DRP2A Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Specificity

DRP2A antibody detects the Arabidopsis dynamin-related protein DRP2A, which belongs to the evolutionarily conserved DRP2 family. These antibodies are typically polyclonal and raised against peptide sequences specific to DRP2A, though cross-reactivity with its paralog DRP2B is common due to their 76% amino acid sequence identity . DRP2A functions in clathrin-mediated endocytosis (CME), post-Golgi trafficking, and immune responses against pathogens like Pseudomonas syringae .

Antibody Development and Validation

DRP2A antibodies have been validated through:

  • Immunoblotting: Detecting DRP2A protein in wild-type plants and confirming its absence in drp2a mutants .

  • Co-immunoprecipitation (Co-IP): Identifying DRP2A interaction partners, including DRP2B and clathrin light chain 2 (CLC2) .

  • Mutant Analysis: Using T-DNA insertion lines (e.g., drp2a-1, drp2a-3) to verify antibody specificity (Table 1) .

Table 1: Key drp2a Mutant Lines Used in Antibody Validation

AlleleInsert PositionGenotyping PrimersSource
drp2a-12246SALK_071036 (LP: 1164)Smith et al.
drp2a-33979SALK_011319 (LP: 1171)Backues et al.
drp2a-41374SALK_018859 (LP: 858)Backues et al.

Research Applications

DRP2A antibody has been pivotal in elucidating:

Immune Signaling

  • DRP2A contributes to pattern-triggered immunity (PTI) by modulating late flg22 responses, including PATHOGENESIS-RELATED1 (PR1) gene induction and suppression of callose deposition .

  • drp2a mutants exhibit reduced resistance to P. syringae and altered flg22-induced reactive oxygen species (ROS) accumulation .

Endocytosis and Membrane Trafficking

  • DRP2A colocalizes with clathrin-coated vesicles (60% colocalization with CLC2) and transiently associates with the plasma membrane during vesicle scission .

  • Pharmacological inhibition (e.g., wortmannin, latrunculin B) increases DRP2A retention at the plasma membrane, highlighting its role in cytoskeleton-dependent trafficking .

Genetic Interactions with DRP2B

  • DRP2A and DRP2B function synergistically in plant growth and immune responses. Double mutants (drp1a drp2b) show seedling lethality, stunted roots, and hyperaccumulation of FLS2 receptor in the plasma membrane .

Table 2: Functional Roles of DRP2A in Immune Responses

ProcessDRP2A ContributionExperimental Evidence
Flg22-induced endocytosisRegulates FLS2 internalizationReduced FLS2 uptake in mutants
ROS signalingModulates NADPH oxidase activityEnhanced ROS in drp2a
SA-mediated defensePositively regulates PR1 expressionqRT-PCR data
Callose depositionSuppresses excessive cell wall thickeningMicroscopy quantification

Limitations and Considerations

  • Cross-Reactivity: DRP2A antibodies often detect DRP2B due to high sequence homology, necessitating mutant controls .

  • Functional Redundancy: DRP2A’s roles overlap with DRP2B, complicating phenotype interpretation in single mutants .

Future Directions

  • Develop isoform-specific antibodies to disentangle DRP2A/DRP2B functions.

  • Investigate DRP2A’s role in lipid microenvironment modulation at membrane domains .

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
DRP2A antibody; ADL6 antibody; At1g10290 antibody; F14N23.17Dynamin-2A antibody; EC 3.6.5.5 antibody; Dynamin-like protein 6 antibody; Dynamin-related protein 2A antibody
Target Names
DRP2A
Uniprot No.

