The antibody is validated for multiple techniques:
Abcam (EP1809Y): Observed 90 kDa band in HepG2 lysates; phosphatase treatment abolishes signal .
Boster (P00078): 100 kDa band in human cell lines (HepG2, HeLa, SiHa) .
Creative Biolabs (2E6): 90 kDa band in HepG2 and 293 cells treated with Calyculin A/EGF .
Abcam (EP1809Y): Strong nuclear staining in breast carcinoma; absent in non-atypical epithelium .
Boster (P00078): Detected in mouse/rat kidney tissue (with caveats about species reactivity) .
Abcam (EP1809Y): Confocal imaging shows cytoplasm-to-nucleus translocation in dh404-treated cells .
Creative Biolabs (2E6): Nuclear localization in HeLa cells stained with Alexa Fluor® 488 .
Phosphorylation at S40 is mediated by PKC under oxidative stress, facilitating NRF2 release from KEAP1 and nuclear translocation . This process is critical for:
Antioxidant Response: Upregulating ARE-dependent detoxifying enzymes (e.g., glutathione S-transferase) .
Innate Immunity: Suppressing pro-inflammatory cytokines (e.g., IL6) and viral replication (e.g., SARS-CoV-2, HSV) .
Cancer Research: Studying NRF2 hyperactivation in carcinomas and its role in chemoresistance .
Species Cross-Reactivity: Limited to human for most clones; Boster’s P00078 shows partial rodent reactivity but requires validation .
Band Discrepancy: Observed 90–100 kDa bands likely reflect phosphorylation-dependent conformational changes or ubiquitination .
Controls: Use phosphatase-treated lysates and non-phospho peptides to confirm specificity .
CUSABIO's phospho-NFE2L2 (S40) monoclonal antibody is a recombinant rabbit IgG antibody. It was generated using splenocytes from rabbits immunized with a phosphopeptide corresponding to human NFE2L2 (phosphorylated at Serine 40). The antibody's DNA sequence was cloned, expressed in vitro, and purified via affinity chromatography. This antibody is suitable for Western blotting (WB), immunohistochemistry (IHC), and immunofluorescence (IF) applications. It specifically recognizes human NFE2L2 phosphorylated at Serine 40.
NFE2L2 is a crucial transcription factor regulating cellular responses to reactive oxygen species (ROS) and oxidative stress. Protein kinase C (PKC) phosphorylates NFE2L2 at Serine 40 in response to oxidative stress. This phosphorylation facilitates NFE2L2's release from the cytoplasmic KEAP1 anchor, triggering nuclear translocation. Subsequently, the phosphorylated NFE2L2 at Serine 40 mediates the cellular antioxidant response via the antioxidant response element (ARE).
NFE2L2 (also known as NRF2) is a transcription factor essential for the cellular response to oxidative stress. It binds to antioxidant response elements (AREs) in the promoter regions of numerous cytoprotective genes, including phase 2 detoxifying enzymes. This binding upregulates their expression, thus neutralizing reactive electrophiles. Under normal conditions, NFE2L2 is ubiquitinated and degraded in the cytoplasm by the KEAP1-CUL3 E3 ubiquitin ligase complex (BCR). However, oxidative stress inhibits the KEAP1-CUL3 complex, resulting in NFE2L2 nuclear accumulation. There, it heterodimerizes with small Maf proteins and binds to AREs, activating the transcription of cytoprotective genes. Selective autophagy also activates the NFE2L2 pathway: autophagy enhances the KEAP1-SQSTM1/p62 interaction, inactivating the KEAP1-CUL3 complex and promoting NFE2L2 nuclear accumulation. Beyond oxidative stress response, NFE2L2 contributes to beta-globin gene cluster activation, innate immune regulation, and antiviral cytosolic DNA sensing. Specifically, it is a critical regulator of innate immunity and sepsis survival by maintaining redox homeostasis and preventing dysregulation of pro-inflammatory signaling pathways such as MyD88-dependent and -independent TNF-alpha signaling. NFE2L2 suppresses the macrophage inflammatory response by inhibiting pro-inflammatory cytokine transcription and IL6 induction. This occurs through binding near pro-inflammatory genes, independently of the NRF2-binding motif and ROS levels. Moreover, NFE2L2 represses antiviral cytosolic DNA sensing by suppressing STING1 expression and reducing responsiveness to STING1 agonists, increasing susceptibility to DNA viruses. Upon activation, NFE2L2 limits pro-inflammatory cytokine release in response to SARS-CoV-2 infection and its ligands by inhibiting IRF3 dimerization. It also inhibits SARS-CoV-2 and other pathogenic virus replication (e.g., HSV-1, HSV-2, Vaccinia virus, Zika virus) via a type I interferon (IFN)-independent mechanism.
