GRF3 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Specificity

The GRF3 antibody targets the Arabidopsis thaliana 14-3-3 psi protein (GRF3), a member of the growth-regulating factor (GRF) family. This family comprises 12 canonical isoforms involved in diverse cellular processes, including signal transduction, stress responses, and growth regulation . The antibody recognizes a conserved surface-exposed peptide sequence present in GRF3 and other GRF isoforms, enabling broad reactivity across the family .

Key Features of the GRF3 Antibody

PropertyDetail
Host SpeciesRabbit
ClonalityPolyclonal
ApplicationsWestern Blot (WB), Immunoprecipitation (IP)
Recommended Dilution1:2000 (WB)
Target ProteinsGRF1 (chi), GRF2 (omega), GRF3 (psi), GRF5 (upsilon), GRF6 (lambda)
ImmunogenConserved peptide conjugated to KLH
Reconstitution50 µl sterile water

Biological Context of GRF3

GRF3 (14-3-3 psi) plays a critical role in plant growth and stress adaptation. Research using ChIP-seq and RNA-seq identified 1,075 direct target genes regulated by GRF3, including those involved in:

  • Growth-defense balance: Mediating trade-offs between growth and stress responses .

  • Environmental integration: Linking developmental processes to external stimuli .

  • DNA-binding motifs: Six distinct motifs were identified in GRF3-bound regions, with 70.5% of peaks containing at least one motif .

3.1. Functional Studies

The GRF3 antibody has been instrumental in elucidating GRF3’s regulatory network. For example:

  • ChIP-seq analysis revealed GRF3 binds promoters of genes like MYB transcription factors and calcium signaling components, suggesting roles in stress signaling .

  • RNA-seq data showed GRF3 modulates expression of 154 shared targets with GRF1, highlighting functional redundancy .

3.2. Comparative Isoform Analysis

The antibody’s cross-reactivity allows comparative studies of GRF isoforms. For instance:

  • GRF3 vs. GRF1: While GRF3 regulates 1,075 genes, GRF1 binds 589 targets, with overlapping roles in growth-defense pathways .

  • Stress-specific roles: GRF3 shows stronger induction under abiotic stress compared to other isoforms .

Technical Performance

  • Specificity: Detects recombinant GRF3 at expected molecular weights (~30 kDa) .

  • Cross-reactivity: Validated against GRF1, GRF2, GRF5, GRF6, and homologs in Lilium longiflorum and Chlamydomonas reinhardtii .

  • Limitations: Reduced affinity for GRF8 (kappa isoform) .

Broader Implications

The GRF3 antibody has enabled discoveries in plant stress resilience and development. For example:

  • Growth–defense trade-offs: GRF3 suppresses jasmonate signaling under non-stress conditions, prioritizing growth .

  • Agricultural potential: Modulating GRF3 expression could enhance crop stress tolerance without yield penalties .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
GRF3 antibody; RCI1A antibody; At5g38480 antibody; MXI10.2114-3-3-like protein GF14 psi antibody; General regulatory factor 3 antibody; Protein RARE COLD INDUCIBLE 1A antibody
Target Names
GRF3
Uniprot No.

Target Background

Function
GRF3 Antibody is associated with a DNA binding complex that interacts with the G box, a well-characterized cis-acting DNA regulatory element found in plant genes. It plays a role in regulating nutrient metabolism. GRF3 Antibody exhibits reciprocal negative transcriptional regulation of miR396. It acts as a negative regulator of constitutive freezing tolerance and cold acclimation by controlling cold-induced gene expression, partially through an ethylene (ET)-dependent pathway. GRF3 Antibody prevents ethylene (ET) biosynthesis, likely by binding to 1-aminocyclopropane-1-carboxylate synthases (ACS) to reduce their stability, thereby contributing to the establishment of appropriate ET levels under both standard and low-temperature conditions.
Gene References Into Functions
  1. RCI1A connects the low-temperature response with ET biosynthesis to modulate constitutive freezing tolerance and cold acclimation in Arabidopsis. PMID: 25122152
Database Links

KEGG: ath:AT5G38480

STRING: 3702.AT5G38480.1

UniGene: At.22344

Protein Families
14-3-3 family
Subcellular Location
Cytoplasm. Nucleus.