Target Background

Function
DRP2A is a microtubule-associated force-producing protein involved in clathrin-mediated vesicle trafficking from the trans-Golgi network to the central vacuole. It is capable of binding and hydrolyzing GTP and exhibits specific binding affinity for phosphatidylinositol 3-phosphate (PtdIns3P).
Gene References Into Functions
  1. Studies have demonstrated that all tested variants of AVR3a effectively suppress defense responses triggered by flg22 and reduce the internalization of activated FLS2. Notably, AVR3a associates with Dynamin-Related Protein 2 (DRP2), a plant GTPase implicated in receptor-mediated endocytosis. [DRP2A] PMID: 26348328
  2. DRP2A and DRP2B function cooperatively in multiple pathways of post-Golgi trafficking, influenced by phosphatidylinositol 3- or 4-kinase activity and cytoskeleton polymerization. PMID: 25462567
  3. Research findings support a role for DRP2A/B in endocytosis, highlighting the essentiality of at least one of these closely related proteins for plant growth. PMID: 21461976
  4. Double mutants lacking both DRP2A and DRP2B exhibit an early developmental arrest prior to the first mitotic cell division during both male and female gametophytic development, resulting in gametophytic lethality. PMID: 20959563
  5. Based on experimental evidence, it is proposed that Seh1 plays a role in regulating the cycling of DRP2A between membrane-bound and soluble forms. PMID: 17085974

Show More

Hide All

Database Links

KEGG: ath:AT1G10290

STRING: 3702.AT1G10290.1

UniGene: At.11881

Protein Families
TRAFAC class dynamin-like GTPase superfamily, Dynamin/Fzo/YdjA family
Subcellular Location
Cytoplasm, cytosol. Golgi apparatus membrane; Peripheral membrane protein. Cytoplasm, cytoskeleton, phragmoplast. Cytoplasmic vesicle, clathrin-coated vesicle. Note=Localized in the forming cell plate during cytokinesis.

Q&A

What is DRP2A and what is its function in plants?

DRP2A is a member of the dynamin-related protein family in Arabidopsis thaliana, exhibiting significant homology with DRP2B. These proteins function in membrane trafficking processes critical for plant development and immune responses. DRP2A has approximately 29% amino acid identity with the GTPase domain of human dynamin, suggesting conserved mechanistic functions in membrane remodeling . The protein participates in vesicle formation and scission during endocytosis, particularly in the context of immune receptor trafficking. While DRP2A and DRP2B share high sequence similarity, they display some distinct and overlapping functions in plant cellular processes, with DRP2B showing more pronounced roles in immune responses compared to DRP2A .

How does DRP2A antibody cross-reactivity with DRP2B impact experimental interpretation?

The high amino acid sequence identity between DRP2A and DRP2B presents significant challenges for antibody specificity. Commercial polyclonal αDRP2 peptide antibodies typically detect both DRP2A and DRP2B proteins due to their sequence similarity . This cross-reactivity necessitates careful experimental design and interpretation of results. When analyzing immunoblot data, researchers should account for the combined signal from both proteins unless using highly specific monoclonal antibodies. The cross-reactivity is evident in immunoblot analyses where reduced levels of both proteins can be observed in either single mutant compared to wild-type plants . To distinguish between the two proteins, complementary approaches such as genetic knockouts, transcript analysis, or epitope-tagged versions of each protein should be employed alongside antibody-based detection.

What controls should be included when using DRP2A antibodies in immunoblotting?

When conducting immunoblot analyses with DRP2A antibodies, several critical controls must be incorporated. First, include both drp2a and drp2b single mutant lines alongside wild-type samples to assess antibody specificity and cross-reactivity profiles . This approach helps determine the contribution of each protein to the total signal. Second, utilize loading controls such as αCalnexin antibodies, which have been successfully employed in published studies . Third, include positive controls with recombinant DRP2A protein when available to confirm antibody sensitivity. For quantitative analyses, prepare a dilution series of total protein extracts to ensure signal linearity within the detection range. Finally, when possible, include double mutant samples (drp1a drp2b) as negative controls, though note that these plants may exhibit severe developmental defects that could complicate tissue collection and protein extraction .

How can researchers validate the specificity of commercial DRP2A antibodies?

To validate DRP2A antibody specificity, researchers should implement a multi-faceted approach. Begin with immunoblot analysis comparing wild-type plants with drp2a and drp2b single mutants to assess cross-reactivity patterns and relative signal reduction . Subsequently, perform immunoprecipitation followed by mass spectrometry to confirm the identity of pulled-down proteins. For immunolocalization studies, compare antibody labeling patterns with subcellular localization of fluorescently-tagged DRP2A in transgenic lines. When possible, conduct peptide competition assays using the immunizing peptide to demonstrate signal specificity. Additionally, validate antibody performance across different plant tissues and developmental stages, as expression patterns may vary. Finally, complement antibody-based approaches with transcript analysis using qRT-PCR to correlate protein and mRNA levels, although protein abundance may not always directly correspond to transcript levels .

