NFE2L2 Antibody

Shipped with Ice Packs
In Stock

Description

Research Applications

The antibody is widely used in studies investigating oxidative stress, inflammation, and degenerative diseases:

  • Immunohistochemistry: Detected elevated NFE2L2 expression in neutrophils of ankylosing spondylitis (AS) patients, correlating with ferroptosis regulation .

  • Western Blot: Validated NRF2 stabilization in macroautophagy studies, showing impaired autophagy gene expression in NFE2L2-deficient mice .

  • ChIP Assays: Identified ARE-binding sites in autophagy-related genes (e.g., SQSTM1, ULK1) using a V5-tagged NFE2L2 construct due to limitations in endogenous antibody efficiency .

Performance in Diverse Studies

Study TypeKey FindingCitation
Ferroptosis in ASNFE2L2 upregulation in neutrophils of AS patients linked to disease progression .
Cuproptosis ResistanceNFE2L2-mediated GSH synthesis drives resistance to copper-induced cell death .
Autophagy RegulationNRF2 deficiency impairs autophagy flux and aggregates clearance in Alzheimer’s models .

Limitations and Considerations

  • Cross-reactivity: Confirmed specificity for human and mouse NFE2L2, but requires optimization for other species .

  • Epitope Competition: Neh7-targeting may interfere with transcriptional repression assays .

  • Optimization: Recommended concentrations vary by application (e.g., 0.2–0.5 µg/mL for WB, 2–5 µg/mL for IHC) .

Therapeutic Implications

Research highlights NFE2L2 as a target for drugs modulating oxidative stress in diseases like cancer and neurodegeneration. For example, cinnamaldehyde exhibits strong binding affinity to NFE2L2 (−3.2 kcal/mol), stabilizing its activity .

Product Specs

Buffer
Preservative: 0.03% ProClin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Order fulfillment typically takes 1-3 business days. Shipping times vary depending on the order method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
erythroid derived 2 antibody; HEBP1 antibody; like 2 antibody; NF E2 related factor 2 antibody; NF-E2-related factor 2 antibody; NF2L2_HUMAN antibody; NFE2 related factor 2 antibody; NFE2-related factor 2 antibody; Nfe2l2 antibody; Nrf 2 antibody; NRF2 antibody; Nuclear factor (erythroid derived 2) like 2 antibody; Nuclear factor antibody; nuclear factor erythroid 2 like 2 antibody; Nuclear factor erythroid 2 related factor 2 antibody; Nuclear factor erythroid 2-related factor 2 antibody; Nuclear factor erythroid derived 2 like 2 antibody
Target Names
Uniprot No.

Target Background

Function

The NFE2L2 (NRF2) antibody targets a transcription factor crucial for the cellular response to oxidative stress. NRF2 binds to antioxidant response elements (AREs) in the promoter regions of numerous cytoprotective genes, including phase 2 detoxifying enzymes. This binding stimulates gene expression, thereby neutralizing reactive electrophiles. Under normal conditions, NRF2 is ubiquitinated and degraded in the cytoplasm by the KEAP1-CUL3 E3 ubiquitin ligase complex. Oxidative stress inhibits KEAP1 activity, leading to NRF2 nuclear accumulation, heterodimerization with small Maf proteins, and subsequent ARE binding for the activation of cytoprotective target genes. Selective autophagy also activates the NFE2L2/NRF2 pathway; autophagy facilitates the KEAP1-SQSTM1/p62 interaction and KEAP1 complex inactivation, resulting in NRF2 nuclear translocation and cytoprotective gene expression. NRF2 may also regulate beta-globin cluster gene transcription by mediating enhancer activity within the beta-globin locus control region. Additionally, NRF2 plays a significant role in innate immune responses and antiviral cytosolic DNA sensing. It is a critical regulator of innate immunity and sepsis survival, maintaining redox homeostasis and preventing dysregulation of proinflammatory signaling pathways (MyD88-dependent and -independent, and TNF-α signaling). NRF2 suppresses macrophage inflammatory responses by inhibiting proinflammatory cytokine transcription and IL-6 induction. It binds near proinflammatory genes in macrophages, hindering RNA polymerase II recruitment—a process independent of the NRF2-binding motif and reactive oxygen species levels. Furthermore, NRF2 represses antiviral cytosolic DNA sensing by suppressing STING1 expression, reducing responsiveness to STING1 agonists, and increasing susceptibility to DNA virus infection. Upon activation, NRF2 limits the release of pro-inflammatory cytokines in response to SARS-CoV-2 infection and virus-derived ligands by inhibiting IRF3 dimerization. Importantly, NRF2 inhibits SARS-CoV-2 replication and the replication of other pathogenic viruses (Herpes Simplex Virus-1 and -2, Vaccinia virus, and Zika virus) via a type I interferon (IFN)-independent mechanism.

