphorylation?

Mutation of the RelA T505 residue to alanine (preventing phosphorylation) results in:

  • Enhanced resistance to cisplatin and other DNA replication stress-inducing agents

  • Increased cell proliferation and migration in cell culture models

  • Aberrant hepatocyte proliferation following liver partial hepatectomy or damage in mouse models

  • Earlier onset of hepatocellular carcinoma in the N-nitrosodiethylamine mouse model

  • Reduced survival in the Eμ-Myc mouse model of B-cell lymphoma

  • Resistance to CHK1 inhibitors in cancer models

  • Significant remodeling of the actin cytoskeleton with cells appearing larger and displaying more intense actin staining

What are the optimal conditions for detecting phospho-RelA T505 in Western blots?

For optimal detection of phospho-RelA T505 in Western blots:

  • Use fresh cell lysates prepared with phosphatase inhibitors to prevent dephosphorylation

  • The recommended dilution range is 1:500-1:2000 for most commercially available antibodies

  • Blocking should be performed with 5% BSA in TBST rather than milk (which contains phosphatases)

  • Use positive controls such as cisplatin-treated cells, which show approximately 2-fold enhancement of T505 phosphorylation after 12 hours of treatment

  • Consider enriching nuclear fractions since phospho-T505 RelA is primarily nuclear

  • Include appropriate loading controls and total RelA antibody on parallel blots to normalize phospho-signal

How can I validate the specificity of a phospho-RelA T505 antibody?

Validating phospho-RelA T505 antibody specificity requires:

  • Comparing signal in wild-type cells versus rela -/- cells reconstituted with T505A mutant RelA, which should show significantly reduced signal

  • Performing peptide competition assays using phospho-T505 peptide versus non-phosphorylated peptide

  • Treating samples with lambda phosphatase to confirm phosphorylation-dependent signal

  • Testing induction with known stimuli (e.g., cisplatin) versus non-inducing stimuli (e.g., TNFα)

  • Using phospho-ELISA with both phosphorylated and non-phosphorylated peptides covering the T505 region to demonstrate specificity

  • For immunohistochemistry applications, blocking with the phospho-peptide should eliminate specific staining as demonstrated in human breast carcinoma samples

What are the key differences between various commercial phospho-RelA T505 antibodies?

While multiple vendors offer phospho-RelA T505 antibodies, they differ in several aspects:

  • Host species (typically rabbit)

  • Clonality (most are polyclonal though some monoclonal options exist)

  • Validated applications (most support WB and ELISA, some support IHC)

  • Cross-reactivity (most react with human, mouse, and rat species)

  • Immunogen design (synthetic phosphopeptides around T505 position, but exact sequence may vary)

  • Storage buffer composition (typically PBS with glycerol, BSA and sodium azide)

  • Recommended dilutions for different applications

What cell types and experimental models are most appropriate for studying RelA T505 phosphorylation?

Based on published research, the following models are particularly suitable:

  • MEF cells (both wild-type and rela -/- reconstituted with wild-type or T505 mutant RelA)

  • Human U-2 OS osteosarcoma cells, which show inducible T505 phosphorylation after cisplatin treatment

  • Mouse models of hepatocellular carcinoma, where T505A mutation shows clear phenotypes

  • Eμ-Myc mouse model of B-cell lymphoma, which demonstrates the role of T505 in cancer progression

  • Cell cycle synchronized cultures to study the S-phase specific phosphorylation

  • Systems with active DNA replication stress, as T505 phosphorylation is particularly relevant in this context

How can I effectively study the functional impact of RelA T505 phosphorylation?

To study functional impacts of T505 phosphorylation:

  • Generate T505A (phospho-deficient) and T505D (phospho-mimetic) mutants of RelA

  • Reconstitute rela -/- cells with these mutants for comparative studies

  • Measure functional outcomes including:

    • Apoptotic response to cisplatin (annexin staining, caspase 3 activation)

    • Cell proliferation rates (MTS assay, cell cycle analysis by FACS)

    • Migration capacity (wound-healing assay with mitomycin C to block proliferation)

    • Actin cytoskeleton organization (phalloidin staining)

    • Gene expression changes using qPCR or RNA-seq (focusing on genes like NOXA, Bcl-xL, WAVE3, α-actinin 4)

  • Use ChIP assays to examine RelA binding to target gene promoters in T505A vs wild-type cells

  • Perform ReChIP experiments to identify T505 phosphorylation-dependent cofactor recruitment (e.g., HDAC1)

What stimuli effectively induce RelA T505 phosphorylation in experimental settings?