How should researchers optimize immunoblotting protocols specifically for DRP2A detection?

Optimizing immunoblotting for DRP2A requires attention to several critical parameters. First, select an appropriate protein extraction buffer that preserves DRP2A integrity while efficiently solubilizing membrane-associated proteins - typically containing 1% Triton X-100 or NP-40 and protease inhibitor cocktails. Second, carefully determine optimal protein loading concentrations; published protocols have successfully detected DRP2 proteins in total protein extracts from un-elicited leaf tissue . Third, select the appropriate gel percentage (typically 8-10% SDS-PAGE) to efficiently resolve the approximately 100 kDa DRP2A protein. Fourth, optimize transfer conditions, often using wet transfer systems with extended transfer times (>1 hour) for large proteins. Fifth, implement a robust blocking protocol (5% non-fat dry milk or BSA) to minimize background signal. Finally, determine the optimal primary antibody dilution through systematic testing, with published studies successfully using αDRP2 antibody concentrations that detect both DRP2A and DRP2B . Document the complete set of optimized parameters for reproducibility across experiments.

What methodological approaches can differentiate between DRP2A and DRP2B functions in membrane trafficking?

Differentiating between DRP2A and DRP2B functions requires sophisticated methodological approaches. First, employ genetic analysis with single and double mutants to identify unique and overlapping phenotypes, noting that drp1a drp2b double mutants show synergistic developmental defects not observed in single mutants . Second, utilize complementation studies with protein-specific mutations in conserved domains to identify functional differences. Third, implement live-cell imaging with differentially tagged versions of each protein (e.g., DRP2A-GFP, DRP2B-RFP) to monitor spatiotemporal dynamics during membrane trafficking events. Fourth, perform immunoprecipitation studies to identify protein-specific interaction partners that might explain functional differences. Fifth, employ quantitative phenotypic assays measuring specific cellular processes like endocytosis rates of defined cargo proteins in single mutant backgrounds. Finally, use pharmacological inhibitors of specific trafficking pathways combined with single mutant analysis to determine differential roles in distinct membrane trafficking routes.

What sample preparation techniques improve DRP2A detection in different plant tissues?

Effective sample preparation is critical for consistent DRP2A detection across tissue types. For aerial tissues like leaves and cotyledons, snap-freezing in liquid nitrogen followed by grinding to a fine powder preserves protein integrity. For seedlings, where studies have successfully detected DRP2 proteins, gentle homogenization in extraction buffer containing protease inhibitors yields reliable results . When working with roots and other tissues with high proteolytic activity, incorporate additional protease inhibitors and maintain consistently cold temperatures during extraction. For membrane fraction enrichment, implement differential centrifugation protocols to separate plasma membrane fractions where DRP2A is known to localize during membrane trafficking events . This approach has successfully demonstrated FLS2 hyperaccumulation in the plasma membrane of drp1a drp2b double mutants . For immunohistochemistry, optimize fixation conditions (typically 4% paraformaldehyde) and epitope retrieval methods to maintain antibody accessibility while preserving tissue architecture.

How can researchers utilize DRP2A antibodies to study protein dynamics during pathogen challenge?

Studying DRP2A dynamics during pathogen challenge requires integration of multiple technical approaches. First, implement time-course immunoblot analysis following pathogen exposure, such as flg22 elicitation, to track changes in DRP2A protein abundance and post-translational modifications . Second, combine biochemical fractionation with immunoblotting to monitor DRP2A redistribution between subcellular compartments during immune responses. Third, utilize immunoprecipitation with DRP2A antibodies before and after pathogen treatment to identify changing protein interaction networks. Fourth, employ immunofluorescence microscopy to visualize altered DRP2A localization patterns during infection, particularly at sites of pathogen contact. Fifth, implement in situ proximity ligation assays to detect transient interactions between DRP2A and immune signaling components. Finally, consider phospho-specific antibody development if phosphorylation regulates DRP2A activity during immune responses, as suggested by the dynamic nature of membrane trafficking during pathogen challenge .

