Based on available literature, "ERF7 Antibody" can refer to antibodies targeting different proteins, depending on the context. One possibility is antibodies targeting Ethylene Response Factor 7 (ERF7), a plant-specific transcription factor involved in various stress responses . Another possibility is that ERF7 Antibody may be related to Estrogen Receptor-β (ERβ) antibodies, which are used in cancer research . It is crucial to clarify the specific target of the "ERF7 Antibody" to provide accurate information.
ERF7 is a member of the ethylene response factor (ERF) family of transcription factors in plants . These proteins play crucial roles in:
Mature ERF-VII proteins possess an N-terminal Cys (Cys2), which acts as a degradation signal, targeting these proteins to the 26S proteasome .
Estrogen Receptor-β (ERβ) is implicated in several cancers, and its function is debated in prostate and breast cancer . Genetic studies suggest a role in cancer progression . Research has shown that many commercially available ERβ antibodies have limited specificity or are non-specific .
Antibodies, including those targeting ERF7 or ERβ, are utilized in various experimental modalities . Common applications include:
Selecting the appropriate antibody is crucial for accurate and reliable research outcomes. Important factors to consider include:
Specificity: Ensuring the antibody binds to the intended target protein and not to others .
Reactivity: Verifying the antibody's reactivity with the target protein in the species of interest .
Validation: Checking if the antibody has been validated for the intended application .
HERV-K Env Antibodies: Monoclonal antibodies against the Human Endogenous Retrovirus-K envelope protein show potential as immunotherapeutic agents for breast cancer therapy .
EGFR Inhibitors: Studies have identified potent Epidermal Growth Factor Receptor (EGFR) inhibitors using fragment-based drug design, offering potential strategies to overcome drug resistance in non-small-cell lung cancer .
ERα Reporters: Fluorescent protein reporter cell lines for Estrogen Receptor alpha (ERα) have been established to assess the carcinogenic hazards of estrogenic compounds .
EGFR Degraders: Research has led to the discovery of potent and selective EGFR bifunctional small-molecule degraders, which can effectively degrade mutant EGFR proteins in cancer cells .
The below tables show how antibodies are selected by reactivity, application and host.
ERF7 (Ethylene Response Factor 7) belongs to the ERF/AP2 transcription factor family, which plays crucial roles in plant stress responses. ERF proteins contain the AP2 DNA-binding domain and are classified into various subfamilies. The ERF-VII subfamily includes key members like RAP2.2, RAP2.12, RAP2.3, HRE1 (ERF73), and HRE2 that regulate hypoxia-responsive genes . While our search results do not specifically detail ERF7, based on the ERF family characteristics, it likely functions as a transcription factor involved in ethylene signaling pathways and stress responses similar to other family members such as ERF71 and ERF73 .
ERF7 antibodies are valuable tools for:
Protein detection via Western blotting
Protein localization using immunohistochemistry
Chromatin immunoprecipitation (ChIP) assays to identify DNA binding sites
Protein-protein interaction studies
The methodological approach for these applications would be similar to those used with other ERF family antibodies, such as the chromatin immunopurification techniques used with RAP2.2 and RAP2.12 to study binding to hypoxia-responsive promoter elements (HRPEs) .
For experimental validity when using ERF7 antibodies, researchers should include:
| Control Type | Purpose | Implementation |
|---|---|---|
| Positive control | Confirms antibody functionality | Tissue with known ERF7 expression |
| Negative control | Assesses non-specific binding | ERF7 knockout/knockdown tissue |
| Loading control | Ensures equal protein loading | Housekeeping protein detection (e.g., TUBULIN) |
| Secondary antibody control | Evaluates background signal | Primary antibody omission |
Similar controls were employed in ERF-VII transcription factor research, where researchers used wild-type Col-0 as negative controls when analyzing FLAG-tagged RAP2.2 or RAP2.12 binding to promoter regions .
ERF7 antibodies can be employed in chromatin immunoprecipitation (ChIP) experiments to identify genomic regions bound by ERF7. This approach would be similar to the ChIP methodology used for RAP2.2 and RAP2.12, which demonstrated binding to specific promoter regions containing hypoxia-responsive promoter elements (HRPEs) .
The procedure would involve:
Cross-linking proteins to DNA in plant tissues
Shearing chromatin to 200-400bp fragments
Immunoprecipitating with ERF7 antibody
Purifying and analyzing co-precipitated DNA by qPCR or sequencing
Identifying ERF7 binding sites based on enrichment patterns
For robust results, researchers should include appropriate controls such as wild-type samples without tagged proteins, as demonstrated in the FLAG-RAP2.2 and FLAG-RAP2.12 ChIP experiments .