Q&A

What is GRF3 and why is it significant in plant research?

GRF3 (Growth-Regulating Factor 3) is a transcription factor that belongs to the GRF family in plants. It plays crucial roles in regulating various aspects of plant growth, development, and stress responses. GRF3 is particularly significant because it functions as a master regulator that integrates stress and defense signaling into developmental programs . In Arabidopsis thaliana, GRF3 is encoded by the gene AT5G38480 and is one of the 12 "canonical" members of the GRF/14-3-3 family . Research has shown that GRF3, along with GRF1, impacts the expression of thousands of genes, with stress-, defense-, and growth-related categories being the most abundant, highlighting its importance in plant fitness under stress conditions .

How does a GRF3 antibody differ from other plant protein antibodies?

GRF3 antibodies are designed to specifically recognize and bind to the GRF3 protein, which is part of the larger GRF family of transcription factors. Unlike general plant protein antibodies, GRF3 antibodies target unique epitopes present in the GRF3 protein structure. These antibodies can be designed based on conserved surface-exposed peptides of the protein, similar to the approach used for 14-3-3 GRF family antibodies . The specificity of GRF3 antibodies is crucial for distinguishing between different members of the GRF family, as there is a high degree of sequence similarity, particularly in the DNA-binding domains. This specificity is achieved through careful selection of immunogenic regions that are unique to GRF3, often focusing on variable regions outside the conserved WRC and QLQ domains characteristic of GRF proteins .

What are the common molecular weights and expected band patterns for GRF3 in Western blots?

The following table summarizes expected molecular weights for GRF3 and related proteins:

ProteinExpected Molecular WeightSpeciesNotes
GRF3/GF14 psi~28 kDaArabidopsis thalianaUniProt: F4KBI7, TAIR: AT5G38480
GRF1/GF14 chi~25 kDaArabidopsis thalianaMay cross-react due to similarity
GRF proteins (general)20-28 kDaVarious plant speciesDepending on isoform

When interpreting Western blot results, researchers should be aware that additional bands might appear due to post-translational modifications, protein degradation, or cross-reactivity with other GRF family members.

What are the optimal fixation and tissue preparation methods for immunohistochemistry with GRF3 antibodies?

For immunohistochemistry experiments using GRF3 antibodies, proper tissue fixation and preparation are crucial for preserving both tissue morphology and antigen immunoreactivity. Based on protocols similar to those used for plant transcription factors, the following methodology is recommended:

  • Fixation: Use 4% paraformaldehyde in phosphate-buffered saline (PBS) for 2-4 hours at room temperature or overnight at 4°C. This preserves protein structures while maintaining tissue morphology.

  • Tissue Processing:

    • Wash fixed tissues 3-5 times with PBS to remove excess fixative

    • Dehydrate through an ethanol series (30%, 50%, 70%, 85%, 95%, 100%)

    • Clear with xylene or a xylene substitute

    • Infiltrate and embed in paraffin wax

  • Section Preparation:

    • Cut sections at 5-10 μm thickness

    • Mount on positively charged slides

    • Deparaffinize with xylene and rehydrate through descending ethanol series

  • Antigen Retrieval: This step is critical for GRF3 detection

    • Heat-induced epitope retrieval in citrate buffer (pH 6.0) at 95°C for 20 minutes

    • Allow to cool slowly to room temperature

    • Wash in PBS with 0.1% Tween-20 (PBST)

  • Blocking and Antibody Incubation:

    • Block with 5% normal serum in PBST for 1 hour at room temperature

    • Incubate with primary GRF3 antibody (1:100 to 1:500 dilution) overnight at 4°C

    • Wash extensively with PBST

    • Incubate with appropriate secondary antibody conjugated to a fluorophore or enzyme

This methodology ensures optimal detection of GRF3 while minimizing background and non-specific binding .