How should experiments be designed to investigate DRP2A's role in endocytosis of immune receptors?

Investigating DRP2A's role in immune receptor endocytosis requires systematic experimental design. Begin by establishing transgenic lines expressing fluorescently-tagged immune receptors (such as FLS2-GFP) in wild-type, drp2a, and drp2b genetic backgrounds to facilitate comparative analysis . Implement confocal microscopy-based endocytosis assays with appropriate ligands (e.g., flg22 peptide for FLS2) and track receptor internalization over time courses (typically 0-120 minutes post-elicitation). Quantify endocytic vesicle formation using automated image analysis to ensure unbiased assessment. Complement imaging with biochemical fractionation and subsequent immunoblotting with DRP2A antibodies to correlate changes in protein localization with receptor endocytosis kinetics . Include appropriate controls such as inactive ligand variants (flg22*) and endocytosis inhibitors to validate experimental systems . Finally, assess functional outcomes by measuring downstream immune responses such as ROS production in these genetic backgrounds, noting that drp2b mutants show increased ROS production in response to flg22 despite decreased receptor endocytosis .

What approaches can resolve contradictory findings regarding DRP2A's impact on different immune signaling pathways?

Resolving contradictions in DRP2A's impact on immune signaling requires systematic investigation across multiple signaling branches. First, implement parallel analysis of all major immune signaling outputs (ROS production, MAPK activation, callose deposition, and transcriptional responses) within the same experimental system and developmental stage . Second, conduct time-course experiments with fine temporal resolution to detect pathway-specific kinetic differences, as different signaling branches may be affected at distinct time points. Third, utilize tissue-specific expression systems to determine if contradictory findings result from cell-type-specific functions of DRP2A. Fourth, examine the impact of different PAMP concentrations, as signaling thresholds may differ between pathways. Fifth, directly compare drp2a and drp2b single mutants alongside double mutants to identify protein-specific contributions to each signaling branch . Finally, implement phosphoproteomic analysis to identify pathway-specific post-translational modifications that might explain differential effects of DRP2A on distinct signaling components.

How can researchers assess DRP2A-DRP1A functional interactions in membrane trafficking during immune responses?

Investigating DRP2A-DRP1A functional interactions requires sophisticated experimental approaches. First, generate and characterize drp2a drp1a double mutants alongside single mutants, although severe developmental phenotypes may complicate analysis as observed with drp1a drp2b double mutants . Second, implement co-immunoprecipitation experiments using DRP2A antibodies to confirm physical interactions with DRP1A and identify additional complex components. Third, employ bimolecular fluorescence complementation (BiFC) or fluorescence resonance energy transfer (FRET) to visualize protein interactions in vivo during immune responses. Fourth, conduct live-cell imaging with differentially tagged proteins to track co-localization dynamics following pathogen elicitation. Fifth, perform domain swapping experiments to identify interaction interfaces critical for function. Sixth, use biochemical approaches to determine if interactions affect biochemical properties like GTPase activity. Finally, correlate interaction patterns with functional readouts such as receptor endocytosis efficiency and downstream immune responses in genetic backgrounds with different combinations of wild-type and mutant proteins .

What experimental setup best captures the dynamic relationship between DRP2A and plant plasma membrane composition?

Investigating DRP2A's relationship with plasma membrane composition requires specialized experimental setups. First, implement lipidomic analysis comparing plasma membrane fractions from wild-type and drp2a mutant plants, with particular attention to sterol-rich domains that may facilitate protein recruitment . Second, utilize super-resolution microscopy techniques (STORM/PALM) with appropriate membrane dyes and tagged DRP2A to visualize nanoscale association with specific membrane domains. Third, apply fluorescence recovery after photobleaching (FRAP) to measure DRP2A mobility within different membrane domains before and after pathogen elicitation. Fourth, employ biochemical fractionation methods specifically optimized to isolate detergent-resistant membrane fractions, where DRP2A may preferentially associate. Fifth, investigate changes in lipid composition following controlled expression of DRP2A to establish causality. Finally, correlate changes in membrane composition with functional outcomes such as immune receptor distribution and signaling efficiency, particularly focusing on whether DRP2A influences the lipid environment in defined plasma membrane domains that may support immune receptor function .