Phosphorylation of NFE2L2 (also known as Nrf2) at serine 40 (S40) represents a critical regulatory mechanism in the cellular oxidative stress response pathway. This specific post-translational modification disrupts the interaction between NFE2L2 and KEAP1, preventing ubiquitination and subsequent proteasomal degradation of NFE2L2. Consequently, phosphorylated NFE2L2 accumulates in the nucleus where it heterodimerizes with small Maf proteins and binds to antioxidant response elements (ARE) in the promoter regions of cytoprotective genes. This activation triggers the expression of phase 2 detoxifying enzymes and antioxidant proteins that neutralize reactive electrophiles and protect cells from oxidative damage . The S40 phosphorylation site is particularly important as a direct target of protein kinase C (PKC) during oxidative stress conditions, making it a key biomarker for monitoring NFE2L2 activation status in various experimental contexts.
The NFE2L2 pathway demonstrates remarkable integration with multiple cellular stress response mechanisms:
Oxidative Stress Response: NFE2L2 acts as the master regulator of antioxidant defenses by upregulating genes that neutralize reactive oxygen species and electrophiles .
Selective Autophagy: The NFE2L2 pathway is activated during selective autophagy, where autophagy promotes interaction between KEAP1 and SQSTM1/p62, leading to inactivation of the BCR(KEAP1) complex and subsequent NFE2L2 nuclear accumulation .
Unfolded Protein Response (UPR): During endoplasmic reticulum stress, the NFE2L2 pathway is activated to maintain redox homeostasis and promote cell survival .
Inflammatory Response: NFE2L2 suppresses macrophage inflammatory responses by blocking pro-inflammatory signaling pathways, serving as a critical regulator of innate immune responses and survival during sepsis .
Ferroptosis Regulation: NFE2L2 activation can inhibit ferroptosis, a form of regulated cell death characterized by iron-dependent lipid peroxidation, by upregulating various target genes involved in iron metabolism and antioxidant defense .
This multifaceted involvement makes phospho-NFE2L2 detection crucial for studying integrated stress responses across various experimental models.
Phospho-NFE2L2 (S40) Recombinant Monoclonal Antibody demonstrates validated utility across multiple experimental techniques:
Western Blot (WB): Provides quantitative analysis of phospho-NFE2L2 levels in cell and tissue lysates, allowing assessment of activation status across different experimental conditions .
Immunohistochemistry (IHC): Enables visualization of phospho-NFE2L2 localization within tissue sections, particularly useful for assessing nuclear translocation as an indicator of activation .
Immunocytochemistry/Immunofluorescence (ICC/IF): Allows high-resolution subcellular localization studies to track phospho-NFE2L2 translocation between cytoplasm and nucleus in response to various stimuli .
Flow Cytometry: Permits quantitative single-cell analysis of phospho-NFE2L2 levels within heterogeneous cell populations, enabling identification of responsive subpopulations .
Chromatin Immunoprecipitation (ChIP): While not explicitly mentioned in the search results, this technique can be employed to assess phospho-NFE2L2 binding to ARE sequences in target gene promoters.
The choice of application should be guided by specific research questions, with western blot and immunofluorescence being particularly valuable for monitoring activation dynamics in response to oxidative challenges.
Optimal sample preparation for phospho-NFE2L2 (S40) detection requires careful consideration of the following methodological aspects:
Lysis Buffer Composition: Use phosphatase inhibitor-enriched lysis buffers (containing sodium fluoride, sodium orthovanadate, and β-glycerophosphate) to preserve phosphorylation status during extraction.
Nuclear Fraction Isolation: Since activated phospho-NFE2L2 translocates to the nucleus, nuclear extraction protocols yield enriched samples for detection of the activated form. Standard protocols using hypotonic lysis followed by high-salt nuclear extraction are recommended.
Rapid Processing: Process samples quickly at 4°C to minimize dephosphorylation by endogenous phosphatases.
Fixation for Microscopy: For immunofluorescence detection, paraformaldehyde fixation (4%) followed by permeabilization with 0.1% Triton X-100 preserves phospho-epitopes while enabling antibody access.
Positive Controls: Include samples treated with known NFE2L2 activators (such as sulforaphane or tert-butylhydroquinone) as positive controls to validate antibody performance.