Gene References Into Functions

Relevant Research Highlights:

  1. Upregulation of key antioxidant enzymes, the Nrf2 transcription factor, and downregulation of NOX5 during the development of drug resistance in cisplatin-treated tumor cells. PMID: 30225719
  2. HIF1A upregulation in breast and bladder tumors with high NRF2 activity, indicating a direct regulatory link between these oxygen-responsive transcription factors. PMID: 30241031
  3. Identification of a core set of 32 direct NRF2 targets consistently upregulated in NRF2-hyperactivated tumors. PMID: 30195190
  4. Decreased expression of Nrf2-ARE molecules and related antioxidases in patients with obstructive sleep apnea hypopnea syndrome (OSAHS), correlating with neurocognitive dysfunction. PMID: 30159112
  5. A link between constitutive Nrf2 activation, tumor aerobic glycolysis, and progression of urothelial transitional cell carcinoma (UTUC). PMID: 29716554
  6. Review article discussing NRF2 as a transcription factor transducing chemical signals to regulate cytoprotective genes, with KEAP1 repressing NRF2 activity under normal conditions. PMID: 29717933
  7. Confirmation of porphyra-334 and shinorine's ability to dissociate Nrf2 from Keap1, resulting in increased mRNA expression of Nrf2-targeted genes in primary skin fibroblasts. PMID: 30071261
  8. Common expression of NRF2, DJ1, and SRNX1 in diffusely infiltrating astrocytomas, with prognostic implications. PMID: 29441509
  9. Pterostilbene's attenuation of high glucose-induced central nervous system injury via Nrf2 signaling activation, protecting against mitochondrial dysfunction-derived oxidative stress. PMID: 28089584
  10. Upregulation of xCT antiporter expression via Nrf2 in some breast cancer cells, contributing to their glucose dependence. PMID: 28429737
  11. Evidence for the involvement of NFE2L2 and PPARGC1α in Parkinson's disease susceptibility and progression, potentially through maneb and paraquat exposure pathways. PMID: 29630901
  12. A novel interaction between Nrf2 and ATF4 under oxidative and endoplasmic reticulum stress, driving antioxidant mechanisms. PMID: 29421327
  13. Senescence-associated downregulation of NRF2 decreasing endothelial glycolytic activity and stress tolerance, restored upon NRF2 reinstatement. PMID: 29986211
  14. Potential impact of aberrant Nrf2/Keap1 system integrity on oxidative stress defense mechanisms in primary biliary cholangitis. PMID: 28333129
  15. Inflammation, oxidative stress, and higher Nrf2 and NQO1 protein expression levels in the airways of women exposed to biomass fuel smoke. PMID: 29363060
  16. NRF2/NFE2L2 promotes breast cancer progression by enhancing glycolysis through HIF1A co-activation; elevated NRF2 and HIF1A levels in breast cancer cells compared to benign cells. PMID: 29275212
  17. NRF2 expression regulation by NRG1 in papillary thyroid cancer (PTC). PMID: 29901070
  18. 27-OH-induced autophagy's dependence on the Nrf2-dependent antioxidant response and p62. PMID: 29879549
  19. Evidence for a direct role of NRF2 in globin gene regulation. PMID: 28473619
  20. The CD44-NRF2 axis as a potential therapeutic target for controlling stress resistance and survival of CD44(high) cancer stem cells in breast tumors. PMID: 29729523
  21. Changes in NRF2 expression levels induced by cell-free DNA in different cell types. PMID: 29743966
  22. Nrf2-dependent activation of MCT1-driven lactate exchange inducing metabolism-dependent clonal growth of HCT15 colorectal cancer cells. PMID: 28846107
  23. Rational design of an epitope-specific antibody binding to Keap1, blocking the Keap1-Nrf2 interaction. PMID: 28128368
  24. Review article focusing on Nrf2 effects on redox systems, mitochondrial function, and proteostasis. PMID: 28424271
  25. Therapeutic inhibition of Nrf2/ABCB1 signaling as a potential novel therapeutic strategy. PMID: 29793178
  26. Review article discussing the cross-talk between HIF1A, NRF2, and NF-κB in adapting to oxygen availability changes. PMID: 29485192