Stimuli that effectively induce RelA T505 phosphorylation include:

  • Cisplatin (shows approximately 2-fold enhancement after 12h treatment)

  • p14 ARF tumor suppressor induction

  • Cell synchronization in S-phase

  • DNA replication stress inducers including:

    • Mitomycin C (DNA cross-linking agent)

    • Hydroxyurea (ribonucleotide reductase inhibitor)

  • Notably, TNFα, UV radiation, and topoisomerase inhibitors like etoposide and SN38 do NOT significantly induce T505 phosphorylation

How should I interpret changes in RelA T505 phosphorylation in the context of cancer studies?

When interpreting changes in RelA T505 phosphorylation in cancer:

  • Increased phosphorylation generally suggests activation of negative regulatory mechanisms that may suppress tumor-promoting NF-κB activities

  • Reduced phosphorylation may indicate loss of this tumor-suppressive mechanism, potentially contributing to enhanced oncogenic NF-κB activity

  • Context matters: in some cancers, RelA T505 phosphorylation correlates with resistance to specific chemotherapeutics

  • Consider RelA T505 phosphorylation alongside other markers of DNA replication stress (γH2AX, phospho-RPA2)

  • Changes should be interpreted within the larger context of ATR/CHK1 pathway activation, especially when considering response to CHK1 inhibitors

  • In mouse models, loss of T505 phosphorylation (T505A mutation) accelerates cancer progression in both liver cancer and B-cell lymphoma models

What are common challenges in detecting RelA T505 phosphorylation and how can they be overcome?

Common challenges and solutions include:

ChallengeSolution
Low signal-to-noise ratioUse nuclear enrichment protocols; increase antibody concentration; enhance ECL detection system
High backgroundOptimize blocking (5% BSA); increase washing steps; dilute primary antibody
Inconsistent phosphorylationStandardize treatment time; harvest cells at consistent cell cycle phase
Rapid dephosphorylationUse fresh lysates; include phosphatase inhibitors in all buffers
Cross-reactivity with unphosphorylated RelAPerform peptide competition controls; compare with T505A mutant samples
Variability between experimentsInclude positive controls in each experiment; normalize to total RelA

How can phospho-RelA T505 data be correlated with gene expression changes?

To correlate phospho-RelA T505 with gene expression:

  • Perform ChIP-seq with phospho-T505 specific antibodies to identify genome-wide binding sites

  • Use ReChIP techniques to identify co-factor recruitment differences between phosphorylated and non-phosphorylated RelA

  • Correlate binding with RNA-seq data to identify genes specifically regulated by T505-phosphorylated RelA

  • Focus on key genes known to be regulated in a T505-dependent manner:

    • Anti-apoptotic genes (e.g., Bcl-xL) are often repressed by T505 phosphorylation

    • Pro-apoptotic genes (e.g., NOXA) are induced in a T505-dependent manner

    • Migration-associated genes (WAVE3, α-actinin 4, cortactin, N-Wasp, integrin-α5) show increased expression when T505 phosphorylation is prevented

  • Validate findings with reporter assays using promoters of identified target genes in cells expressing wild-type versus T505A RelA

How does RelA T505 phosphorylation integrate with the DNA damage and replication stress response pathways?

RelA T505 phosphorylation represents a critical node connecting NF-κB signaling with DNA damage and replication stress responses:

  • T505 is phosphorylated by Chk1, a key kinase activated during the DNA replication checkpoint

  • This phosphorylation occurs primarily during S-phase when DNA replication is active

  • It forms part of a complex regulatory network where different phosphorylated forms of RelA control cell cycle progression

  • In S-phase, Akt (which normally activates IKK) becomes inactivated, while Chk1 becomes activated and phosphorylates RelA

  • This phosphorylation influences RelA's interaction with p100/p52 (NF-κB2), which plays a role in regulating key cell cycle genes

  • Mutation of RelA T505 disrupts the DNA replication stress response and leads to resistance to CHK1 inhibitors

  • This suggests a feedback loop where CHK1 modulates NF-κB activity through T505 phosphorylation, which in turn affects sensitivity to CHK1 inhibition

What is the potential therapeutic relevance of targeting pathways involving RelA T505 phosphorylation?

