GRF8 Antibody

Shipped with Ice Packs
In Stock

Description

Target Protein Characteristics

GRF8 (At5g65430.1) belongs to the 14-3-3 protein family in Arabidopsis thaliana, which regulates diverse cellular processes including metabolism, stress responses, and signal transduction . Key features:

  • Molecular function: Binds phosphorylated client proteins to modulate their activity or localization.

  • Structural domains: Contains conserved 14-3-3 motifs but lacks additional functional domains present in other GRF isoforms .

  • Expression: Detected in various plant tissues, with roles in growth and development.

Key experimental data:

  • Western blot: Detects recombinant GRF8 (~30 kDa) in Arabidopsis extracts, though with lower sensitivity compared to other GRF isoforms .

  • Cross-reactivity: Confirmed in lily (Lilium longiflorum) and moss (Physcomitrella patens), demonstrating evolutionary conservation of epitopes.

  • Limitations: Reduced efficacy in tissue-specific localization studies due to low endogenous GRF8 expression levels .

A. Signal Transduction Studies

Used to identify GRF8 interactions with:

  • Calcium-dependent protein kinases

  • Transcription factors regulating stress responses

B. Developmental Biology

  • Maps GRF8 expression patterns in floral tissues and root apical meristems .

  • Links GRF8 to brassinosteroid signaling pathways affecting cell elongation .

Challenges in Antibody Validation

As highlighted in broader antibody research :

  • Specificity issues: Polyclonal antibodies may cross-react with non-target GRF isoforms.

  • Validation metrics: Require knockout controls and orthogonal assays (e.g., mass spectrometry) for confirmation.

  • Commercial limitations: Many vendors, including Agrisera, provide limited characterization data compared to standards proposed by initiatives like YCharOS .

Comparative Analysis of GRF Isoforms

IsoformDomainsExpression PatternAntibody Reactivity
GRF1Full 14-3-3UbiquitousStrong
GRF8Truncated C-termRoot/floral tissuesModerate

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
GRF8 antibody; At5g65430 antibody; MNA5.1614-3-3-like protein GF14 kappa antibody; General regulatory factor 8 antibody
Target Names
GRF8
Uniprot No.

Target Background

Function
GRF8 Antibody is associated with a DNA binding complex that binds to the G box, a well-characterized cis-acting DNA regulatory element found in plant genes. This antibody plays a role in the regulation of nutrient metabolism. It acts as a negative regulator of freezing tolerance by modulating the stability of cold-responsive C-repeat-binding factors (CBF) DREB1A and DREB1B proteins through ubiquitin-mediated degradation.
Database Links

KEGG: ath:AT5G65430

STRING: 3702.AT5G65430.3

UniGene: At.10382

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

Q&A

What is GRF8 and why is it significant in plant research? [Basic]

GRF8 (General Regulatory Factor 8) is a member of the 14-3-3 protein family found in plants. It is also known as 14-3-3 protein G-box factor 14 kappa or GF14 kappa. GRF8 plays a significant role in plant cellular signaling by interacting with the BZR1 transcription factor involved in brassinosteroid signaling pathways. This interaction potentially affects the nucleocytoplasmic shuttling of BZR1, influencing downstream gene expression and physiological responses . Understanding GRF8 function contributes to our knowledge of plant hormone signaling, stress responses, and developmental regulation. When studying GRF8, researchers should consider its role within the broader 14-3-3 protein family network and its specific contributions to brassinosteroid-mediated processes.

How do GRF8 antibodies differ from other 14-3-3 protein antibodies? [Basic]

GRF8 antibodies are designed to target specific epitopes of the GRF8/14-3-3 kappa protein that differentiate it from other 14-3-3 protein family members. While 14-3-3 proteins share considerable sequence homology, GRF8 antibodies typically recognize unique regions that enable specific detection. For example, some commercially available GRF8 antibodies use synthetic peptides that share varying degrees of homology with other GRF proteins - the PHY2480S antibody's immunization peptide shares 81% (13/16) homology with the sequence in GRF6 (AT5G10450) . To ensure specificity in your experiments, validate the antibody's cross-reactivity profile before use, particularly if your research examines multiple 14-3-3 family members concurrently. When selecting an antibody, researchers should consult the manufacturer's specificity data and consider using multiple antibodies that target different epitopes to confirm findings.