  27. Lutein's antiproliferative effect mediated by NrF2/ARE pathway activation and NF-κB signaling pathway blocking. PMID: 29336610
  28. Nuclear factor erythroid-2-related factor 2 (NRF2) regulating LRWD1 expression and cellular adaptation to oxidative stress in human embryonal carcinoma cells. PMID: 29544732
  29. Perillaldehyde's inhibition of BaP-induced AHR activation and ROS production, inhibition of BaP/AHR-mediated CCL2 chemokine release, and activation of the NRF2/HO1 antioxidant pathway in human keratinocytes. PMID: 29643980
  30. NRF2's regulation of basal and inducible HER1 expression in ovarian cancer cells. PMID: 29410730
  31. Nrf2 deregulation's association with aging and pathogenesis of chronic diseases, including neurodegenerative diseases. PMID: 29969760
  32. Pc-induced HO-1 expression mediated by the PKCA-Nrf-2/HO-1 pathway, inhibiting UVB-induced apoptotic cell death in primary skin cells. PMID: 29470442
  33. Identification of a gene signature regulated by the KEAP1-NRF2-CUL3 axis associated with tumorigenesis and drug resistance in head and neck squamous cell cancer. PMID: 29306329
  34. Association of Nrf2 overexpression with tumor size, histological grade, and metastasis in gastric cancer patients. PMID: 29091877
  35. ChREBPα's upregulation of NRF2 expression and activity, initiating mitochondrial biogenesis in beta-cells; NRF2 induction required for ChREBPα-mediated effects and glucose-stimulated beta-cell proliferation. PMID: 29764859
  36. The NFE2L2 promotor variant rs6721961's potential protective effect against hearing loss in cisplatin-receiving cancer patients. PMID: 27457817
  37. Arachidin-1's suppression of TNF-α-induced inflammation in endothelial cells via upregulation of Nrf-2-related phase II enzyme expression. PMID: 29115410
  38. Review article on the Nrf2-Keap1 system, Nrf2 functions, Nrf2-NF-κB cross-talk, and the system's effects in striated muscle physiology and pathophysiology. PMID: 29499228
  39. Smoke-induced HO-1 expression modulation through the NRF2/BACH1 axis, showing significant nuclear NRF2 translocation but only a slight decrease in nuclear BACH1. PMID: 29125538
  40. Significant downregulation of NRF2 expression in a hepatic ischemia-reperfusion (HIR) model. PMID: 28708282
  41. BRAF-mediated NRF2 gene transcription and Histone Acetyltransferases-mediated NRF2 protein acetylation contributing to ABCC1-mediated chemoresistance and glutathione-mediated survival in acquired topoisomerase II poison-resistant cancer cells. PMID: 29080842
  42. Respiratory syncytial virus-induced NRF2 degradation through a PML-RNF4 pathway. PMID: 29107745
  43. Positive regulation of NRF2 activity by miR-432-3p through KEAP1 downregulation and increased NRF2 stability in squamous cell carcinoma. PMID: 28760781
  44. PGAM5-KEAP1-Nrf2 complex preservation of mitochondrial motility by suppressing dominant-negative KEAP1 activity. PMID: 28839075
  45. Hydrogen sulfide's attenuation of vascular smooth muscle cell calcification via the KEAP1-NRF2 system by enhancing NQO1 expression. PMID: 28865326
  46. 5-hydroxyeicosatetraenoic acid and 5-hydroxyeicosapentaenoic acid acting as Nrf2 activators via the metabolite 5-oxo-ETE in human umbilical vein endothelial cells (HUVECs). PMID: 28892009
  47. The Nrf2-VEGF interplay's contribution to venous hypertension-induced angiogenesis in brain arteriovenous malformations. PMID: 27869147
  48. Association of high Nrf2 expression in alveolar type I pneumocytes with low recurrences in primary spontaneous pneumothorax. PMID: 28962820
  49. NRF2 activation in cancer cells increasing dependency on exogenous glutamine via glutamate consumption for glutathione synthesis and glutamate secretion by xc(-) antiporter system. PMID: 28967864
  50. Inhibition of Wnt3A secretion blocking the Wnt signaling pathway and preventing Nrf2 signaling; potential use of Wnt inhibitors as radiosensitizing drugs. PMID: 28627706
Database Links