How can researchers address inconsistent DRP2A antibody labeling between experiments?

Addressing inconsistent DRP2A antibody labeling requires systematic troubleshooting. First, implement standardized protein extraction protocols with precise buffer compositions and handling procedures to minimize variation in sample preparation. Second, establish consistent protein quantification methods and loading controls across experiments, with calnexin serving as a reliable loading control in published studies . Third, prepare larger antibody stock aliquots to reduce freeze-thaw cycles that may affect antibody performance. Fourth, optimize blocking conditions to minimize background signals that might mask specific labeling. Fifth, consider sample-specific factors such as tissue type and developmental stage that might affect DRP2A expression levels and antibody accessibility. Sixth, maintain detailed records of antibody lot numbers, as lot-to-lot variation can significantly impact performance. Finally, implement positive controls with recombinant DRP2A protein or samples with known high expression to validate each experiment. If inconsistencies persist, consider transitioning to epitope-tagged transgenic lines that allow detection with highly standardized commercial tag antibodies.

What explains contradictory findings between transcript and protein levels when studying DRP2A?

Discrepancies between DRP2A transcript and protein levels can arise from multiple factors. First, post-transcriptional regulatory mechanisms may modulate mRNA stability or translation efficiency, as seen in studies where unchanged FLS2 transcript levels did not correlate with protein abundance changes . Second, protein stability and turnover rates may vary under different experimental conditions, affecting steady-state protein levels independently of transcription. Third, technical limitations in detection sensitivity between qRT-PCR and immunoblotting may create apparent discrepancies when one technique operates near its detection limit. Fourth, spatial or temporal variation in gene expression might not be captured by whole-tissue analyses, masking cell-specific correlation between transcript and protein. Fifth, experimental timing is critical, as transcriptional changes typically precede protein-level changes, requiring time-course experiments for proper correlation. To address these complexities, researchers should implement parallel transcript and protein analyses within the same biological samples, utilize cell-type-specific approaches where possible, and consider mathematical modeling to account for differences in synthesis and degradation rates when interpreting results.

How can researchers quantitatively analyze DRP2A protein levels in immune response time-course studies?

Quantitative analysis of DRP2A in immune response time-courses requires rigorous methodological approaches. First, implement standardized protein extraction and immunoblotting protocols with appropriate normalization controls. Second, utilize digital image acquisition with care to avoid signal saturation, ensuring all samples fall within the linear detection range. Third, apply appropriate normalization using loading controls like calnexin that remain stable during immune responses . Fourth, employ densitometry software with consistent analysis parameters across all experimental replicates. Fifth, represent data using fold-change relative to time zero rather than absolute values to facilitate cross-experimental comparison. Sixth, apply appropriate statistical tests for time-course data, typically repeated measures ANOVA with post-hoc tests. Finally, correlate protein level changes with functional readouts such as ROS production or callose deposition to establish biological significance . For more sophisticated analysis, consider implementing computational modeling to describe the dynamic relationship between DRP2A levels and downstream immune responses across the time-course.

What statistical approaches are most appropriate for analyzing DRP2A function in disease resistance assays?

When analyzing DRP2A function in disease resistance, appropriate statistical approaches are essential. For bacterial growth assays comparing wild-type and drp2a mutants, implement two-way ANOVA to account for both genotype and bacterial strain effects, followed by Tukey's post-hoc test for multiple comparisons . Calculate bacterial titers using log-transformed colony-forming unit (CFU) values to meet normality assumptions required for parametric tests. For disease symptom scoring, utilize non-parametric tests such as Mann-Whitney U or Kruskal-Wallis when data does not follow normal distribution. In time-course experiments measuring responses like ROS production, apply repeated measures ANOVA or mixed-effects models to account for the non-independence of measurements . For correlation analyses between DRP2A levels and disease outcomes, use Pearson's or Spearman's correlation coefficients depending on data distribution. Finally, implement power analysis prior to experimental design to ensure sufficient biological replicates for detecting biologically meaningful differences, particularly important when working with subtle phenotypes that might emerge from functional redundancy between DRP2A and related proteins .