Additionally, when performing western blot analysis, use freshly prepared samples whenever possible, as freeze-thaw cycles can diminish phospho-epitope integrity and affect detection sensitivity.
Researchers frequently encounter several technical challenges when detecting phospho-NFE2L2 (S40), which can be addressed through the following approaches:
High Background Signal:
Increase blocking time (5% BSA in TBST for 2 hours)
Optimize primary antibody dilution (typically 1:500-1:2000)
Include additional washing steps (5 x 5 minutes in TBST)
Use more specific secondary antibodies with minimal cross-reactivity
Weak or Absent Signal:
Confirm NFE2L2 activation with positive controls (e.g., H₂O₂ treatment)
Enrich nuclear fractions where activated phospho-NFE2L2 accumulates
Optimize protein loading (50-100 μg for whole cell lysates)
Increase exposure time for chemiluminescence detection
Multiple Bands in Western Blot:
Use gradient gels (4-12%) to better resolve NFE2L2 isoforms
Include protease inhibitors in lysis buffer to prevent degradation
Validate bands with positive controls and knockout/knockdown samples
Poor Nuclear Staining in Immunofluorescence:
These methodological refinements significantly improve detection reliability while minimizing artifacts that could lead to misinterpretation of experimental results.
Rigorous experimental design requires incorporation of several key controls to ensure valid interpretation of phospho-NFE2L2 (S40) detection:
Positive Controls:
Cells/tissues treated with known NFE2L2 activators (sulforaphane, tert-butylhydroquinone)
Recombinant phosphorylated NFE2L2 protein (for western blot standard curves)
Negative Controls:
Genetic models: NFE2L2 knockout/knockdown samples
Dephosphorylation controls: Lysate aliquots treated with lambda phosphatase
Secondary antibody-only controls to assess non-specific binding
Specificity Controls:
Peptide competition assays using phospho-S40 peptides
Parallel detection with total NFE2L2 antibody to calculate phosphorylation ratio
Cross-validation with alternative phospho-NFE2L2 antibody clones
Functional Validation:
Implementation of these controls provides crucial evidence for antibody specificity and ensures that observed signals genuinely represent phosphorylated NFE2L2 rather than artifacts or non-specific binding.
The application of phospho-NFE2L2 (S40) antibodies offers sophisticated approaches to investigate NFE2L2's critical role in ferroptosis resistance:
Mechanistic Analysis: These antibodies enable direct monitoring of NFE2L2 activation status in response to ferroptosis inducers (e.g., erastin, RSL3), providing temporal resolution of the antioxidant response. This approach reveals whether NFE2L2 activation precedes, coincides with, or follows lipid peroxidation events, helping establish causality in protection mechanisms .
Target Gene Expression Correlation: By combining phospho-NFE2L2 detection with analysis of downstream targets specifically involved in ferroptosis protection (SLC7A11, GPX4, FTH1, FTL), researchers can establish direct functional relationships between NFE2L2 activation and the expression of ferroptosis defense genes .
Cancer Therapy Resistance Models: In cancer cell lines and patient-derived xenografts showing therapy resistance, phospho-NFE2L2 antibodies can identify whether constitutive activation contributes to ferroptosis evasion. This information guides rational design of combination therapies targeting both NFE2L2 and ferroptosis pathways .
Structure-Function Analysis: When combined with site-directed mutagenesis of NFE2L2 domains, these antibodies help determine how S40 phosphorylation specifically contributes to ferroptosis resistance compared to other activation mechanisms.
Drug Discovery Applications: The antibodies facilitate high-throughput screening of compounds that modulate NFE2L2 phosphorylation status, potentially identifying novel ferroptosis sensitizers for cancer therapy .
A methodical research approach using these techniques has revealed that NFE2L2 activation upregulates multiple ferroptosis defense genes including SLC7A11, GCL, GSS, GSR, GPX4, and metabolic enzymes that support glutathione synthesis and utilization .