HGNC: 7782

OMIM: 600492

KEGG: hsa:4780

STRING: 9606.ENSP00000380252

UniGene: Hs.744006

Protein Families
BZIP family, CNC subfamily
Subcellular Location
Cytoplasm, cytosol. Nucleus.
Tissue Specificity
Widely expressed. Highest expression in adult muscle, kidney, lung, liver and in fetal muscle.

Customer Reviews

Overall Rating 5.0 Out Of 5
,
B.A
By Anonymous
★★★★★

Applications : Immunoblotting

Sample type: cells

Review: immunoblotting revealed that IL- 1β stimulation reduced the mRNA and protein levels of the anti-oxidative factors Nrf2, HO-1, NQO-1, SOD1, and SOD2,but kaempferol only partially mitigated IL-1-β induced oxidative changes.

Q&A

What is the molecular weight pattern expected when detecting NFE2L2/NRF2 by Western blot?

NFE2L2/NRF2 typically displays multiple banding patterns in Western blot experiments, which can cause confusion among researchers. The predicted molecular weight is approximately 68 kDa, but the observed molecular weight often appears at both 68 kDa and 95-110 kDa due to post-translational modifications . According to validation studies, the consensus in scientific literature confirms that the actual observed molecular weights of NFE2L2 are approximately 70 kDa and 95-110 kDa . This variation in banding pattern can lead to uncertainties about specificity, which requires careful validation of antibodies used.

What are the recommended dilutions for NFE2L2 antibodies across different applications?

Different experimental applications require specific antibody dilution ranges for optimal results:

ApplicationRecommended Dilution
Western Blot (WB)1:2000-1:12000
Immunoprecipitation (IP)0.5-4.0 μg for 1.0-3.0 mg of total protein lysate
Immunohistochemistry (IHC)1:50-1:500
Immunofluorescence (IF)/ICC1:50-1:500

It's important to note that these dilutions should be optimized for each specific experimental system, as reactivity can vary depending on sample type and processing methods . For recombinant monoclonal antibodies, manufacturers may recommend different dilutions, such as 1:20-1:200 for IF applications .

How can I improve detection of endogenous NFE2L2 protein under basal conditions?

Under normal physiological conditions, NFE2L2 is rapidly ubiquitinated and degraded in the cytoplasm, making it difficult to detect by Western blot . Methodological approaches to improve detection include:

  • Adding proteasome inhibitors like MG132 to cell cultures prior to lysis, which effectively inhibits NFE2L2 degradation and increases detection signal

  • Using nuclear extraction protocols since NFE2L2 accumulates in the nucleus during oxidative stress

  • Employing treatments that induce oxidative stress (e.g., H₂O₂) or chemical activators like sulforaphane (SFN) to stabilize NFE2L2

  • Optimizing lysis buffers to include phosphatase inhibitors that preserve post-translational modifications

These approaches can significantly enhance the detection of endogenous NFE2L2 in experimental systems .

What cell types and tissues are recommended for validating NFE2L2 antibody specificity?

