EGR2 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Stored at -20°C, Avoid freeze-thaw cycles.
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your orders. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
AT591 antibody; CMT1D antibody; CMT4E antibody; DKFZp686J1957 antibody; E3 SUMO-protein ligase EGR2 antibody; Early growth response 2 antibody; Early growth response protein 2 antibody; EGR-2 antibody; egr2 antibody; EGR2_HUMAN antibody; FLJ14547 antibody; KROX 20 Drosophila homolog antibody; Krox 20 homolog Drosophila antibody; KROX-20; Drosophila; homolog (early growth response-2) antibody; KROX20 antibody; Krox20 protein antibody; Zinc finger protein Krox-20 antibody
Target Names
Uniprot No.

Target Background

Function
EGR2, also known as Krox20, is a sequence-specific DNA-binding transcription factor. It plays a critical role in hindbrain segmentation by regulating the expression of a subset of homeobox-containing genes. Additionally, EGR2 is involved in Schwann cell myelination, regulating the expression of genes responsible for myelin formation and maintenance.

EGR2 binds to two EGR2-consensus sites, EGR2A (5'-CTGTAGGAG-3') and EGR2B (5'-ATGTAGGTG-3'), within the HOXB3 enhancer, promoting HOXB3 transcriptional activation. It also binds to specific DNA sites located in the promoter region of HOXA4, HOXB2, and ERBB2.

EGR2 regulates hindbrain segmentation by controlling the expression of Hox genes, such as HOXA4, HOXB3, and HOXB2, thereby specifying odd and even rhombomeres. It specifically promotes the expression of HOXB3 in the rhombomere r5 of the hindbrain.

Following birth, EGR2 regulates myelination in the peripheral nervous system. This is likely achieved through regulating the expression of myelin proteins, such as MPZ, and promoting the differentiation of Schwann cells.

EGR2 participates in the development of the jaw opener musculature, potentially by influencing its innervation through trigeminal motor neurons. It may also play a role in adipogenesis, possibly by regulating the expression of CEBPB.