Distinguishing between KEAP1-dependent and KEAP1-independent NFE2L2 activation requires sophisticated methodological approaches:
Phosphorylation Status Analysis: Phospho-NFE2L2 (S40) antibodies are particularly valuable as S40 phosphorylation represents a KEAP1-independent activation mechanism. Comparative analysis of total NFE2L2 versus phospho-S40 NFE2L2 levels can establish the relative contribution of each pathway:
Activation Mechanism | Total NFE2L2 | Phospho-S40 NFE2L2 | Nuclear/Cytoplasmic Ratio |
---|---|---|---|
KEAP1-dependent | Increased | Minimal change | Increased |
Phosphorylation-dependent | Moderate increase | Significantly increased | Increased |
Combined mechanisms | Substantially increased | Significantly increased | Substantially increased |
Protein-Protein Interaction Analysis:
Co-immunoprecipitation assays using phospho-NFE2L2 (S40) antibodies to assess interaction with KEAP1
Proximity ligation assays to visualize NFE2L2-KEAP1 interactions in situ
FRET-based biosensors to monitor real-time dissociation kinetics
Genetic Approaches:
KEAP1 knockout or knockdown studies combined with phospho-NFE2L2 (S40) detection
Expression of phospho-mimetic (S40D) or phospho-deficient (S40A) NFE2L2 mutants
CRISPR-Cas9 genome editing of endogenous NFE2L2 phosphorylation sites
Pathway-Specific Activators:
These approaches revealed that multiple regulatory mechanisms often operate simultaneously, with phosphorylation at S40 facilitating the escape from KEAP1-mediated degradation while also independently enhancing NFE2L2 transcriptional activity through altered protein-protein interactions with transcriptional machinery .
Investigating the complex relationship between NFE2L2 phosphorylation and innate immune regulation requires multidisciplinary experimental approaches:
Temporal Activation Studies:
Use phospho-NFE2L2 (S40) antibodies to track activation kinetics during immune challenges
Perform parallel analysis of NFE2L2 phosphorylation and inflammatory cytokine production
Establish time-dependent relationships between NFE2L2 activation and resolution phases of inflammation
Cell Type-Specific Analysis:
Flow cytometry with phospho-NFE2L2 (S40) antibodies to identify responsive immune cell populations
Single-cell resolution of NFE2L2 activation in heterogeneous immune cell populations
Correlation of phospho-NFE2L2 levels with immune cell functional states
Signaling Pathway Cross-talk:
Functional Outcome Assessment:
Measure how manipulation of NFE2L2 phosphorylation affects phagocytosis, ROS production, and cytokine secretion
Correlate phospho-NFE2L2 levels with bacterial clearance or viral replication in infection models
Assess the impact on resolution of inflammation and tissue damage
Disease Model Applications:
Research using these approaches has demonstrated that NFE2L2 acts as a critical regulator of innate immune responses during sepsis by maintaining redox homeostasis and restraining dysregulated pro-inflammatory signaling pathways, including MyD88-dependent and -independent pathways and TNF-alpha signaling . The phosphorylation status of NFE2L2 serves as a molecular switch that balances protective inflammatory responses with excessive immune activation.
Phospho-NFE2L2 (S40) antibodies provide powerful tools for elucidating the mechanisms of therapeutic resistance in cancer:
Biomarker Development:
Quantitative analysis of phospho-NFE2L2 (S40) levels in patient tumor samples to predict therapy response
Correlation of NFE2L2 phosphorylation status with clinical outcomes and resistance patterns
Longitudinal monitoring of phospho-NFE2L2 levels during treatment to detect emerging resistance
Resistance Mechanism Characterization:
Comparative phospho-NFE2L2 profiling between sensitive and resistant cell lines
Analysis of NFE2L2 activation following exposure to chemotherapeutics, radiation, or targeted therapies
Identification of resistance-associated NFE2L2 target gene signatures
Combination Therapy Development:
Mechanistic Studies:
Investigation of how tumor microenvironment factors regulate NFE2L2 phosphorylation
Analysis of cross-talk between NFE2L2 and other resistance-associated signaling pathways
Identification of kinases responsible for therapy-induced NFE2L2 phosphorylation
Research has demonstrated that elevated NFE2L2 activity supports tumor progression, metastasis, and therapy resistance in various malignancies. NFE2L2 activation contributes to drug resistance by upregulating antioxidant defenses and inhibiting ferroptosis through regulation of multiple target genes including NQO1, HMOX1, FTH1, FTL, SLC7A11, GCL, GSS, GSR, GPX4, and AIFM2 . Phospho-NFE2L2 (S40) antibodies enable precise tracking of this activation, making them invaluable tools for developing strategies to overcome NFE2L2-mediated therapeutic resistance.