Based on extensive validation studies, specific cell types and tissues have been consistently used to verify NFE2L2 antibody specificity:

Sample TypeValidated Cell Lines/Tissues
WB-positive cell linesHepG2 cells, A549 cells, DMSO-treated HeLa cells
IP-positive cellsHeLa cells
IHC-positive tissuesHuman liver cancer tissue, human breast cancer tissue, human colon cancer tissue, human kidney tissue, human pancreas cancer tissue, human renal cell carcinoma tissue
IF/ICC-positive cellsMG132-treated HepG2 cells, HepG2 cells

These specific sample types have demonstrated reliable NFE2L2 detection and should be considered as positive controls when validating new antibodies or experimental conditions . Notably, antibodies may show differential reactivity across species, with most validated for human samples .

How can I design proper controls when studying NFE2L2 translocation in response to stress conditions?

Designing appropriate controls for NFE2L2 translocation studies requires multiple approaches:

  • Positive controls: Include known NFE2L2 activators such as sulforaphane (SFN) treatment, which has been demonstrated to induce NFE2L2 nuclear accumulation and target gene expression

  • Negative controls: Use Nfe2l2-knockout cells or tissues as definitive negative controls, which have been shown to exhibit reduced expression of autophagy genes including Sqstm1, Calcoco2, Ulk1, Atg2b, Atg4d, Atg5, Atg7, and Gabarapl1

  • Time-course experiments: Monitor NFE2L2 translocation at multiple time points following stress induction (e.g., 0, 1, 2, 4, and 8 hours post-treatment)

  • Subcellular fractionation: Compare cytoplasmic versus nuclear fractions to quantify translocation efficiency

  • Rescue experiments: Transfection of NFE2L2 expression constructs (particularly those lacking the KEAP1 regulatory ETGE domain) into knockout cells can restore target gene expression and serve as functional validation

These control strategies ensure reliable interpretation of NFE2L2 translocation and downstream effects in response to various stimuli.

What are validated approaches for ChIP experiments targeting NFE2L2 given the challenges with endogenous immunoprecipitation?

ChIP experiments with NFE2L2 present unique challenges due to difficulties in immunoprecipitating endogenous NFE2L2 efficiently. Published protocols have successfully addressed this limitation through the following approaches:

  • Using HEK293T cells transfected with an expression vector for V5-tagged NFE2L2 that lacks the KEAP1 regulatory domain (ETGE), facilitating NFE2L2 stabilization, nuclear translocation, and binding to target genes

  • Performing ChIP with anti-V5 antibody (for tagged NFE2L2) and anti-IgG as negative control

  • Analyzing immunoprecipitated DNA by quantitative real-time PCR (qRT-PCR) with specific primers surrounding putative Antioxidant Response Elements (AREs)

  • Screening chromatin immunoprecipitation databases like ENCODE for proteins that bind NFE2L2-regulated enhancer AREs (such as MAFK and BACH1) as alternative approaches to identify NFE2L2 binding sites

  • Validating newly identified ARE sequences in autophagy genes through enrichment analysis in V5-immunoprecipitated chromatin

These approaches have successfully identified NFE2L2 binding to regulatory regions of multiple autophagy genes, establishing its role in transcriptional regulation beyond oxidative stress response.

How can NFE2L2 antibodies be used to investigate its role in cancer and therapeutic resistance?

NFE2L2 antibodies serve as critical tools for investigating its role in cancer development and treatment resistance:

  • Mutation analysis: NFE2L2 mutations, particularly in the ETGE or DLG domains that abrogate KEAP1 association, can be detected through immunoblotting to assess nuclear accumulation patterns

  • Prognostic biomarker assessment: Studies have shown that NFE2L2/KEAP1/CUL3 mutations are associated with significantly more local failure (HR = 3.50, 95% CI: 1.56–7.89, P = 0.0025) and locoregional failure (HR = 3.80, 95% CI: 1.80–8.03, P = 0.0005) in cancer radiotherapy

  • Therapeutic resistance monitoring: NFE2L2 nuclear accumulation can be quantified by immunohistochemistry or immunofluorescence to correlate with radiotherapy outcomes, as demonstrated in T2N0 glottic squamous cell carcinoma patients where disease-free survival was significantly worse for patients with NFE2L2 pathway mutations (HR = 2.88, 95% CI: 1.46–5.66, P = 0.0022)

  • Oncogenic driver analysis: Patient-derived NFE2L2 mutants (e.g., L30P and R34P) have been shown to dramatically accelerate tumorigenesis when co-expressed with other oncogenic factors, which can be tracked using specific antibodies

These applications highlight NFE2L2 antibodies as valuable tools for understanding cancer biology and therapeutic resistance mechanisms.