EGR2 functions as an E3 SUMO-protein ligase, facilitating SUMO1 conjugation to its coregulators NAB1 and NAB2. Sumoylation of these coregulators downregulates EGR2's transcriptional activity.
Gene References Into Functions
  1. Low EGR2 expression is associated with hepatocellular carcinoma. PMID: 28542387
  2. The study suggests that acetylation of EGR2 is regulated independently of nucleosome remodeling and deacetylase. PMID: 28576496
  3. Egr2-driven cell surface proteins LAG-3 and 4-1BB can identify dysfunctional tumor antigen-specific CD8(+) TIL. PMID: 28115575
  4. Egr2 and Egr3 have emerged as regulatory molecules that suppress excessive immune responses. Mice deficient for Egr2 and Egr3 develop a lupus-like disease with dysregulated activation of effector T cells. Egr2 and Egr3 confer suppressive activity to CD4(+) T cells and regulate the production of inhibitory cytokines such as IL-10 and TGF-beta1. PMID: 27856665
  5. Analysis of consensus EGR-binding elements (EBEs) showed that the immediate early response 3 gene (IER3) is a novel transcriptional target gene of EGR2 as confirmed by the luciferase assay, electrophoretic mobility-shift assay (EMSA), chromatin immunoprecipitation (ChIP), and western blot analysis. PMID: 27890615
  6. In the PPI network, genes may be involved in Down syndrome (DS) by interacting with others, including nuclear receptor subfamily 4 group A member 2 (NR4A2)early growth response (EGR)2 and NR4A2EGR3. Therefore, RUNX1, NR4A2, EGR2, EGR3 and ID4 may be key genes associated with the pathogenesis of DS. PMID: 27667480
  7. EGR2 mutation presents as an axonal Charcot-Marie-Tooth phenotype with variable severity. PMID: 26204789
  8. MicroRNA20a promotes the proliferation and cell cycle of human osteosarcoma cells by suppressing EGR2 expression. PMID: 26238942
  9. These results suggested that EGR2 overexpression has a pivotal role in the downregulation of cytokines implicated in the pathophysiology of Guillain-Barre syndrome PMID: 26718337
  10. A recurrent mutation was identified in EGR2 which appears to be associated with the pathogenesis of schizophrenia. PMID: 26119399
  11. Knock-down of EGR2 with siRNA was demonstrated to have a similar effect as the over-expression of miR-330-3p in NSCLC cell lines PMID: 25935837
  12. Dysregulated Egr-2 is observed in some human autoimmune disorders. PMID: 25381473
  13. EGR2 knockdown inhibited proliferation, clonogenicity and spheroidal growth in vitro and induced regression of Ewing sarcoma xenografts. PMID: 26214589
  14. transcription factor early growth response gene-2 is a novel molecular switch regulating known immunomodulatory molecules in human mesenchymal stem cells. PMID: 24007274
  15. Overexpression of EGR2 significantly attenuated the oncogenic effect of miR-20a. PMID: 23924943
  16. Lack of association of EGR2 variants with bipolar disorder in Japanese population. PMID: 23747400
  17. EGR2 and EGR3 are regulated by NFkappaB and MAPK signalling pathways downstream of TNFalpha stimulation in breast adipose fibroblasts, and this in turn is upstream of CYP19A1 transcription via PI.4 PMID: 23485457
  18. data suggest a tendency of gender-specific association of EGR2 and EGR4 in schizophrenia. PMID: 22691714
  19. Charcot-Marie-Tooth neuropathy due to a novel EGR2 gene mutation exhibits a mild phenotype in a Czech family. PMID: 22546699
  20. Egr2 expression is translationally regulated via an IRES element, which is responsive to an inflammatory environment. PMID: 22915601
  21. we report on a heterozygous mutation in EGR2 in a family with a mild demyelinating form of adult-onset Charcot-Marie-Tooth disease PMID: 22734907
  22. Combined with a plausible biological function of EGR2, the EGR2 gene is a possible susceptibility gene in bipolar disorder. PMID: 22089088
  23. data further demonstrate the major role of the myelinating Schwann cell element in the regulation of EGR2 expression in the human peripheral nervous system PMID: 22522483
  24. Results identify a mutation in EGR2 in a Charcot-Marie-tooth disease patient with hypersensitivity to vincristine. PMID: 22271166
  25. Common variants near TARDBP and EGR2 are associated with susceptibility to Ewing sarcoma. PMID: 22327514
  26. Krox20 functions as a SUMO ligase for its coregulators--the Nab proteins--and that Nab sumoylation negatively modulates Krox20 transcriptional activity in vivo. PMID: 21836637
  27. TGF-beta3 regulated Egr-2 gene expression in uterine leiomyoma cells. PMID: 21703609
  28. these findings suggest that Egr-2 plays an important nonredundant role in the pathogenesis of fibrosis. PMID: 21514423
  29. EGR2 is a direct transcriptional target of p53 family that can in part mediate the p53-dependent apoptotic pathway. PMID: 21042708
  30. Schwann cells expressed myelin proteins and Krox20 which is an important regulator of peripheral myelination. PMID: 21057508
  31. EGR2 is a genetic risk factor for Systemic lupus erythematosus (SLE), in which increased gene expression may contribute to SLE pathogenesis. PMID: 20194224
  32. EGR2 mutation(amino aacid substitution) identifies a case of Charcot-Marie-Totth disease. PMID: 20513111
  33. overexpression of miR-150 in gastric cancer could promote proliferation and growth of cancer cells at least partially through directly targeting the tumor-suppressor EGR2. PMID: 20067763
  34. Frequency of mutations in the early growth response 2 gene associated with peripheral demyelinating neuropathies PMID: 12471219
  35. EGR2 mutant D355V induces a Cx32 promoter, mutant R381H doesn't. A sequence located at -216, recognized by the wild-type and the mutant D355V recombinant proteins, is relevant for promoter transactivation. PMID: 12609493
  36. we found that EGR2 could induce apoptosis in a large proportion of these lines by altering the permeability of mitochondrial membranes, releasing cytochrome c and activating caspase-3, -8, and -9. PMID: 12687019
  37. This report detected a heterozygous Asp383Tyr mutation of EGR2 in one patient with severe CMT1, Dejerine-Sottas syndrome. PMID: 12736090
  38. a possible molecular mechanism to account for down-regulation of EGR2 in tumor cells PMID: 14596916
  39. Egr-2 and Egr-3 transcription is enhanced by Hepatitis B virus X protein, which induces fasL gene expression PMID: 15173177
  40. A Charcot-Marie-Tooth syndrome in a family with a missense mutation in EGR2. PMID: 15947997
  41. One novel de-novo mutation (Arg359Gln, R359Q) was identified in heterozygous state in a patient with a typical CMT1 phenotype, progressive moderate thoracolumbar scoliosis and without clinical signs of cranial nerve dysfunction. PMID: 16198564
  42. Control of Krox20 transcription relies on three very long-range enhancer elements (A, B and C) that are conserved in chick, mouse and human genomes. PMID: 16495311
  43. morphological studies in the context of the I268N homozygous recessive mutation affecting the NAB repressor binding site and the R359W dominant-negative mutation in the zinc-finger domain PMID: 17717711
  44. Collectively, these data indicate that EGR2 is specifically upregulated by a parasite-derived secreted factor that is most likely a resident rhoptry protein. PMID: 18678671
  45. Dysregulation of EGR2 and MeCP2 plays roles in in Rett syndrome and autism. PMID: 19000991
  46. AIP2 regulates activation-induced T-cell death by suppressing EGR2-mediated FasL expression via the ubiquitin pathway PMID: 19651900

Show More

Hide All

Database Links

HGNC: 3239

OMIM: 129010

KEGG: hsa:1959

STRING: 9606.ENSP00000242480

UniGene: Hs.1395

Involvement In Disease
Neuropathy, congenital hypomyelinating or amyelinating (CHN); Charcot-Marie-Tooth disease 1D (CMT1D); Dejerine-Sottas syndrome (DSS)
Protein Families
EGR C2H2-type zinc-finger protein family
Subcellular Location
Nucleus.