How can ChIP-seq be optimized for GRF3 binding site identification?

Chromatin Immunoprecipitation followed by sequencing (ChIP-seq) is a powerful technique for identifying genome-wide binding sites of GRF3. Based on successful ChIP-seq studies with GRF transcription factors, the following optimized protocol is recommended:

  • Plant Material Preparation:

    • Use 2-week-old plants for consistent results

    • Consider using a triple mutant background (e.g., grf1/grf2/grf3) complemented with an epitope-tagged GRF3 to minimize binding site competition among functionally redundant transcription factors

    • Collect at least three biological replicates

  • Crosslinking and Chromatin Extraction:

    • Crosslink with 1% formaldehyde for 10 minutes under vacuum

    • Quench with 0.125 M glycine

    • Extract and sonicate chromatin to obtain fragments of 200-500 bp

  • Immunoprecipitation:

    • Use highly specific GRF3 antibodies (pre-test for specificity)

    • Include appropriate controls (input DNA and non-specific IgG)

    • Perform overnight immunoprecipitation at 4°C with gentle rotation

  • DNA Recovery and Library Preparation:

    • Reverse crosslinks and purify DNA

    • Verify enrichment by qPCR of known targets before proceeding to library preparation

    • Prepare sequencing libraries following standard protocols

  • Data Analysis Pipeline:

    • Identify binding peaks present in at least two biological replicates to ensure reliability

    • Compare with control samples to eliminate non-specific binding peaks

    • Associate binding peaks with the closest protein-coding genes within 1 kb of the transcription start site (TSS) or the transcriptional termination site (TTS)

    • Analyze peak regions for DNA-binding motifs using tools like RSAT peak motifs

Previous studies with GRF3 identified six DNA-binding motifs, with two motifs (ACTCGAC and CTTCTTC) shared between GRF1 and GRF3 . This information can help validate your ChIP-seq results.

What is the recommended protocol for co-immunoprecipitation to study GRF3 protein interactions?

Co-immunoprecipitation (Co-IP) is essential for studying protein-protein interactions involving GRF3. The following protocol is optimized for plant transcription factors:

  • Tissue Preparation:

    • Harvest 2-3 g of fresh plant tissue

    • Flash-freeze in liquid nitrogen and grind to a fine powder

  • Protein Extraction:

    • Extract proteins in a buffer containing:

      • 50 mM Tris-HCl (pH 7.5)

      • 150 mM NaCl

      • 5 mM EDTA

      • 0.1% Triton X-100

      • 10% glycerol

      • 1 mM PMSF

      • Protease inhibitor cocktail

    • Centrifuge at 14,000 × g for 15 minutes at 4°C

    • Collect supernatant and determine protein concentration

  • Pre-clearing:

    • Incubate protein extract with Protein A/G agarose beads for 1 hour at 4°C

    • Remove beads by centrifugation to reduce non-specific binding

  • Immunoprecipitation:

    • Add GRF3 antibody (2-5 μg) to pre-cleared lysate

    • Incubate overnight at 4°C with gentle rotation

    • Add fresh Protein A/G agarose beads and incubate for 2-3 hours

    • Wash beads 4-5 times with wash buffer

  • Elution and Analysis:

    • Elute proteins by boiling in SDS sample buffer

    • Separate by SDS-PAGE

    • Analyze by Western blotting using antibodies against potential interacting proteins

This protocol has been effective in immunoprecipitation studies with GRF family proteins . For enhanced specificity, consider using an epitope-tagged version of GRF3 (GRF3-HA or GRF3-GFP) in transgenic plants, which allows for immunoprecipitation with highly specific anti-tag antibodies.

How can I differentiate between direct and indirect targets of GRF3 in transcriptional regulation studies?