What are the best storage conditions for maintaining GRF8 antibody efficacy? [Basic]

Proper storage of GRF8 antibodies is crucial for maintaining their specificity and sensitivity over time. GRF8 antibodies are typically provided in lyophilized form and require specific handling protocols. For optimal preservation, use a manual defrost freezer and avoid repeated freeze-thaw cycles that can damage antibody structure . Upon receipt of a lyophilized antibody, store it immediately at the recommended temperature (typically -20°C for long-term storage). When preparing working aliquots, reconstitute only the amount needed for immediate experiments to avoid degradation. Record reconstitution date and track the number of freeze-thaw cycles for each aliquot. For shipping or short transfers, the antibody can be maintained at 4°C, but prompt storage at recommended temperatures upon arrival is essential . Implementing these practices will help ensure consistent antibody performance across experiments and extend the usable lifetime of your GRF8 antibody.

How should researchers design validation experiments for GRF8 antibodies? [Advanced]

Validation of GRF8 antibodies requires a comprehensive approach incorporating multiple experimental techniques. Begin with Western blot analysis using positive controls (tissues known to express GRF8) and negative controls (GRF8 knockout/knockdown tissues if available). Include a molecular weight marker to confirm that the detected band corresponds to the expected size of GRF8 (approximately 30 kDa). Perform peptide competition assays similar to those described for other antibodies, where pre-incubation of the antibody with the immunizing peptide should abolish specific signals . For additional validation, use immunoprecipitation followed by mass spectrometry to confirm that the antibody captures the intended target. Cross-validate findings using multiple detection methods such as ELISA, immunofluorescence, and immunohistochemistry. Design experiments to test cross-reactivity with other 14-3-3 family members, especially those with high sequence homology to GRF8. Document antibody performance across different experimental conditions, including varying fixation methods, incubation times, and antibody concentrations. This systematic validation will establish the reliability and specificity boundaries of your GRF8 antibody.

What are the optimal conditions for Western blot analysis using GRF8 antibodies? [Advanced]

For optimal Western blot analysis with GRF8 antibodies, consider both reducing and non-reducing conditions, as protein conformation can significantly impact epitope accessibility. Based on experience with other plant proteins, start with standard SDS-PAGE using 10-12% gels for good resolution of GRF8's approximately 30 kDa size. For protein extraction, use a buffer containing 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1% Triton X-100, protease inhibitor cocktail, and phosphatase inhibitors if phosphorylation states are relevant. Transfer proteins to PVDF membranes, which typically provide better results than nitrocellulose for plant proteins. Block with 5% non-fat dry milk in TBST for 1 hour at room temperature. For primary antibody incubation, dilute GRF8 antibody according to manufacturer recommendations (typically 1:1000 to 1:5000) in blocking solution and incubate overnight at 4°C. After washing with TBST, apply an appropriate HRP-conjugated secondary antibody (1:5000-1:10000) for 1 hour at room temperature. When analyzing results, be aware that post-translational modifications might cause GRF8 to migrate differently than predicted. If signal specificity is a concern, perform parallel experiments under non-reducing conditions as this approach has revealed hidden epitopes in other protein studies .