Q&A

What is EGR2 and what are its key biological functions?

EGR2 (Early Growth Response Protein 2) is a zinc finger-containing transcription factor that plays crucial roles in multiple biological systems. It functions as a regulator of gene expression involved in cellular differentiation and development, particularly in the nervous and immune systems. EGR2 is predominantly expressed in the thymus and nervous system, where it contributes to T cell development and Schwann cell myelination . In the immune system, EGR2 is upregulated following T cell receptor (TCR) cross-linking and can both antagonize T cell activation and promote apoptosis under certain conditions . The protein is also known by several synonyms including KROX20, AT591, E3 SUMO-protein ligase EGR2, and Zinc finger protein Krox-20, which reflects its diverse functions across different research contexts .

How do EGR2 antibodies differ in terms of reactivity and applications?

EGR2 antibodies are available in multiple formats with varying species reactivity and application compatibility. They can be broadly categorized as monoclonal or polyclonal antibodies, each with specific advantages depending on the experimental context. Monoclonal antibodies offer high specificity for particular epitopes, while polyclonal antibodies provide broader epitope recognition which can be advantageous for certain applications .

Antibody TypeSpecies ReactivityPrimary ApplicationsAdvantages
Mouse Monoclonal (e.g., OTI1B12)Human, Mouse, RatIF, IHC, WBHigh specificity, consistent results across experiments
Rabbit Monoclonal (e.g., ARC1932)Human, Mouse, RatWB, IPExcellent for protein detection and isolation
Rabbit PolyclonalHuman, Mouse, RatWBBroad epitope recognition
Goat PolyclonalHumanELISA, WBAlternative host species for multiplexing

The selection of an appropriate antibody should be guided by the specific experimental requirements, including the target species, application method, and the need for conjugation to reporter molecules .

Why are EGR2 antibodies commonly used as Schwann cell markers?

EGR2 antibodies serve as reliable markers for Schwann cells due to the critical role of EGR2 in Schwann cell development and myelination processes. EGR2 is specifically expressed during the transition from immature to myelinating Schwann cells and is essential for the expression of myelin-specific genes . Mutations in the EGR2 gene are associated with demyelinating neuropathies such as Charcot-Marie-Tooth disease (types 1D and 4E), further highlighting its importance in Schwann cell function .

For researchers studying peripheral nerve development, injury, or diseases affecting myelin, EGR2 antibodies provide a specific tool to identify and track Schwann cells in various experimental models. When designing experiments using EGR2 as a Schwann cell marker, it is important to consider the developmental stage of the cells, as EGR2 expression is dynamically regulated during Schwann cell maturation and myelination processes.

How should researchers select the most appropriate EGR2 antibody for their specific experimental needs?

Selecting the optimal EGR2 antibody requires careful consideration of multiple factors to ensure experimental success:

  • Experimental Application: Different antibodies perform optimally in specific applications. For instance, if performing Western blotting, choose antibodies validated for WB. The rabbit monoclonal [ARC1932] antibody is suitable for Western blot and immunoprecipitation, while mouse monoclonal [OTI1B12] antibodies are validated for immunofluorescence, immunohistochemistry, and Western blot applications .

  • Species Reactivity: Ensure the antibody recognizes EGR2 in your experimental species. While some antibodies react with human, mouse, and rat EGR2 (like OTI1F10), others (such as A84367) may only react with human samples .

  • Clonality Considerations: Monoclonal antibodies provide consistent lot-to-lot reproducibility and specific epitope recognition, while polyclonal antibodies offer broader epitope recognition which can be beneficial for detecting proteins with post-translational modifications or in denaturing conditions.

  • Validation Status: Review the validation data provided by manufacturers, particularly for your specific application. For EGR2 antibodies used in flow cytometry, ensuring they've been tested in intracellular staining protocols is essential, as exemplified by the erongr2 monoclonal antibody .

  • Specificity Testing: Consider antibodies that have been tested for cross-reactivity. For instance, the erongr2 antibody has been verified not to cross-react with EGR3 based on immunoblot analysis .

The experimental question should ultimately guide antibody selection, with consideration for the cellular context in which EGR2 will be studied.

What are the optimal conditions for detecting EGR2 expression in different cell types using flow cytometry?

For optimal detection of EGR2 by flow cytometry, researchers should implement the following methodological considerations:

  • Fixation and Permeabilization: Since EGR2 is a transcription factor primarily localized in the nucleus, effective intracellular staining requires proper cell fixation and permeabilization. The Foxp3/Transcription Factor Staining Buffer Set is recommended for this purpose, as demonstrated with the erongr2 antibody .

  • Antibody Titration: Carefully titrate the antibody to determine optimal concentration. For the PE-conjugated erongr2 antibody, a recommended starting point is ≤0.25 μg per test (with a test defined as the amount staining a cell sample in 100 μL) .

  • Stimulation Conditions: When studying EGR2 in immune cells, consider that expression may require stimulation. In T cells, EGR2 is upregulated following TCR cross-linking. Non-adherent mouse splenocytes show increased EGR2 expression after stimulation with phorbol 12-myristate 13-acetate (PMA) and ionomycin .

