RND3 antibody targets the RND3 protein, a member of the Rho GTPase family involved in regulating actin dynamics, cell migration, and apoptosis . Unlike typical Rho GTPases, RND3 remains constitutively active (GTP-bound) and acts as an antagonist of RhoA/ROCK signaling . It is widely expressed in tissues, including the brain and epithelial cells, and plays roles in neuronal development, cancer progression, and inflammatory responses .
The RND3 antibody has been instrumental in:
Cancer Studies: Demonstrating RND3’s tumor-suppressive role in non-small cell lung cancer (NSCLC). Overexpression of RND3 in H520 and H358 cells reduced proliferation by downregulating Notch intracellular domain (NICD) and Rho kinase activity .
Neuroscience: Linking RND3 deficiency to upregulated NF-κB signaling in mouse brains, as shown by increased P65 protein levels .
Cell Cycle Regulation: Revealing RND3’s phosphorylation by TAOK kinases during mitosis, which promotes cytosolic relocation and mitotic cell rounding .
Storage: Stable at -20°C to -70°C; avoid freeze-thaw cycles .
Specificity: Recognizes both endogenous and exogenous RND3 in human and mouse samples .
Limitations: Does not distinguish between phosphorylated and non-phosphorylated forms unless combined with phospho-specific assays .
Research using the RND3 antibody has highlighted its potential as a biomarker or therapeutic target:
RND3 (also known as RhoE) is a small GTPase with a molecular mass of approximately 25-27.4 kDa that belongs to the Rho family of GTPases . Unlike typical GTPases, RND3 binds GTP but lacks intrinsic GTPase activity and is resistant to Rho-specific GTPase-activating proteins . This makes it functionally distinct from other Rho family members. Researchers should be aware that RND3 has diverse regulatory properties independent of the traditional RhoA/ROCK1 pathway, particularly in governing oxidative stress, inflammation, and lipid metabolism .
RND3 antibodies have been validated for multiple experimental applications with varying degrees of effectiveness. Western blot (WB) is the most commonly validated application, with antibodies typically detecting a band at approximately 27 kDa . Immunohistochemistry (IHC) is another common application, allowing visualization of RND3 in tissue sections . For protein interaction studies, RND3 antibodies are effectively used in co-immunoprecipitation (Co-IP) assays . Additionally, immunofluorescence techniques can be employed to examine cellular localization patterns and co-localization with interacting proteins like TRAF6 .
When selecting an RND3 antibody, researchers should consider several critical factors:
Species reactivity: Most commercial RND3 antibodies have reactivity against human and mouse samples, with some also reactive against rat samples . Verify the antibody's reactivity matches your experimental model.
Application validation: Ensure the antibody has been validated for your specific application (WB, IHC, Co-IP, etc.) .
Epitope recognition: Consider whether the antibody targets a specific domain or the full-length protein. For example, some antibodies target the RD1 peptide region of RND3 .
Conjugation requirements: Determine whether you need an unconjugated antibody or one conjugated to specific tags (biotin, FITC, HRP, etc.) depending on your detection method .
Validation evidence: Review published literature and manufacturer data showing the antibody's specificity and performance in your application of interest .
To investigate RND3's role in endothelial cell pyroptosis during atherosclerosis, researchers should implement a multi-faceted experimental approach:
Animal models: Utilize Apoe KO mice as an atherosclerosis model, comparing with endothelium-specific RND3 transgenic or knockout mice to establish causality .
Primary cell isolation: Isolate primary aortic endothelial cells (ECs) from these models for in vitro studies .
Pyroptosis induction: Challenge ECs with oxidized low-density lipoprotein (oxLDL) to induce pyroptosis in vitro .
Molecular analysis: Examine pyroptosis markers including NLRP3, Caspase1, and GSDMD-N through Western blot and flow cytometry (using propidium iodide and Caspase-1 double staining) .
Gain/loss-of-function studies: Use adenoviral vectors (e.g., Ad-Flag-Rnd3) for overexpression studies and genetic knockouts for loss-of-function studies .
Mechanistic investigation: Employ liquid chromatography tandem mass spectrometry (LC-MS/MS), co-immunoprecipitation assays, and molecular docking to identify interaction partners and signaling mechanisms .