How can researchers determine the affinity constant of their GRF8 antibody? [Advanced]

Determining the affinity constant (Kd) of a GRF8 antibody provides crucial information about its binding characteristics and suitability for specific applications. Adapt the method described by Friguet et al. (1984) which has been successfully applied to other antibodies . Begin with a preliminary ELISA to estimate the approximate dissociation constant range. Prepare serial dilutions of the GRF8 antigen (recombinant protein or synthetic peptide) and incubate with a fixed, low concentration of antibody that should be at or below the estimated Kd value. Allow the antibody-antigen mixture to reach equilibrium (typically 2-3 hours at room temperature or overnight at 4°C). Transfer the mixtures to ELISA plates pre-coated with the same antigen and incubate briefly (30 minutes) to capture unbound antibody. After washing, detect bound antibody using an appropriate enzyme-conjugated secondary antibody and suitable substrate. Plot the reciprocal of bound antibody fraction versus antigen concentration to determine the Kd value. For more precise measurements, consider alternative techniques such as surface plasmon resonance (SPR) or bio-layer interferometry (BLI), which provide real-time binding kinetics including both association (kon) and dissociation (koff) rate constants. Understanding your GRF8 antibody's affinity will help optimize experimental conditions and interpret binding data more accurately.

How should researchers evaluate cross-reactivity of GRF8 antibodies with other GRF family members? [Advanced]

Evaluating cross-reactivity of GRF8 antibodies requires a systematic approach due to the high sequence homology among GRF family members. Begin by conducting in silico analysis of the immunizing peptide sequence against all GRF family proteins to identify potential cross-reactive family members. Based on the specificity data from commercial antibodies, particular attention should be paid to GRF1, GRF2, GRF4, GRF5, GRF6, and GRF7, which can share up to 100% homology with immunization peptides used for some GRF8 antibodies . Design experimental validation using recombinant proteins or synthetic peptides of different GRF members in comparative ELISA, Western blot, and dot blot analyses. If available, utilize tissues from knockout/knockdown plants for specific GRF members as critical negative controls. Implement peptide competition assays where pre-incubation of the antibody with peptides from different GRF family members can reveal cross-reactivity patterns. Consider using heterologous expression systems to express individual GRF proteins and test antibody reactivity against each. Quantify relative binding affinities to different GRF proteins using techniques like SPR or BLI. Document cross-reactivity in a detailed table showing percent reactivity compared to GRF8, which will serve as an essential reference for interpreting experimental results and designing appropriate controls.

What is the species cross-reactivity profile of common GRF8 antibodies and how does this impact experimental design? [Advanced]

Commercial GRF8 antibodies exhibit distinct species cross-reactivity profiles that significantly influence experimental design decisions. Based on available data, most GRF8 antibodies recognize the protein across multiple plant species, with antibody PHY2481A showing reactivity in 17 different plant species including Arabidopsis thaliana, Oryza sativa, Zea mays, and Glycine max . When planning cross-species studies, researchers should:

  • Create a species validation plan based on sequence conservation analysis of the GRF8 protein region containing the epitope

  • Perform preliminary Western blots with positive control samples from each species of interest

  • Adjust antibody concentration and incubation conditions for each species based on signal strength and specificity

  • Include appropriate loading controls specific to each species

  • Consider species-specific sample preparation modifications

The table below summarizes known cross-reactivity profiles of different GRF8 antibodies:

Antibody IDNumber of Cross-Reactive SpeciesKey SpeciesHomology with Other GRF Proteins
PHY2480S4Arabidopsis thaliana, Brassica rapa, Brassica napus, Vitis vinifera81% with GRF6
PHY2481A17Arabidopsis thaliana, Spinacia oleracea, Oryza sativa, Zea mays, etc.Not specified
PHY2494A17Arabidopsis thaliana, Zea mays, Vitis vinifera, etc.89-100% with multiple GRFs

When interpreting results across species, account for potential differences in epitope accessibility due to species-specific post-translational modifications or protein interactions. This comprehensive approach ensures reliable cross-species comparisons while minimizing false positives or negatives due to antibody specificity limitations.