  • Laser and Filter Settings: When using PE-conjugated antibodies, utilize appropriate excitation wavelengths (488-561 nm) and emission filters (centered around 578 nm) for optimal signal detection .

  • Controls: Include fluorescence minus one (FMO) controls and isotype controls to accurately identify positive populations, particularly when analyzing heterogeneous samples like splenocytes where EGR2 may be expressed in subpopulations of T cells and B cells .

For analyzing EGR2 in the context of T cell exhaustion or tumor-infiltrating lymphocytes, consider dual staining with markers like LAG-3 and 4-1BB, as these have been identified as surface markers associated with EGR2 expression in dysfunctional T cells .

What methodological approaches ensure reliable results when using EGR2 antibodies in Western blot applications?

To achieve reliable and reproducible Western blot results with EGR2 antibodies, researchers should implement the following methodological approaches:

  • Sample Preparation: EGR2 is a nuclear protein, so effective nuclear extraction protocols are essential. Consider using specialized nuclear extraction buffers containing appropriate protease inhibitors to preserve protein integrity.

  • Denaturation Conditions: Optimize denaturation conditions based on the specific EGR2 antibody. Some antibodies perform better under reducing conditions, while others may require non-reducing conditions to preserve epitope recognition.

  • Gel Percentage Selection: EGR2 has a molecular weight of approximately 50-55 kDa, making 10% acrylamide gels suitable for optimal separation and resolution.

  • Transfer Parameters: Use PVDF membranes for optimal protein binding and consider semi-dry transfer systems for efficient transfer of transcription factors like EGR2.

  • Blocking Optimization: Test both BSA and non-fat milk blocking solutions, as some EGR2 antibodies may show cross-reactivity or background with specific blocking agents.

  • Antibody Dilution and Incubation: For primary EGR2 antibodies, start with manufacturer-recommended dilutions (typically 1:500 to 1:1000) and optimize as needed. Extended overnight incubation at 4°C often improves signal quality for transcription factors.

  • Signal Development Strategy: For low-abundance expression, consider enhanced chemiluminescence (ECL) systems with extended exposure times or fluorescent secondary antibodies for quantitative analysis.

  • Positive Controls: Include positive control lysates from cells known to express EGR2, such as stimulated T cells or Schwann cell lines, to validate antibody performance .

When troubleshooting weak signals, consider concentrating nuclear extracts or using antibodies validated specifically for Western blot applications, such as the rabbit monoclonal [ARC1932] antibody or polyclonal antibodies that recognize multiple epitopes .

How can EGR2 antibodies be used to investigate the role of EGR2 in T cell exhaustion?

EGR2 has emerged as a critical regulator of T cell exhaustion, making EGR2 antibodies valuable tools for studying this immunological phenomenon. Researchers can implement the following approaches:

  • Flow Cytometric Identification of Exhausted T Cell Populations: EGR2 antibodies can be used in conjunction with surface markers LAG-3 and 4-1BB to identify exhausted T cells. This combination has been demonstrated to effectively identify dysfunctional tumor antigen-specific CD8+ tumor-infiltrating lymphocytes (TILs) . Multi-parameter flow cytometry panels can be designed to simultaneously analyze EGR2 expression alongside exhaustion markers (PD-1, TIM-3, LAG-3, 4-1BB) and functional readouts (IFNγ, TNFα, IL-2).

  • Chromatin Immunoprecipitation (ChIP) Analysis: EGR2 antibodies can be employed in ChIP experiments to identify the genomic binding sites of EGR2 in exhausted T cells, revealing direct transcriptional targets that mediate the exhaustion program. Recent research indicates that EGR2 both directly controls key exhaustion-associated genes and indirectly maintains the exhausted epigenetic state .

  • Temporal Analysis of Exhaustion Development: By examining EGR2 expression at different time points during chronic infection or in tumor models, researchers can track the kinetics of exhaustion development. Studies have shown that chronic antigen induces EGR2 selectively within progenitor exhausted cells in both chronic LCMV infection and tumor models .

  • Functional Studies with Genetic Manipulation: Combining EGR2 antibody staining with genetic approaches (conditional knockout, CRISPR-Cas9) allows for functional validation of EGR2's role in T cell exhaustion. T cell-specific deletion of Egr2 has been shown to alter the differentiation trajectory of exhausted cells .

  • Analysis of Progenitor vs. Terminal Exhausted Cells: EGR2 antibodies can help distinguish between exhaustion states, as EGR2 maintains the differentiation competency of progenitor exhausted cells and enables terminal exhaustion .

These approaches have revealed that EGR2 plays a crucial role in stabilizing the exhausted transcriptional state, making it a potential therapeutic target for reinvigorating exhausted T cells in cancer immunotherapy contexts.

What insights have been gained about EGR2's role in autoimmune disease pathogenesis through antibody-based studies?