Resolving contradictory data regarding NFE2L2 activation across different experimental models requires systematic methodological approaches:
Standardized Detection Protocols:
Implement consistent antibody validation procedures across laboratories
Establish standardized positive and negative controls for phospho-NFE2L2 detection
Develop quantitative assays with defined dynamic ranges and detection limits
Comprehensive Activation Assessment:
Measure multiple NFE2L2 activation parameters simultaneously:
a) Total protein levels (stability)
b) Phosphorylation status (S40 and other sites)
c) Nuclear translocation (subcellular fractionation)
d) DNA binding activity (ChIP or EMSA)
e) Transcriptional output (target gene expression)
Context-Dependent Analysis:
Systematically evaluate how experimental variables affect NFE2L2 activation:
a) Cell type and differentiation state
b) Culture conditions (2D vs. 3D, oxygen tension)
c) Acute vs. chronic stress exposure
d) Presence of competing signaling pathways
Time-Course Resolution:
Multi-Omics Integration:
Combine phospho-proteomics, transcriptomics, and metabolomics data
Develop computational models that predict context-dependent NFE2L2 responses
Identify factors that modify NFE2L2 activity in different experimental systems
This methodical approach has revealed that apparent contradictions often reflect genuine biological complexity rather than technical artifacts. For example, in certain contexts, NFE2L2 phosphorylation may be disconnected from nuclear translocation due to additional regulatory mechanisms. Similarly, the transcriptional output of phospho-NFE2L2 can vary dramatically depending on the availability of co-factors and the epigenetic landscape of target genes .
A comprehensive experimental design to robustly assess NFE2L2 activation status should incorporate multiple complementary approaches:
Multi-parameter Assessment Strategy:
Parameter | Technique | Information Provided |
---|---|---|
Phosphorylation (S40) | Western blot with phospho-specific antibody | Direct measure of activation-associated modification |
Nuclear Translocation | Nuclear/cytoplasmic fractionation followed by western blot or immunofluorescence | Subcellular localization indicating activation |
KEAP1 Interaction | Co-immunoprecipitation or proximity ligation assay | Release from inhibitory complex |
DNA Binding | Chromatin immunoprecipitation (ChIP) or electrophoretic mobility shift assay (EMSA) | Functional engagement with target genes |
Transcriptional Activity | qPCR of target genes or ARE-luciferase reporter assay | Functional output of activation |
Temporal Resolution:
Early activation markers: S40 phosphorylation (minutes to hours)
Intermediate markers: Nuclear accumulation (hours)
Late markers: Target gene expression (hours to days)
Include multiple time points to capture the complete activation cycle
Stimulus-Specific Responses:
Compare phospho-NFE2L2 responses across different activators:
a) Electrophiles (sulforaphane, tBHQ)
b) Oxidative stressors (H₂O₂, paraquat)
c) Pathway-specific activators (PKC activators for phosphorylation)
Determine stimulus-specific activation signatures
Genetic Validation:
This integrated approach provides a comprehensive assessment of NFE2L2 activation status while controlling for technical artifacts and biological variability. Studies employing such robust designs have revealed that NFE2L2 activation involves a coordinated sequence of events, with phosphorylation at S40 often serving as an early indicator that precedes and facilitates subsequent steps in the activation cascade .
Interpreting discrepancies between phospho-NFE2L2 (S40) levels and target gene expression requires systematic analysis of potential mechanistic explanations:
Temporal Disconnection:
Phosphorylation typically precedes transcriptional changes by several hours
Establish time-course relationships before concluding genuine disconnection
Consider target gene mRNA half-life and protein turnover rates when interpreting expression data
Threshold Effects:
Determine whether minimum threshold levels of phospho-NFE2L2 are required for transcriptional activation
Establish dose-response relationships between phosphorylation levels and target gene expression
Consider that different target genes may have different activation thresholds
Competing Regulatory Mechanisms:
Investigate epigenetic regulation of target gene promoters (histone modifications, DNA methylation)
Assess availability of essential co-factors (small Maf proteins, co-activators)
Examine presence of competing transcription factors at ARE sites
Technical Considerations:
Verify antibody specificity using appropriate controls
Ensure nuclear extraction protocols effectively isolate transcriptionally active phospho-NFE2L2
Validate target gene primer specificity and efficiency
Integrated Analysis Approach:
Implement single-cell analyses to resolve population heterogeneity
Use ChIP-seq to map genome-wide NFE2L2 binding in relation to phosphorylation status
Apply mathematical modeling to integrate multiple regulatory layers
Research utilizing these analytical approaches has revealed that while S40 phosphorylation enhances NFE2L2 transcriptional activity, it is not always sufficient for target gene induction. For example, studies examining NFE2L2 binding to ARE sequences in chagasic myocardium demonstrated that despite the presence of phospho-NFE2L2, binding capacity was decreased by 52%, leading to reduced expression of downstream targets like MnSOD, γGCS, and HO1 . This indicates that additional factors beyond phosphorylation status regulate the functional output of the NFE2L2 pathway.