Validating antibody specificity is critical for meaningful research outcomes. For ERF7 antibodies, consider:
Western blot analysis using:
Recombinant ERF7 protein
Plant tissues with ERF7 overexpression
erf7 mutant tissues (should show no signal)
Competition assays with immunizing peptide
Cross-reactivity assessment:
Testing against closely related ERF proteins like other subfamily members
Performing immunoprecipitation followed by mass spectrometry
Genetic validation:
Comparing antibody signals between wild-type and erf7 knockout lines
Using complementation lines to verify restored detection
This approach aligns with methods that would be used to validate other ERF antibodies, such as those against ERF71 or ERF73 .
To investigate ERF7's role in transcriptional complexes:
Co-immunoprecipitation (Co-IP):
Use ERF7 antibodies to pull down ERF7 and associated proteins
Analyze by mass spectrometry to identify interaction partners
Confirm interactions using reverse Co-IP with antibodies against candidate partners
Proximity labeling approaches:
Express ERF7 fused to BioID or APEX2
Identify proximal proteins through biotinylation and streptavidin pulldown
Validate candidates using ERF7 antibodies
Bimolecular fluorescence complementation:
Express ERF7 fused to split fluorescent protein fragment
Co-express candidate interactors fused to complementary fragment
Validate interactions observed through fluorescence using ERF7 antibodies
Similar approaches would be applicable to studying protein interactions of other ERF family members like RAP2.2 and RAP2.12, which were shown to function redundantly in hypoxia response regulation .
For optimal ERF7 detection by Western blot:
Sample preparation:
Extract proteins in buffer containing 50mM Tris-HCl (pH 7.5), 150mM NaCl, 1% Triton X-100
Include protease inhibitors (PMSF, leupeptin, pepstatin)
Add phosphatase inhibitors if phosphorylation status is important
For nuclear proteins like ERF7, use nuclear extraction protocols
Gel electrophoresis parameters:
Use 10-12% SDS-PAGE gels
Load 20-40μg total protein per lane
Include molecular weight markers
Transfer and detection:
Transfer to PVDF membrane (better for nuclear proteins)
Block with 5% non-fat milk or BSA
Use ERF7 antibody at optimized dilution (typically 1:1000)
Include controls as outlined in section 1.3
These recommendations are based on general practices for transcription factor detection and would be similar to those used for other ERF family proteins such as ERF71 and ERF73 .
When facing cross-reactivity issues with ERF7 antibodies:
Antibody purification techniques:
Perform affinity purification against specific ERF7 epitopes
Use competitive elution with immunizing peptide
Consider cross-adsorption against related ERF proteins
Experimental modifications:
Increase stringency of washing steps (higher salt concentration)
Optimize antibody concentration (use titration experiments)
Adjust blocking conditions (try different blocking agents)
Alternative validation approaches:
Compare results with orthogonal detection methods
Use tagged ERF7 constructs in parallel experiments
Employ genetic controls (erf7 mutants, complementation lines)
When selecting antibodies, researchers should carefully evaluate specificity information provided by suppliers, similar to what's shown for ERF71 and ERF73 antibodies in their product information .
To investigate post-translational modifications (PTMs) of ERF7:
Modification-specific detection:
Use phospho-specific antibodies if available
Combine ERF7 antibodies with PTM-specific antibodies
Employ 2D gel electrophoresis to separate modified forms
Enrichment approaches:
Immunoprecipitate with ERF7 antibodies followed by PTM detection
Use PTM-specific enrichment (e.g., phospho-peptide enrichment) before ERF7 detection
Perform sequential immunoprecipitation with ERF7 and PTM antibodies
Analytical techniques:
Mass spectrometry analysis after ERF7 immunoprecipitation
Use mobility shift assays to detect modified forms
Employ Phos-tag gels for phosphorylated protein separation
This approach would be particularly relevant for studying ERF7, as ERF-VII family proteins are known to undergo regulated proteolysis via the N-end rule pathway in an oxygen-dependent manner, similar to what has been observed with RAP2.2 and RAP2.12 .