Antibody-based studies investigating EGR2 have provided significant insights into its role in autoimmune disease pathogenesis, particularly in multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE):

  • Elevated Expression in Autoimmune Conditions: EGR2 antibody staining has revealed significantly elevated EGR2 levels in myelin-reactive CD4+ T cells from multiple sclerosis patients and mice with autoimmune neuroinflammation . This suggests EGR2 may be a biomarker for pathogenic T cell responses in autoimmune disorders.

  • Regulation of Pathogenic TH17 Cell Programs: Immunostaining and flow cytometry with EGR2 antibodies have demonstrated that EGR2 controls the transcriptional program of pathogenic TH17 cells in the central nervous system (CNS), but not protective TH17 cells at barrier sites . This tissue-specific function highlights EGR2's context-dependent role.

  • Mechanistic Analysis of CNS Inflammation: Studies have shown that approximately 30% of CNS-infiltrating CD4+ T cells express EGR2 protein at the peak of EAE disease. Further functional studies with T cell-specific deletion of Egr2 resulted in attenuated EAE in mouse models .

  • Regulation of Inflammatory Chemokines: EGR2 has been found to enhance TH17 cell differentiation and myeloid cell recruitment to the CNS by upregulating pathogenesis-associated genes and myelomonocytic chemokines . This suggests that EGR2 antibodies could potentially be used to monitor disease activity or treatment response.

  • Inflammatory IFNγ Production: Studies have shown that EGR2 positively regulates inflammatory IFNγ production , providing another mechanism by which EGR2 may contribute to autoimmune pathogenesis.

These findings collectively suggest that modulation of EGR2 activity could represent a therapeutic approach for autoimmune diseases, particularly those involving pathogenic T cell responses. EGR2 antibodies serve as valuable tools for monitoring EGR2 expression as a biomarker of disease activity or treatment response in research models and potentially in clinical settings.

How can researchers effectively analyze the interaction between EGR2 and its transcriptional targets using antibody-based approaches?

Analyzing interactions between EGR2 and its transcriptional targets requires sophisticated antibody-based approaches that extend beyond simple detection. Researchers can implement the following methodological strategies:

  • Chromatin Immunoprecipitation (ChIP): EGR2 antibodies can be used in ChIP assays to identify genomic binding sites. This technique allows for genome-wide mapping of EGR2 binding sites when coupled with next-generation sequencing (ChIP-seq) or analysis of specific target regions when followed by quantitative PCR (ChIP-qPCR). Research has revealed that EGR2 binding is intricately woven within the TH17 cell transcriptional regulatory network and shows high interconnectivity with core TH17 cell-specific transcription factors .

  • Sequential ChIP (ChIP-reChIP): This technique involves sequential immunoprecipitation with EGR2 antibodies followed by antibodies against other transcription factors to identify genomic regions where EGR2 co-localizes with other regulatory proteins, revealing collaborative transcriptional regulation.

  • Proximity Ligation Assay (PLA): This technique can detect protein-protein interactions between EGR2 and other transcriptional regulators at the single-cell level, providing spatial information about where these interactions occur within cells.

  • Co-Immunoprecipitation (Co-IP): EGR2 antibodies can be used to immunoprecipitate EGR2 along with its interacting proteins, followed by mass spectrometry or Western blotting to identify protein complexes that include EGR2.

  • Reporter Gene Assays: When combined with site-directed mutagenesis of EGR2 binding sites, these assays can determine the functional significance of EGR2 binding to specific promoter regions.

A study examining Egr2-driven gene expression in CD8+ TILs identified several target genes, demonstrating the utility of these approaches. Analysis of 4-1BB+LAG-3+ CD8+ TILs showed increased expression of 10 out of 43 Egr2 target genes that had been previously identified in in vitro anergic CD4+ T cell clones . This finding illustrates how antibody-based approaches can connect transcriptional programs across different T cell states and lineages.

What are common challenges when working with EGR2 antibodies and how can researchers address them?

Researchers may encounter several challenges when working with EGR2 antibodies. Here are common issues and their methodological solutions:

  • Low Signal Intensity in Western Blots:

    • Challenge: EGR2 is often expressed at relatively low levels in some cell types.

    • Solution: Optimize protein extraction with specialized nuclear extraction buffers, concentrate samples, increase antibody concentration, extend incubation time, or use enhanced chemiluminescence detection systems with higher sensitivity. Consider using monoclonal antibodies specifically validated for Western blot, such as the rabbit monoclonal [ARC1932] antibody .

  • Background Staining in Immunohistochemistry:

    • Challenge: Non-specific binding can obscure true EGR2 signals.

    • Solution: Implement more stringent blocking procedures using both protein blockers (BSA, normal serum) and chemical blockers to reduce endogenous peroxidase activity. Optimize antibody dilution and consider using monoclonal antibodies like [OTI1B12] which are validated for IHC applications .

  • Cross-Reactivity with Related Proteins:

    • Challenge: EGR family members share structural similarities.

    • Solution: Select antibodies specifically tested for cross-reactivity, such as the erongr2 antibody which has been verified not to cross-react with EGR3 . Include appropriate controls, including samples from EGR2-knockout models when available.

  • Variable Detection in Flow Cytometry:

    • Challenge: EGR2 is a nuclear protein requiring effective permeabilization.