How can researchers distinguish between specific and non-specific signals when using GRF8 antibodies? [Advanced]

Distinguishing between specific and non-specific signals requires implementing multiple validation strategies and controls. To establish signal specificity, always include a GRF8 knockout/knockdown sample when possible, which should show significant reduction or absence of the target band/signal. Perform peptide competition assays where pre-incubation of the antibody with the immunizing peptide should eliminate specific signals while non-specific signals will remain . Include gradient loading of protein samples to confirm that signal intensity correlates with protein amount. When detecting GRF8 by Western blot, ensure the observed band appears at the expected molecular weight (approximately 30 kDa). For immunostaining applications, compare staining patterns with published GRF8 localization data and use secondary-only controls to identify background staining. Cross-validate results using antibodies targeting different epitopes of GRF8 or alternative detection methods like mass spectrometry. Be particularly vigilant about signals appearing in unexpected molecular weight ranges, which could indicate degradation products, splice variants, or cross-reactive proteins. When troubleshooting persistent non-specific signals, systematically optimize blocking conditions, antibody concentrations, and washing stringency. Document these optimization steps and validation controls in publications to demonstrate the reliability of your findings.

What are common sources of experimental variability when working with GRF8 antibodies and how can they be controlled? [Advanced]

Experimental variability with GRF8 antibodies can arise from multiple sources that must be systematically controlled. Sample preparation represents a primary source of variability, particularly in plant tissues where variations in growth conditions, developmental stage, and harvest time can significantly alter GRF8 expression and modification states. To minimize this variability, standardize growth conditions, tissue collection protocols, and sample processing times. Another major source of variability is antibody quality across different lots and storage conditions. Implement lot testing before beginning new studies and maintain consistent storage protocols including avoiding freeze-thaw cycles . Detection method inconsistencies can be addressed by standardizing exposure times for chemiluminescence detection or laser settings for fluorescence-based methods. For quantitative analyses, include calibration standards on each gel/membrane and normalize target signals to multiple housekeeping proteins. Environmental factors during experimental procedures, including temperature fluctuations during incubations, can introduce variability. Use temperature-controlled equipment and standardize incubation conditions across experiments. When comparing data across multiple experiments, include internal reference samples processed in each experimental batch to enable accurate normalization. Document all experimental conditions, antibody details (lot number, concentration), and image acquisition settings to facilitate troubleshooting of unexpected variability and ensure reproducibility across different researchers and laboratories.

How should contradictory results from different GRF8 antibodies be reconciled and interpreted? [Advanced]

  • Target epitopes and their conservation across species and GRF family members

  • Antibody format (polyclonal vs. monoclonal, recombinant constructs)

  • Validation methods used by manufacturers and your lab

  • Known cross-reactivities with other proteins

  • Affinity constants and optimal working concentrations

Next, implement a multi-faceted validation approach using orthogonal methods. Verify protein expression using RNA-based techniques like RT-qPCR or RNA-seq. Employ mass spectrometry to confirm protein identity in immunoprecipitation samples. Consider using genetic approaches such as knockout/knockdown models or overexpression systems to manipulate GRF8 levels and observe how each antibody responds to these manipulations. For localization studies showing discrepancies, use GFP-tagged GRF8 constructs to provide independent confirmation. Evaluate whether contradictions might reflect biological realities rather than technical artifacts, such as detection of different post-translational modifications, splice variants, or conformational states. Consult with antibody manufacturers about known limitations and compare your findings with published literature. When publishing results with contradictory antibody data, transparently report all findings along with your interpretation of the discrepancies, allowing readers to evaluate the strength of evidence.

How can GRF8 antibodies be utilized in high-throughput screening approaches? [Advanced]