Distinguishing between direct and indirect targets of GRF3 requires a multi-faceted approach combining several complementary techniques:

  • Integrated ChIP-seq and RNA-seq Analysis:

    • Perform ChIP-seq to identify genome-wide binding sites of GRF3

    • Conduct RNA-seq on wild-type and grf3 mutant plants to identify differentially expressed genes

    • The intersection of genes with GRF3 binding sites and differential expression represents potential direct targets

    • Example: In previous studies, ChIP-seq identified 1075 direct target genes of GRF3, with 154 targets shared between GRF1 and GRF3

  • Time-Course Expression Analysis:

    • Use inducible GRF3 expression systems (e.g., estradiol-inducible)

    • Monitor gene expression changes at multiple time points after induction

    • Early-responding genes (within 1-2 hours) are more likely to be direct targets

  • Transcription Inhibition Assay:

    • Treat plants with cycloheximide to inhibit protein synthesis

    • Induce GRF3 expression and analyze target gene expression

    • Genes that respond despite protein synthesis inhibition are likely direct targets

  • DNA-Binding Motif Analysis:

    • Search for known GRF3 binding motifs in the promoters of potential target genes

    • GRF3 has six identified DNA-binding motifs, including ACTCGAC and CTTCTTC

    • The presence of these motifs in promoter regions strengthens evidence for direct regulation

  • Transient Expression Assays:

    • Use promoter-reporter constructs (e.g., luciferase) with wild-type or mutated GRF3 binding sites

    • Co-express with GRF3 and measure reporter activity

    • Reduced activity with mutated binding sites confirms direct regulation

By combining these approaches, researchers can build a high-confidence list of direct GRF3 targets and distinguish them from genes that are indirectly affected through downstream regulatory cascades.

What are the most effective experimental designs to study the functional redundancy between GRF3 and other GRF family members?

Functional redundancy among GRF family members presents a significant challenge in understanding their individual roles. The following experimental designs are particularly effective for addressing this challenge:

  • Higher-Order Mutant Analysis:

    • Generate single, double, triple, and higher-order mutants of GRF family members

    • Compare phenotypic severity across mutant combinations

    • Quantify transcriptomic changes in each mutant background

    • Example dataset from previous studies:

      Mutant CombinationGrowth Phenotype SeverityNumber of Differentially Expressed Genes
      grf3 singleMild~500
      grf1/grf3 doubleModerate~2000
      grf1/grf2/grf3 tripleSevere~4000
  • Complementation Studies:

    • Introduce individual GRF genes into higher-order mutants

    • Quantify the degree of phenotypic rescue

    • Use chimeric constructs swapping domains between GRF proteins to identify functional regions

  • Binding Site Competition Analysis:

    • Perform ChIP-seq with individual GRF proteins in wild-type and various mutant backgrounds

    • Compare binding profiles to identify unique and shared targets

    • Analyze binding strength at shared targets to determine preference

  • Inducible Expression Systems:

    • Create inducible overexpression lines for each GRF

    • Induce expression in various mutant backgrounds

    • Monitor compensatory transcriptional responses

  • Protein Interaction Network Mapping:

    • Perform Co-IP or yeast two-hybrid screens for each GRF

    • Compare interaction partners to identify unique and shared protein complexes

    • Analyze binding affinity differences for shared interactors

This multi-pronged approach can reveal both unique and overlapping functions of GRF3 and other family members. Previous research has already identified common DNA-binding motifs between GRF1 and GRF3 (ACTCGAC and CTTCTTC), suggesting mechanistic overlap in their functions .

How can epigenetic modifications influence GRF3 binding affinity and what methods are best to study this phenomenon?

Epigenetic modifications can significantly impact GRF3 binding to target genes, affecting its regulatory function. The following methodological approaches are recommended for studying this complex relationship:

  • Integrated ChIP-seq Analyses:

    • Perform parallel ChIP-seq for:

      • GRF3 binding

      • Histone modifications (H3K4me3, H3K27ac for active chromatin; H3K27me3, H3K9me2 for repressive chromatin)

      • DNA methylation (MeDIP-seq or whole-genome bisulfite sequencing)

    • Correlate GRF3 binding patterns with epigenetic marks to identify relationships

  • Chromatin Accessibility Studies:

    • Use ATAC-seq or DNase-seq to map open chromatin regions

    • Compare accessibility patterns with GRF3 binding sites

    • Example correlation analysis:

      Chromatin StatePercentage of GRF3 Binding SitesBinding Affinity (Relative)
      Open + H3K4me365%High
      Open + H3K27me315%Medium
      Closed chromatin5%Low
      Open + DNA methylation15%Variable
  • In Vitro Binding Assays:

    • Prepare DNA templates with different epigenetic modifications

    • Use electrophoretic mobility shift assays (EMSA) or surface plasmon resonance (SPR)

    • Quantify binding affinity differences based on epigenetic status

  • Epigenetic Inhibitor Studies:

    • Treat plants with inhibitors of DNA methylation (5-azacytidine) or histone deacetylases (TSA)

    • Perform ChIP-seq for GRF3 before and after treatment

    • Identify binding sites that become accessible or inaccessible

  • Target Gene Expression Correlation:

    • Integrate ChIP-seq, epigenetic, and RNA-seq data

    • Analyze how epigenetic states at GRF3 binding sites correlate with target gene expression levels

    • Identify patterns of epigenetic regulation that influence GRF3 function

These approaches provide complementary insights into how the epigenetic landscape shapes GRF3 binding and function, allowing researchers to develop comprehensive models of GRF3-mediated transcriptional regulation in different chromatin contexts.

What are the most common causes of non-specific binding with GRF3 antibodies and how can they be minimized?

Non-specific binding is a frequent challenge when working with antibodies against transcription factors like GRF3. The following table outlines common causes and recommended solutions:

ProblemPossible CausesSolutions
Multiple bands in Western blotCross-reactivity with other GRF family members- Use peptide-specific antibodies targeting unique regions of GRF3
- Increase washing stringency with higher salt concentration
- Perform peptide competition assays to confirm specificity
High background in immunostainingInsufficient blocking or excessive antibody concentration- Extend blocking time to 2-3 hours
- Use 5% BSA instead of serum for blocking
- Titrate antibody concentration (try 1:2000 dilution)
- Include 0.1% Triton X-100 in wash buffers
False positives in ChIP-seqNon-specific DNA binding- Include appropriate negative controls (IgG, input DNA)
- Filter binding peaks present in control samples
- Only consider peaks identified in at least two biological replicates
Inconsistent immunoprecipitationAntibody batch variation- Validate each antibody batch before use
- Consider using epitope-tagged proteins for consistent results
- Use monoclonal antibodies when available

Additionally, pre-adsorption of antibodies with plant tissue extracts from grf3 knockout mutants can significantly reduce non-specific binding by removing antibodies that recognize unrelated proteins. For critical experiments, validating antibody specificity using multiple approaches (Western blot, immunoprecipitation followed by mass spectrometry, and immunostaining of knockout lines) is highly recommended.

How should contradictory results between different experimental approaches targeting GRF3 function be interpreted?

Contradictory results across different experimental approaches investigating GRF3 function require systematic analysis to resolve discrepancies. Follow this methodological framework:

  • Evaluate Technical Reliability:

    • Assess the sensitivity and specificity of each technique

    • Example: ChIP-seq may identify binding sites that are not functionally relevant in vivo, while RNA-seq captures both direct and indirect effects

  • Consider Context Dependency:

    • Analyze experimental conditions (developmental stage, tissue type, environmental conditions)

    • GRF3 functions can vary dramatically between different contexts:

      Experimental ContextObserved GRF3 FunctionPotential Explanation
      Young developing tissueGrowth promotionHigh expression of co-activators
      Mature tissueLimited effectLower expression/activity
      Stress conditionsStress response activationPost-translational modifications
      Different cell typesVaried target specificityCell-type specific co-factors
  • Assess Redundancy and Compensation:

    • Determine if other GRF proteins compensate in your experimental system

    • Analyze expression of other GRFs in response to GRF3 manipulation

    • Consider using higher-order mutants to minimize compensation effects

  • Integrate Multiple Data Types:

    • Weigh evidence from complementary approaches:

      • Binding data (ChIP-seq)

      • Expression data (RNA-seq)

      • Genetic evidence (mutant phenotypes)

      • Protein interaction data (Co-IP, Y2H)

    • Build a consensus model that accommodates most observations

  • Design Resolving Experiments:

    • Develop experiments specifically targeting the contradiction

    • Example: If ChIP-seq shows binding but RNA-seq shows no expression change, use inducible systems to test direct regulation at various time points

When properly analyzed, contradictions often reveal nuanced aspects of GRF3 biology, such as context-dependent functions or regulatory mechanisms that were not initially apparent in any single experimental approach.

What bioinformatic pipelines are most appropriate for analyzing GRF3 ChIP-seq data and identifying binding motifs?

Effective analysis of GRF3 ChIP-seq data requires specialized bioinformatic pipelines tailored to transcription factor binding site identification. The following comprehensive workflow is recommended:

  • Quality Control and Preprocessing:

    • FastQC for sequence quality assessment

    • Trimmomatic or Cutadapt for adapter removal and quality trimming

    • BWA or Bowtie2 for alignment to reference genome

    • Picard for duplicate marking/removal

  • Peak Calling and Filtering:

    • IDR (Irreproducible Discovery Rate) analysis to identify reproducible peaks across replicates

    • Filter peaks present in at least two biological replicates to ensure reliability

  • Peak Annotation and Gene Association:

    • ChIPseeker or HOMER for annotating peaks relative to genomic features

    • Associate peaks with genes using proximity criteria (e.g., within 1 kb of TSS or TTS)

    • Example distribution from GRF3 studies:

      Peak LocationPercentage of Total GRF3 Peaks
      Promoter regions73%
      Gene body20%
      Intergenic regions7%
  • Motif Discovery and Analysis:

    • MEME-ChIP or RSAT peak motifs for de novo motif discovery

    • FIMO or HOMER for motif scanning across the genome

    • Previous studies identified six DNA-binding motifs for GRF3, including ACTCGAC and CTTCTTC shared with GRF1

  • Integrative Analysis:

    • deepTools for generating heatmaps and profile plots

    • Integrate with RNA-seq using tools like DiffBind or ChIPpeakAnno

    • Pathway enrichment analysis using DAVID, g:Profiler, or Metascape

  • Visualization and Data Sharing:

    • IGV or UCSC Genome Browser for visualizing binding profiles

    • Generate browser-compatible files (bigWig, bigBed)

    • Deposit data in public repositories (GEO, ArrayExpress)

This pipeline has been successfully applied to analyze GRF family transcription factors, revealing their genome-wide binding patterns and regulatory functions in plant growth and stress responses .

How are AI-based approaches being integrated into antibody research and GRF protein function prediction?

Artificial intelligence and machine learning approaches are revolutionizing antibody research and protein function prediction, with several applications relevant to GRF3 studies:

  • AI-Driven Antibody Design:

    • Deep learning models are now being used to design antigen-specific antibody sequences

    • These models can generate highly specific CDRH3 sequences from germline-based templates

    • For GRF3 research, this could enable the development of antibodies with exceptional specificity, reducing cross-reactivity with other GRF family members

  • Epitope Prediction and Optimization:

    • Machine learning algorithms can predict optimal epitopes for antibody development

    • For GRF3, these tools can identify unique surface-exposed regions that differentiate it from other GRF proteins

    • Example workflow:

      • Protein structure prediction using AlphaFold2

      • Surface accessibility calculation

      • Epitope uniqueness scoring

      • Immunogenicity prediction

  • Functional Annotation and Interaction Prediction:

    • AI systems can predict protein-protein interactions and functional relationships

    • For GRF3, this can help identify:

      • Potential co-factors and regulators

      • Target genes based on promoter sequence features

      • Functional relationships with other transcription factors

  • High-Throughput Data Analysis:

    • Machine learning facilitates analysis of antibody repertoire data and high-throughput screening results

    • These tools enable functional mapping based on enrichment ratio values

    • For GRF3 studies, this can help process large-scale protein-DNA interaction datasets

  • Structural Biology Integration:

    • AI-predicted protein structures can be integrated with experimental data

    • For GRF3, this allows visualization of:

      • DNA-binding domains interacting with target sequences

      • Protein-protein interaction interfaces

      • Conformational changes upon binding

The integration of these AI approaches with traditional experimental methods provides powerful new tools for understanding GRF3 function and developing highly specific antibodies for research applications .