    • Solution: Use specialized buffers designed for nuclear transcription factors, such as the Foxp3/Transcription Factor Staining Buffer Set . Consider stimulation conditions that upregulate EGR2 expression for more reliable detection.

  • Inconsistent Results Across Experiments:

    • Challenge: Variation in EGR2 expression under different experimental conditions.

    • Solution: Standardize cell stimulation protocols, timing of analysis, and antibody incubation conditions. Include positive controls (stimulated T cells) and negative controls in each experiment.

  • Fixation-Dependent Epitope Masking:

    • Challenge: Some fixation methods can mask EGR2 epitopes.

    • Solution: Test different fixation protocols (paraformaldehyde concentrations, methanol fixation) to determine optimal conditions for epitope preservation and accessibility.

Carefully reviewing the validation data provided with each antibody and performing preliminary optimization experiments can significantly improve the reliability and reproducibility of results when working with EGR2 antibodies.

How should researchers interpret discrepancies in EGR2 detection across different experimental platforms or antibodies?

When faced with discrepancies in EGR2 detection across experimental platforms or antibodies, researchers should adopt a systematic approach to data interpretation:

  • Antibody Epitope Consideration: Different antibodies recognize distinct epitopes on the EGR2 protein. Monoclonal antibodies target specific epitopes, while polyclonal antibodies recognize multiple epitopes. Discrepancies may reflect epitope-specific differences in accessibility, post-translational modifications, or protein conformation states. Compare the epitope information for different antibodies and consider how sample preparation might affect epitope availability.

  • Expression Threshold Analysis: Different detection methods have varying sensitivity thresholds. Western blotting may detect abundant EGR2 expression but miss low-level expression detectable by more sensitive methods like qRT-PCR or highly sensitive immunofluorescence. Consider hierarchical validation using multiple techniques with different detection limits.

  • Context-Dependent Expression Patterns: EGR2 expression is highly context-dependent and dynamically regulated. For instance, in T cells, EGR2 may be transiently expressed and subsequently downregulated after inducing downstream targets like LAG-3 and 4-1BB . Temporal analysis across multiple timepoints can resolve apparent discrepancies.

  • Cellular Heterogeneity Assessment: In heterogeneous samples, bulk analysis techniques may mask cell type-specific expression patterns. Flow cytometry data from CD8+ tumor-infiltrating lymphocytes showed that only a subset expressed EGR2, and even within the EGR2-GFP reporter positive cells, there was heterogeneity in LAG-3 and 4-1BB expression . Single-cell resolution techniques are preferable for heterogeneous populations.

  • Post-Translational Modification Impact: EGR2 function is regulated by post-translational modifications that may affect antibody binding. Different antibodies may have differential sensitivity to modifications like phosphorylation, ubiquitination, or SUMOylation, especially given that EGR2 functions as an E3 SUMO-protein ligase .

  • Integrated Data Interpretation Framework: Develop an integrated framework that considers results from multiple antibodies and detection methods. When discrepancies arise, prioritize data from techniques with appropriate controls, validated antibodies, and methods that include genetic validation (e.g., EGR2 knockout controls).

By systematically analyzing these factors, researchers can transform apparent discrepancies into insights about the complex biology of EGR2 expression and function in different cellular contexts.

What controls are essential when validating the specificity of EGR2 antibodies in experimental systems?

Rigorous validation of EGR2 antibody specificity requires a comprehensive set of controls tailored to each experimental system:

  • Genetic Negative Controls:

    • EGR2 Knockout/Knockdown: The gold standard negative control is cells or tissues with genetic ablation of EGR2. Studies have utilized T cell-specific deletion of Egr2 (Egr2ΔT mice) or more targeted deletion in specific cell populations (Egr2ΔIL17A mice) . For in vitro work, CRISPR-Cas9 knockout or siRNA knockdown of EGR2 provides excellent specificity controls.

    • Heterologous Expression Systems: Comparing EGR2-transfected versus empty vector-transfected cells can confirm antibody specificity, particularly useful for antibodies without available knockout models.

  • Biological Specificity Controls:

    • Expression Modulation: Utilize conditions known to upregulate or downregulate EGR2 expression. For instance, stimulating T cells with phorbol 12-myristate 13-acetate (PMA) and ionomycin has been demonstrated to upregulate EGR2 in non-adherent mouse splenocytes .

    • Developmental Stage Comparison: For Schwann cells, comparing developmental stages with known differences in EGR2 expression provides biological validation.

  • Technical Validation Controls:

    • Peptide Blocking: Pre-incubation of the antibody with the immunizing peptide should eliminate specific staining.

    • Isotype Controls: Include appropriate isotype-matched control antibodies to assess non-specific binding, particularly important for flow cytometry and immunohistochemistry.

    • Secondary Antibody Controls: Samples incubated with secondary antibody alone identify background from non-specific secondary antibody binding.

  • Cross-Reactivity Assessment:

    • Related Protein Expression: Test antibody specificity against related EGR family members (EGR1, EGR3, EGR4) through Western blotting of recombinant proteins or cells selectively expressing each family member. The erongr2 antibody, for example, has been validated not to cross-react with EGR3 through immunoblot analysis .