What strategies can address inconsistent or background issues when using NFE2L2 antibodies in immunohistochemistry?

Researchers encountering inconsistent results or background issues with NFE2L2 antibodies in IHC can implement several validated strategies:

  • Optimized antigen retrieval: For NFE2L2 detection in tissues, suggested protocols include antigen retrieval with TE buffer pH 9.0, although citrate buffer pH 6.0 may be used as an alternative

  • Blocking optimization: Extended blocking periods (1-2 hours) with specialized blocking buffers containing both serum proteins and detergents can reduce nonspecific binding

  • Antibody validation: Using tissues from nfe2l2-knockout mice as negative controls and tissues with known high NFE2L2 expression (e.g., human liver cancer tissue, human breast cancer tissue) as positive controls

  • Secondary antibody selection: Higher dilutions of secondary antibodies (1:1000 or greater) often reduce nonspecific staining while maintaining specific signal

  • Signal amplification systems: For low-expression samples, employing tyramide signal amplification systems can enhance specific signals while maintaining signal-to-noise ratio

These methodological refinements have been successful in multiple published studies and can significantly improve NFE2L2 detection specificity in tissue samples.

What are the best approaches for validating NFE2L2 antibody specificity for research publications?

Comprehensive validation of NFE2L2 antibodies for publication-quality research should include:

  • Genetic controls: Testing antibodies on samples from Nfe2l2-knockout mice or cells with CRISPR/Cas9-mediated NFE2L2 deletion to demonstrate specificity

  • Rescue experiments: Reintroducing NFE2L2 expression in knockout systems using constructs like NFE2L2-ΔETGE-V5, which has been shown to rescue normal basal levels of expression of autophagy genes including Sqstm1, Calcoco2, Ulk1, Atg5, Atg7, and Gabarapl1

  • Multi-technique validation: Confirming consistent results across complementary techniques (e.g., if Western blot shows a 68 kDa band, validate with mass spectrometry or immunoprecipitation followed by protein identification)

  • Known inducer testing: Demonstrating appropriate response to established NFE2L2 activators like sulforaphane or oxidative stress inducers (H₂O₂), which should show increased nuclear localization and target gene expression

  • Cross-antibody validation: Using multiple antibodies targeting different epitopes of NFE2L2 to confirm consistent detection patterns

These validation approaches provide compelling evidence of antibody specificity and reliability, essential for high-quality publications.

How can NFE2L2 antibodies be used to investigate the crosstalk between oxidative stress response and autophagy?

Recent research has established NFE2L2 as a critical regulator of both oxidative stress response and autophagy. To investigate this crosstalk:

  • Chromatin immunoprecipitation: NFE2L2 antibodies can be used in ChIP assays to identify direct binding to antioxidant response elements (AREs) in autophagy gene promoters. Studies have validated 11 ARE regions in autophagy genes that NFE2L2 directly binds, including SQSTM1, CALCOCO2, ULK1, ATG2B, ATG4D, ATG5, ATG7, and GABARAPL1

  • Dual immunofluorescence: Co-staining of NFE2L2 with autophagy markers like SQSTM1/p62 can reveal their spatial relationship during stress conditions. Studies in Alzheimer's disease models show reduced colocalization of APP and MAPT with SQSTM1/p62 in the absence of NFE2L2, suggesting impaired autophagy-mediated clearance

  • Autophagy flux assessment: NFE2L2 antibodies combined with LC3B antibodies can help assess autophagy flux under oxidative stress. Research shows that H₂O₂ treatment increases LC3B-II levels in wild-type cells but to a lesser extent in nfe2l2-KO MEFs, indicating NFE2L2 regulates autophagy specifically under oxidative stress conditions

  • Rescue experiments: Introducing NFE2L2 constructs lacking the KEAP1 regulatory domain into NFE2L2-deficient cells restores expression of autophagy genes, providing a powerful tool to confirm direct regulation

These approaches have revealed that NFE2L2 functions at the intersection of redox homeostasis and protein quality control through direct transcriptional regulation of autophagy genes.