What is the relationship between DRP2A function and different branches of plant immune signaling?

The relationship between DRP2A and immune signaling reveals intriguing pathway-specific effects. Evidence indicates DRP2A and related proteins have differential impacts on distinct branches of PAMP-triggered immunity . For instance, drp2b mutants show enhanced ROS production but reduced PR1 expression after flg22 elicitation, while MAPK-dependent gene expression remains unaffected . This pattern suggests dynamin-related proteins selectively modulate specific signaling branches rather than affecting all immune outputs uniformly. The mechanisms underlying this specificity remain incompletely understood but may involve the spatial organization of signaling components into distinct membrane domains, differential regulation of receptor-complex composition, or pathway-specific endocytic trafficking routes. Additionally, the observation that RbohD-dependent ROS production is elevated in drp2b mutants suggests a specific regulatory connection between dynamin function and NADPH oxidase activity at the plasma membrane . Future research should focus on how DRP2A's membrane remodeling activities might differently impact the spatial segregation or activation thresholds of various immune signaling branches, potentially through biochemical interaction studies with branch-specific components coupled with high-resolution imaging of their distribution.

How do DRP2A and DRP2B cooperate with DRP1A in membrane trafficking during immunity?

The cooperation between DRP2A, DRP2B, and DRP1A reveals a complex functional relationship in membrane trafficking during immunity. Genetic analysis has established that DRP1A and DRP2B function synergistically, as drp1a drp2b double mutants display severe developmental defects including stunted roots, defective cell shape, and cytokinesis abnormalities leading to seedling lethality . In the context of immunity, both DRP1A and DRP2B positively contribute to bacterial resistance, with both single mutants showing increased susceptibility to Pseudomonas syringae . Mechanistically, these proteins collaborate in flg22-induced endocytosis of the FLS2 receptor, with drp1a drp2b double mutants exhibiting complete blockage of ligand-induced FLS2 internalization . The functional relationship may stem from their biochemical properties - DRP1A's GTPase domain shares higher amino acid identity with human dynamin (62%) compared to DRP2B (29%), suggesting DRP1A may contribute more substantially to the GTPase activity required for endocytic vesicle scission . Additionally, DRP1A's role in maintaining high lipid-order sterol-rich domains may complement DRP2B's functions in a manner essential for proper immune receptor trafficking and signaling . Future research should investigate the stoichiometry and assembly dynamics of these proteins during defense responses.

What emerging technologies will advance understanding of DRP2A's role in plant immunity?

Emerging technologies will significantly advance our understanding of DRP2A in plant immunity. First, CRISPR-Cas9 gene editing enables precise modification of endogenous DRP2A to create domain-specific mutations or epitope-tagged versions that maintain native expression patterns. Second, advanced super-resolution microscopy techniques such as STORM/PALM now allow visualization of protein dynamics at nanometer resolution, ideal for tracking DRP2A during vesicle formation events. Third, proximity-dependent labeling methods like BioID or TurboID can identify transient interaction partners of DRP2A during immune responses. Fourth, single-cell transcriptomics and proteomics will reveal cell-type-specific functions of DRP2A that may be masked in whole-tissue analyses. Fifth, cryo-electron microscopy advancement may soon enable structural determination of plant dynamin-related protein complexes, providing mechanistic insights into their membrane remodeling activities. Sixth, optogenetic approaches can temporally control DRP2A activity to dissect its acute functions during immune responses. Finally, advanced computational modeling incorporating membrane physics with protein dynamics will help predict how DRP2A influences receptor distribution and signaling in complex membrane environments. These technologies, particularly when used in combination, promise to resolve the molecular mechanisms by which DRP2A orchestrates membrane dynamics during plant immune responses.

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.