    • Species Cross-Reactivity: Validate antibody reactivity across species if using in comparative studies, as some antibodies are species-specific while others recognize human, mouse, and rat EGR2 .

  • Method-Specific Controls:

    • For ChIP Experiments: Include IgG control immunoprecipitations and test antibody enrichment at known EGR2 binding sites versus non-target regions.

    • For Flow Cytometry: Implement fluorescence minus one (FMO) controls to establish gating strategies, particularly important when analyzing EGR2 in conjunction with other markers like LAG-3 and 4-1BB .

Comprehensive validation using these controls ensures that experimental observations truly reflect EGR2 biology rather than technical artifacts or non-specific antibody interactions.

How might EGR2 antibodies contribute to therapeutic development for autoimmune diseases?

EGR2 antibodies have significant potential to advance therapeutic development for autoimmune diseases through several research pathways:

  • Biomarker Development for Patient Stratification: EGR2 antibodies could enable identification of patient subsets with elevated EGR2 expression in specific T cell populations. Research has shown that EGR2 is significantly elevated in myelin-reactive CD4+ T cells from multiple sclerosis patients . This stratification could identify patients likely to respond to therapies targeting EGR2-dependent pathways.

  • Target Validation in Preclinical Models: EGR2 antibodies can help validate the therapeutic potential of targeting EGR2 in preclinical models. Studies demonstrate that T cell-specific deletion of Egr2 attenuated experimental autoimmune encephalomyelitis (EAE) in mouse models , suggesting that EGR2 inhibition could be therapeutically beneficial.

  • Mechanism of Action Studies for Novel Therapeutics: For compounds designed to modulate EGR2 expression or function, EGR2 antibodies provide essential tools for confirming on-target activity. Flow cytometry with EGR2 antibodies can quantify changes in EGR2 protein levels, while ChIP assays can assess altered binding to target genes.

  • Pharmacodynamic Biomarker Development: EGR2 antibodies could serve as tools for pharmacodynamic monitoring in clinical trials of immunomodulatory therapies. By measuring EGR2 levels in patient samples before and after treatment, researchers can determine whether therapies effectively modulate EGR2-dependent pathways.

  • Theranostic Applications: Coupling imaging agents to EGR2 antibodies could potentially enable visualization of pathogenic T cell infiltration in affected tissues, allowing for both diagnostic assessment and therapeutic monitoring in conditions like multiple sclerosis.

  • Target Gene Analysis for Alternative Therapeutic Approaches: EGR2 antibodies used in ChIP-seq experiments can identify critical downstream targets that might present more accessible therapeutic opportunities than directly targeting EGR2. Research has revealed that EGR2 enhances TH17 cell differentiation and myeloid cell recruitment by upregulating specific pathogenesis-associated genes and myelomonocytic chemokines .

The development of these applications will require further validation of EGR2 antibodies in clinical samples and standardization of protocols for biomarker assessment, but the foundation established by current research suggests promising avenues for translation to therapeutic development.

What are the emerging applications for EGR2 antibodies in cancer immunotherapy research?

EGR2 antibodies are becoming increasingly valuable tools in cancer immunotherapy research, with several emerging applications:

  • Identification and Characterization of Exhausted T Cell Subsets: EGR2 antibodies, particularly when used in conjunction with surface markers like LAG-3 and 4-1BB, enable precise identification of dysfunctional tumor antigen-specific CD8+ tumor-infiltrating lymphocytes (TILs) . This application is critical for understanding the tumor immune microenvironment and the impact of immunotherapies on T cell functionality.

  • Mechanistic Studies of Checkpoint Inhibitor Therapy: EGR2 has been identified as a regulator of exhaustion that maintains the differentiation competency of progenitor exhausted cells . EGR2 antibodies can help investigate how checkpoint inhibitors like anti-PD-1 impact the EGR2-dependent transcriptional and epigenetic programs, providing mechanistic insights into treatment response and resistance.

  • Predictive Biomarker Development: Analysis of EGR2 expression patterns in TILs using flow cytometry or immunohistochemistry with EGR2 antibodies may help predict response to immunotherapy. Since EGR2 controls the exhausted T cell state, its expression profile could indicate the potential for T cell reinvigoration with checkpoint blockade.

  • Target Discovery for Next-Generation Immunotherapies: ChIP-seq experiments using EGR2 antibodies can identify direct transcriptional targets of EGR2 in exhausted T cells. These targets may represent novel therapeutic opportunities to reverse T cell dysfunction in combination with existing immunotherapies.

  • Monitoring Cellular Therapy Products: For adoptive cell therapies (e.g., CAR-T cells, TILs), EGR2 antibodies can help assess the exhaustion status during ex vivo expansion and after infusion, potentially guiding optimization of manufacturing protocols to minimize exhaustion.

  • Studying Resistance Mechanisms: In tumors that develop resistance to immunotherapy, EGR2 antibodies can help investigate whether changes in EGR2 expression or function contribute to resistance by promoting terminal exhaustion of tumor-reactive T cells .