What are the current challenges and solutions for detecting modified forms of NFE2L2 protein?

NFE2L2 undergoes multiple post-translational modifications that affect its stability, localization, and function. Current challenges and solutions include:

  • Phosphorylation detection: NFE2L2 is phosphorylated by multiple kinases, affecting its stability and activity. Phospho-specific antibodies combined with phosphatase inhibitors in lysis buffers can help detect these modified forms

  • Ubiquitination analysis: Under normal conditions, NFE2L2 is heavily ubiquitinated. Using proteasome inhibitors (MG132) combined with immunoprecipitation and ubiquitin-specific antibodies allows detection of ubiquitinated forms

  • Acetylation detection: NFE2L2 acetylation affects DNA binding. Immunoprecipitation with NFE2L2 antibodies followed by detection with acetyl-lysine antibodies can reveal this modification

  • Nuclear vs. cytoplasmic forms: Different modified forms predominate in different cellular compartments. Subcellular fractionation combined with Western blotting can distinguish these populations

  • High molecular weight bands: NFE2L2 often appears at 95-110 kDa in addition to the predicted 68 kDa band due to modifications . Using gradient gels (4-15%) improves resolution of these forms

Understanding these modified forms is critical, as they represent different functional states of NFE2L2 and may serve as more specific biomarkers in disease contexts.

How can NFE2L2 antibodies contribute to understanding neurodegenerative disease mechanisms?

NFE2L2 antibodies are proving valuable for investigating neurodegenerative disease mechanisms:

  • Alzheimer's disease models: In mouse models co-expressing human APP V717I and MAPT P301L, NFE2L2 deficiency leads to increased intraneuronal protein aggregates and reduced levels of autophagy proteins (SQSTM1/p62, CALCOCO2/NDP52, ULK1, ATG5, GABARAPL1). NFE2L2 antibodies allow visualization of these changes through immunohistochemistry and immunofluorescence

  • Human patient samples: In AD patients, neurons expressing high levels of APP or MAPT also express SQSTM1/p62 and nuclear NFE2L2, suggesting attempted clearance of intraneuronal aggregates through autophagy. This can be detected using co-immunostaining approaches

  • Therapeutic response markers: As NFE2L2 activators emerge as potential therapeutics for neurodegenerative diseases, antibodies can be used to confirm target engagement and downstream pathway activation

  • Selective vulnerability assessment: NFE2L2 antibodies can help identify neuronal populations with differential NFE2L2 activity, potentially explaining selective vulnerability in neurodegenerative diseases

These applications highlight NFE2L2 antibodies as important tools for both basic and translational research in neurodegenerative disease mechanisms.

What methodological considerations are important when using NFE2L2 antibodies to evaluate cancer therapeutic resistance mechanisms?

When studying cancer therapeutic resistance using NFE2L2 antibodies, several methodological considerations are crucial:

  • Mutation-specific detection: NFE2L2 mutations in the ETGE or DLG domains that abrogate KEAP1 association lead to constitutive NFE2L2 nuclear translocation and therapeutic resistance. Antibodies must be validated to detect both wild-type and mutant forms equally

  • Quantitative image analysis: For accurate assessment of nuclear/cytoplasmic ratios in patient samples, standardized immunohistochemistry protocols combined with digital image analysis are recommended

  • Combination with genomic analysis: Recent clinical studies demonstrate that mutations in NFE2L2/KEAP1/CUL3 predict radiation treatment failure in T2N0 glottic cancer (HR = 3.50; 95% CI, 1.56–7.89; P = 0.0025 for local failure). Correlating antibody staining patterns with mutation status provides the most comprehensive assessment

  • Temporal dynamics monitoring: Serial sampling during treatment can reveal evolution of NFE2L2 activation as resistance emerges

  • Downstream target assessment: Combining NFE2L2 antibodies with antibodies against downstream targets provides functional validation of pathway activation

These methodological approaches support more precise characterization of NFE2L2's role in treatment resistance, potentially guiding personalized therapy decisions.

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.