The therapeutic relevance of RelA T505 phosphorylation includes:

  • Predicting response to CHK1 inhibitors: tumors with defective T505 phosphorylation show resistance to these agents

  • Combination therapies: DNA-damaging agents that induce T505 phosphorylation (like cisplatin) might synergize with agents that depend on intact T505 phosphorylation

  • Targeting the Chk1-RelA axis: developing drugs that enhance T505 phosphorylation might help suppress tumor-promoting NF-κB activities

  • Biomarker potential: T505 phosphorylation status could serve as a biomarker for predicting response to certain chemotherapeutics

  • Personalized medicine approaches: tumors with mutations affecting the T505 pathway might require alternative treatment strategies

  • Novel drug development: understanding the structural changes induced by T505 phosphorylation could lead to drugs that mimic these effects

How can multi-omics approaches enhance our understanding of RelA T505 phosphorylation networks?

Multi-omics approaches can provide comprehensive insights by:

  • Integrating phosphoproteomics data to identify other proteins modified in response to conditions that induce T505 phosphorylation

  • Combining ChIP-seq of phospho-T505 RelA with RNA-seq to create comprehensive maps of direct and indirect gene targets

  • Using proteomics to identify interaction partners specific to phospho-T505 RelA versus unphosphorylated RelA

  • Applying metabolomics to understand how T505 phosphorylation affects cellular metabolism

  • Conducting single-cell analyses to determine cell-to-cell variability in T505 phosphorylation and its consequences

  • Performing time-course studies to map the temporal dynamics of signaling networks after induction of T505 phosphorylation

  • Using structural biology approaches to understand how T505 phosphorylation alters RelA conformation and protein-protein interactions

What recent discoveries have changed our understanding of RelA T505 phosphorylation?

Recent advances include:

  • Identification of RelA T505 as a critical regulator of the DNA replication stress response in vivo

  • Discovery that RelA T505A mutation confers resistance to CHK1 inhibitors, suggesting a feedback relationship

  • Finding that T505 phosphorylation regulates the expression of migration-associated genes and cytoskeletal organization

  • Demonstration that RelA T505 phosphorylation regulates autophagy, expanding its known cellular functions

  • Identification of NOXA as a key T505-dependent effector in cisplatin-induced cell death

  • Evidence that T505 phosphorylation influences RelA's interactions with chromatin modifiers like HDAC1

  • Confirmation of the physiological significance of T505 phosphorylation in multiple mouse models of cancer

What methodological advances are improving the study of site-specific RelA phosphorylation?

Methodological advances include:

  • Development of highly specific phospho-antibodies for different RelA modification sites

  • CRISPR-Cas9 technology enabling precise genomic editing of endogenous RelA phosphosites

  • Improved mass spectrometry approaches for quantitative phosphoproteomics

  • Single-cell techniques to study heterogeneity in RelA phosphorylation

  • Proximity labeling methods to identify phosphorylation-specific interaction partners

  • Live-cell imaging with phospho-specific sensors to track dynamic changes in RelA modification

  • Enhanced computational methods to predict functional consequences of specific phosphorylation events

  • Development of nuclear isolation protocols that preserve phosphorylation status

What are the key outstanding questions regarding RelA T505 phosphorylation?

Critical unresolved questions include:

  • What is the three-dimensional structural impact of T505 phosphorylation on RelA conformation?

  • How does T505 phosphorylation affect RelA's interaction with other NF-κB family members?

  • Are there tissue-specific differences in the regulation and consequences of T505 phosphorylation?

  • How does T505 phosphorylation interact with other post-translational modifications of RelA?

  • What is the clinical significance of T505 phosphorylation status in human cancers?

  • Could targeting the T505 phosphorylation pathway represent a viable cancer therapy approach?

  • What role does T505 phosphorylation play in non-cancer pathologies involving NF-κB?

  • How is the T505 phosphorylation pathway affected by aging and cellular senescence?

  • What are the epigenetic consequences of altered T505 phosphorylation?

  • Do germline or somatic mutations affecting the T505 residue occur in human diseases?

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.