  • Rational Combination Therapy Design: Understanding the interplay between EGR2 and other pathways using antibody-based approaches can inform the design of rational combination therapies targeting multiple aspects of T cell dysfunction simultaneously.

These emerging applications highlight the importance of EGR2 antibodies as both research tools and potential components of clinical assays in cancer immunotherapy development.

How can researchers integrate EGR2 antibody-based findings with other techniques for comprehensive understanding of EGR2 function?

Researchers can achieve a comprehensive understanding of EGR2 function by integrating antibody-based findings with complementary technologies and approaches:

  • Multi-Omics Integration: Combine antibody-based protein detection with transcriptomic and epigenomic analyses. For instance, integrate ChIP-seq data using EGR2 antibodies with RNA-seq to correlate EGR2 binding with gene expression changes, and with ATAC-seq to understand how EGR2 affects chromatin accessibility. This approach has revealed that EGR2 maintains the exhausted transcriptional state through both direct control of key exhaustion-associated genes and indirect maintenance of the exhausted epigenetic state .

  • Single-Cell Resolution Techniques: Pair antibody-based flow cytometry with single-cell RNA-seq to correlate EGR2 protein levels with transcriptional states at the single-cell level. This approach can reveal heterogeneity within cell populations that appear homogeneous by bulk analysis, as demonstrated by the identification of diverse exhausted T cell states in tumor microenvironments .

  • Spatial Context Analysis: Combine immunohistochemistry using EGR2 antibodies with spatial transcriptomics to understand the relationship between EGR2 expression, cellular localization, and the tissue microenvironment. This is particularly relevant for understanding EGR2's context-dependent roles, such as its function in pathogenic TH17 cells in the CNS versus protective TH17 cells at barrier sites .

  • Functional Genomics Validation: Validate antibody-based findings using genetic approaches like CRISPR-Cas9 gene editing to manipulate EGR2 expression or function. Studies using T cell-specific deletion of Egr2 have confirmed its role in autoimmune neuroinflammation and T cell exhaustion .

  • Temporal Analysis: Implement time-course experiments with antibody detection to track dynamic changes in EGR2 expression and function. This approach has revealed that EGR2 may be transiently expressed before inducing stable expression of surface markers like LAG-3 and 4-1BB .

  • In Vivo Models with Reporter Systems: Combine antibody validation with in vivo models expressing fluorescent reporters for EGR2 (like the Egr2-IRES-GFP knock-in reporter mouse) to track EGR2-expressing cells longitudinally during disease progression or therapeutic intervention .

  • Systems Biology Approaches: Integrate antibody-based data into computational models of gene regulatory networks to understand EGR2's place within broader signaling cascades and transcriptional programs. This has revealed EGR2's interconnectivity with core TH17 cell-specific transcription factors and its role in pathogenic versus protective TH17 cell programs .

By implementing this integrative approach, researchers can overcome the limitations of individual techniques and develop a comprehensive understanding of EGR2's multifaceted roles in development, immune function, and disease pathogenesis.

What critical gaps in EGR2 research could be addressed through improved antibody-based methodologies?

Several critical gaps in EGR2 research could be addressed through improved antibody-based methodologies:

  • Tissue-Specific EGR2 Function: Current research indicates context-dependent roles for EGR2, such as its differential function in pathogenic versus protective TH17 cells . Developing antibodies that recognize tissue-specific post-translational modifications or conformational states of EGR2 could help elucidate how the same transcription factor performs distinct functions in different cellular environments.

  • Temporal Dynamics of EGR2 Activity: Evidence suggests that EGR2 may be transiently expressed in some contexts, like T cell exhaustion, with lasting effects on cellular programming . Improved methods for real-time antibody-based tracking of EGR2 expression and activity in living cells could provide insights into these temporal dynamics.

  • EGR2 Protein Interaction Networks: While EGR2's DNA binding sites are increasingly well-characterized, less is known about its protein interaction partners that may mediate context-specific functions. Developing antibodies optimized for techniques like proximity ligation assays or mass spectrometry-based interactome analysis could reveal these critical interactions.

  • Conformational States and Functional Domains: Antibodies that specifically recognize different conformational states or functional domains of EGR2 could help determine how structural changes or domain-specific interactions regulate EGR2 function, potentially revealing new therapeutic approaches targeting specific EGR2 activities.

  • Translation to Human Diseases: While much has been learned from mouse models, translation to human disease contexts remains challenging. Developing and validating antibodies that specifically recognize human EGR2 with high sensitivity and specificity in clinical samples could bridge this gap, enabling more robust biomarker studies in conditions like multiple sclerosis and cancer.

  • Intracellular Trafficking and Localization: EGR2's subcellular localization may regulate its function, but this aspect remains understudied. Antibodies optimized for high-resolution imaging techniques could reveal how EGR2 trafficking between nuclear and cytoplasmic compartments affects its activity.

  • EGR2 in Non-Immune Cell Types: While EGR2's roles in T cells and Schwann cells are increasingly well-characterized, its functions in other cell types remain obscure. Developing antibodies validated across diverse cell types could expand our understanding of EGR2's broader biological functions.

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.