For ChIP-seq using ERF7 antibodies:
Experimental setup:
Cross-link proteins to DNA (1% formaldehyde, 10 minutes)
Sonicate chromatin to 200-300bp fragments
Immunoprecipitate using ERF7 antibodies
Prepare sequencing libraries from immunoprecipitated DNA
Sequence using next-generation sequencing platforms
Data analysis pipeline:
Align reads to reference genome
Call peaks using MACS2 or similar software
Perform motif discovery on peak regions
Integrate with transcriptome data to identify regulated genes
Validation strategies:
Confirm selected binding sites using ChIP-qPCR
Perform reporter assays for identified promoters
Correlate binding with gene expression changes
This methodology would be similar to the approach used to identify the hypoxia-responsive promoter element (HRPE) bound by RAP2.2 and RAP2.12 in hypoxia-responsive genes, as reported in search result .
When designing experiments to study ERF7's role in stress responses:
Stress treatment design:
Include appropriate time course (early and late responses)
Use physiologically relevant stress conditions
Monitor stress markers to confirm treatment efficacy
Sampling considerations:
Collect tissue-specific samples (roots, leaves, etc.)
Consider developmental stage effects
Use flash-freezing to preserve protein modifications
Comparative analysis:
Include related ERF family members for comparison
Examine different stress types to assess specificity
Study interactions with key signaling components
These considerations would be particularly relevant as ERF family members like RAP2.2, RAP2.12, and HRE1/HRE2 have been shown to respond differentially to hypoxia and other stress conditions .
| Stress Condition | Sampling Timepoints | Key Controls |
|---|---|---|
| Hypoxia | 0, 1, 3, 6, 12, 24 hours | Normoxic samples, hypoxic marker genes |
| Drought | Early (soil moisture 70%), Medium (50%), Severe (30%) | Well-watered controls, RWC measurements |
| Pathogen infection | 0, 6, 12, 24, 48, 72 hours post-infection | Mock-infected, defense marker genes |
Integrating ERF7 antibodies with CRISPR-Cas9 approaches:
Validation of knockout/knockdown:
Use ERF7 antibodies to confirm protein absence in CRISPR-edited lines
Quantify reduction in protein levels in partial knockdowns
Verify specificity by confirming presence of other ERF proteins
Domain function analysis:
Generate domain-specific mutations using CRISPR-Cas9
Use ERF7 antibodies to confirm stable protein expression
Compare binding patterns of wild-type vs. mutant proteins
Complementation studies:
Reintroduce wild-type or mutant ERF7 into knockout backgrounds
Use antibodies to confirm expression levels
Compare functional recovery with protein expression
This integrated approach would be similar to the techniques used to study the redundant functions of RAP2.2 and RAP2.12 through mutant analysis and complementation .
When faced with discrepancies between protein and mRNA data:
Technical considerations:
Verify antibody specificity and sensitivity
Confirm RNA analysis methods and normalization
Check for technical issues in either workflow
Biological explanations:
Consider post-transcriptional regulation mechanisms
Evaluate protein stability and turnover rates
Assess tissue-specific expression patterns
Integrated analysis approach:
Use multiple detection methods to validate results
Perform time-course experiments to capture dynamics
Consider subcellular fractionation to assess protein localization
This analytical approach is particularly relevant for ERF family proteins, as research on ERF-VII members has shown that post-translational regulation via the N-end rule pathway plays a crucial role in their function, with protein stability being regulated by oxygen levels independently of transcript abundance .
For robust statistical analysis of ERF7 ChIP data:
Peak calling statistics:
Use false discovery rate (FDR) < 0.05 for peak identification
Apply fold-enrichment thresholds (typically >2-fold over input)
Consider replicate consistency (peaks present in multiple biological replicates)
Comparative analyses:
Employ differential binding analysis between conditions
Use normalized read counts for quantitative comparisons
Apply appropriate transformations for data normalization
Integration with other data types:
Correlate binding strength with gene expression changes
Perform gene ontology enrichment on target genes
Analyze co-occurrence with other transcription factor binding sites
These statistical approaches align with methodologies that would be used when analyzing ChIP data for other ERF family members, such as the ChIP experiments conducted with FLAG-RAP2.2 and FLAG-RAP2.12 .
To distinguish direct from indirect regulatory effects:
Experimental strategies:
Combine ChIP-seq with RNA-seq from the same conditions
Use inducible expression systems with time-course sampling
Perform reporter assays with wild-type and mutated binding sites
Data integration methods:
Identify genes with both binding sites and expression changes
Analyze temporal patterns of binding and expression changes
Compare effects of ERF7 mutations on binding vs. gene expression
Network analysis approaches:
Build directed regulatory networks
Use causal inference algorithms
Implement mathematical modeling of regulatory dynamics
This approach would be similar to the methods used to identify direct targets of RAP2.2 and RAP2.12 through the identification of the hypoxia-responsive promoter element and subsequent validation using promoter mutations and transactivation assays .