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 .
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
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
Significant remodeling of the actin cytoskeleton with cells appearing larger and displaying more intense actin staining
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
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
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
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
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)
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)
Stimuli that effectively induce RelA T505 phosphorylation include:
Cisplatin (shows approximately 2-fold enhancement after 12h treatment)
DNA replication stress inducers including:
Notably, TNFα, UV radiation, and topoisomerase inhibitors like etoposide and SN38 do NOT significantly induce T505 phosphorylation
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
Common challenges and solutions include:
| Challenge | Solution |
|---|---|
| Low signal-to-noise ratio | Use nuclear enrichment protocols; increase antibody concentration; enhance ECL detection system |
| High background | Optimize blocking (5% BSA); increase washing steps; dilute primary antibody |
| Inconsistent phosphorylation | Standardize treatment time; harvest cells at consistent cell cycle phase |
| Rapid dephosphorylation | Use fresh lysates; include phosphatase inhibitors in all buffers |
| Cross-reactivity with unphosphorylated RelA | Perform peptide competition controls; compare with T505A mutant samples |
| Variability between experiments | Include positive controls in each experiment; normalize to total RelA |
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:
Validate findings with reporter assays using promoters of identified target genes in cells expressing wild-type versus T505A RelA
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
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
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
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
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
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?