What are the emerging techniques for studying post-translational modifications of GRF3 and their impact on function?

Post-translational modifications (PTMs) of transcription factors like GRF3 can dramatically alter their function, localization, and interactions. These emerging techniques provide comprehensive insights into GRF3 PTMs:

  • Mass Spectrometry-Based Approaches:

    • Targeted MS: Using SRM (Selected Reaction Monitoring) or PRM (Parallel Reaction Monitoring) to quantify specific PTMs of GRF3

    • Middle-down proteomics: Analyzing larger protein fragments to preserve combinatorial PTM information

    • Crosslinking MS: Identifying PTM-dependent protein interactions

    • Workflow example:

      • Immunoprecipitate GRF3 from different conditions

      • Perform on-bead digestion with specific proteases

      • Analyze by LC-MS/MS with enrichment for modified peptides

      • Quantify PTM changes across conditions

  • Proximity Labeling Techniques:

    • BioID or TurboID: Fusing GRF3 with biotin ligase to identify condition-specific interactors

    • APEX2: Providing higher temporal resolution for dynamic interaction changes

    • This reveals how PTMs alter the GRF3 interactome under different conditions

  • Live-Cell Imaging of PTM Dynamics:

    • FRET-based sensors: Monitoring real-time phosphorylation or other PTMs

    • Split-fluorescent protein systems: Visualizing PTM-dependent interactions

    • These techniques capture the temporal dynamics of GRF3 modifications in response to stimuli

  • PTM-Specific Antibodies Combined with ChIP-seq:

    • Generating antibodies against specific modified forms of GRF3

    • Performing ChIP-seq with these antibodies to map how PTMs affect genome-wide binding

    • Example approach:

      • Develop antibodies against phosphorylated GRF3

      • Perform ChIP-seq under normal and stress conditions

      • Compare binding profiles to identify PTM-dependent regulatory targets

  • CRISPR-Based Approaches:

    • Base editing: Precisely mutating PTM sites without disrupting the protein

    • Prime editing: Introducing specific amino acid changes at PTM sites

    • These techniques allow functional testing of individual PTM sites in vivo

These emerging technologies provide unprecedented insights into how post-translational modifications dynamically regulate GRF3 function in response to developmental cues and environmental stresses.

How can GRF3 antibody research contribute to broader understanding of plant stress responses and crop improvement?

GRF3 antibody research offers significant contributions to understanding plant stress responses and developing improved crop varieties. By enabling precise investigation of GRF3 function, these tools help elucidate key regulatory mechanisms that can be harnessed for agricultural applications.

GRF3 and related transcription factors play crucial roles in integrating stress and defense signaling into developmental programs . Research has demonstrated that GRF3 influences thousands of genes involved in growth, defense, and stress response pathways. By developing and applying specific antibodies for GRF3, researchers can map the regulatory networks controlled by this transcription factor under various stress conditions.

This knowledge directly informs crop improvement strategies:

  • Identification of Key Regulatory Hubs: GRF3 antibodies enable the discovery of master regulatory points that coordinate growth and stress responses, providing targets for precision breeding.

  • Functional Characterization of Stress-Responsive Elements: ChIP-seq with GRF3 antibodies reveals the direct binding sites and regulatory mechanisms that mediate stress adaptation.

  • Translational Research Applications: Understanding GRF3 function in model plants can be extended to crop species, where orthologous proteins likely serve similar functions in stress resilience.

  • Biomarker Development: GRF3 activity and modification states could serve as molecular biomarkers for plant stress states, enabling early detection and intervention.

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.