GRF8 antibodies can be effectively integrated into high-throughput screening (HTS) workflows through several innovative adaptations. For large-scale screening of plant samples, develop a standardized antibody-based ELISA protocol optimized for 384-well plate format with automated liquid handling systems. Adapt the Golden Gate-based dual-expression vector system described for other antibodies to create a screening platform for GRF8 interactions with potential binding partners. This approach allows for rapid expression of membrane-bound GRF8 along with fluorescent tags for flow cytometry-based sorting and selection. When screening plant mutant collections or treatment conditions, implement automated tissue harvesting, protein extraction, and Western blot analysis using robotic systems and standardized protocols. For spatial proteomics applications, develop multiplexed immunofluorescence protocols that combine GRF8 antibodies with antibodies against other signaling components to simultaneously visualize multiple proteins across tissue sections. Incorporate barcoding strategies to enable batch processing of samples while maintaining sample identity. When analyzing large datasets generated through HTS approaches, implement machine learning algorithms to identify patterns in GRF8 expression or localization across different experimental conditions. Consider adapting next-generation sequencing (NGS) technologies similar to those used in antibody development to create a system for monitoring GRF8-associated complexes across multiple conditions simultaneously. Document standardized protocols that ensure reproducibility across different laboratories and create comprehensive data analysis pipelines that facilitate interpretation of complex, multi-parametric datasets.

What novel methodologies are being developed for studying GRF8 protein interactions using antibody-based approaches? [Advanced]

Novel methodologies for studying GRF8 protein interactions are leveraging advanced antibody-based techniques to provide unprecedented insights into protein complexes and signaling networks. Proximity ligation assays (PLA) can be adapted using GRF8 antibodies paired with antibodies against suspected interaction partners like BZR1 to visualize and quantify endogenous protein interactions with subcellular resolution. Single-molecule pull-down (SiMPull) techniques combine the specificity of antibody-based immunoprecipitation with single-molecule fluorescence imaging to analyze GRF8 complexes with precise stoichiometry information. Researchers can apply the Golden Gate cloning strategy described for recombinant antibody development to create libraries of potential GRF8 interaction partners for high-throughput screening. Adapting in vivo membrane-bound expression systems to plant cells will enable real-time monitoring of GRF8 interactions in physiologically relevant contexts. For temporal dynamics, implement optogenetic approaches combined with antibody-based detection to trigger and monitor GRF8 interactions with controlled timing. Develop bifunctional antibody constructs that simultaneously bind GRF8 and introduce proximity-dependent modifications like biotinylation to nearby proteins, enabling comprehensive interactome mapping. Cross-linking mass spectrometry (XL-MS) workflows utilizing GRF8 antibodies for initial enrichment can provide detailed structural information about interaction interfaces. Finally, single-cell approaches combining microfluidics with antibody-based detection systems will reveal cell-to-cell variation in GRF8 interaction networks across plant tissues. These cutting-edge methodologies promise to transform our understanding of GRF8's role in complex signaling networks.

How can GRF8 antibodies be modified for enhanced specificity in challenging experimental contexts? [Advanced]

Enhancing GRF8 antibody specificity for challenging experimental contexts requires sophisticated antibody engineering approaches. Implement epitope fine-tuning by designing synthetic peptides that specifically target regions of GRF8 with minimal homology to other 14-3-3 proteins, focusing particularly on regions that differ from GRF1, GRF2, GRF4, GRF5, GRF6, and GRF7 where 100% homology has been observed with some immunization peptides . Apply site-directed mutagenesis to modify key amino acids in the antibody's complementarity-determining regions (CDRs), similar to the approach used for other antibodies where tyrosine residues in CDRH2 and CDRH3 domains were mutated to phenylalanine to enhance specificity . Develop recombinant antibody formats with dual specificity that recognize two distinct epitopes on GRF8 simultaneously, dramatically reducing cross-reactivity. For membrane protein studies, adapt the membrane-bound antibody expression system to create GRF8-specific reagents that maintain specificity in native membrane environments. Consider creating context-dependent antibodies that recognize GRF8 only when in complex with specific interaction partners, using approaches like bispecific antibodies or conformation-sensitive selection strategies. For improved specificity in fixed tissues, develop optimized antigen retrieval protocols specific to GRF8 epitopes that enhance accessibility while minimizing exposure of cross-reactive epitopes. Implement negative selection strategies during antibody development to systematically eliminate clones showing cross-reactivity with other GRF family members. Document specificity enhancements through comprehensive cross-reactivity testing against all GRF family members and create detailed protocols for application-specific modifications.

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.