This comprehensive approach has revealed that RND3 negatively regulates pyroptosis signaling by direct interaction with TRAF6, suppressing the TRAF6/NF-κB/NLRP3 pathway .
To effectively study RND3-TRAF6 protein interactions, researchers should implement the following methodological approaches:
Co-immunoprecipitation (Co-IP):
Immunofluorescence co-localization:
Domain mapping:
Functional validation:
Ubiquitination analysis:
Distinguishing between RND3's effects on different ubiquitination pathways, particularly K63-linked versus K48-linked TRAF6 ubiquitination, requires specialized experimental approaches:
Linkage-specific ubiquitin antibodies:
Ubiquitin mutants:
Employ ubiquitin constructs with mutations at specific lysine residues (K48R or K63R)
These mutants prevent the formation of specific linkage types
Time-course experiments:
Proteasome inhibition:
Use proteasome inhibitors (e.g., MG132) to block protein degradation
This helps distinguish between ubiquitination for signaling versus degradation purposes
Functional readouts:
Research has shown that RND3 suppresses K63-linked TRAF6 ubiquitination (which promotes signaling) while enhancing K48-linked TRAF6 ubiquitination (which promotes degradation), thereby inhibiting NF-κB activation and promoting TRAF6 degradation .
For optimal Western blot detection of RND3, researchers should follow these technical parameters:
Additionally, researchers should optimize membrane washing steps and incubation times based on their specific antibody and equipment. For atherosclerosis studies, aortic endothelial cell lysates can be used, noting that RND3 expression may be downregulated in disease models like Apoe KO mice .
When encountering challenges with RND3 antibodies, implement these systematic troubleshooting strategies:
For weak or absent signals:
For non-specific binding:
For inconsistent results between experiments:
For Co-IP specific issues:
Designing experiments to distinguish between RND3 and other Rho family GTPases requires careful consideration of their unique properties:
Antibody selection:
Functional assays:
Molecular approaches:
Design PCR primers or siRNAs targeting unique sequences
Verify knockdown/overexpression specificity by checking effects on other family members
Expression analysis:
Compare expression patterns across tissues or cell types, as different Rho GTPases have distinct expression profiles
Use qRT-PCR with highly specific primers for mRNA analysis
Interaction partners:
To effectively study RND3's role in cardiovascular disease models, researchers should implement this comprehensive experimental framework:
Animal model selection:
Cellular systems:
Molecular analysis techniques:
Functional assessments:
Translational considerations:
This comprehensive approach has revealed RND3's protective role against endothelial pyroptosis in atherosclerosis through regulation of the TRAF6/NF-κB/NLRP3 pathway .
When investigating RND3's role in pyroptosis pathways, these essential experimental controls must be included:
Expression controls:
Cell treatment controls:
Pathway validation controls:
Technical controls:
Animal model controls:
When faced with contradictory findings between in vitro and in vivo RND3 studies, researchers should consider these methodological factors:
Model complexity differences:
Expression level considerations:
Temporal dynamics:
Cell-type specific effects:
Signal integration:
Several promising research directions are emerging for RND3 antibodies in cardiovascular disease investigations:
Biomarker development:
Expanded pathway analysis:
Therapeutic target validation:
Mechanistic investigations:
Translational research:
These emerging areas highlight the potential of RND3 as both a therapeutic target and a mechanistic node in understanding cardiovascular disease pathogenesis .
Methodological improvements that can enhance RND3 detection and analysis in tissue samples include:
Advanced imaging techniques:
Implement super-resolution microscopy for improved subcellular localization
Use multiplex immunofluorescence to simultaneously detect RND3 and interacting partners
Apply tissue clearing techniques to enable 3D visualization in intact tissue samples
Single-cell analysis:
Employ single-cell RNA sequencing to identify cell-specific RND3 expression patterns
Use mass cytometry (CyTOF) with metal-conjugated RND3 antibodies for high-dimensional analysis
Apply spatial transcriptomics to correlate RND3 expression with tissue microenvironment
Improved antibody validation:
Quantitative analysis methods:
Combination approaches:
These methodological improvements will enable more precise characterization of RND3's role in cardiovascular disease progression and potentially reveal new therapeutic targets.