RANGAP1 antibodies are immunoreagents designed to detect and quantify the RanGAP1 protein, a 587-amino-acid enzyme encoded by the RANGAP1 gene. RanGAP1 regulates the Ran GTPase cycle by accelerating GTP hydrolysis, ensuring proper nucleocytoplasmic transport and mitotic spindle assembly . Antibodies targeting RanGAP1 are widely used in Western blotting (WB), immunoprecipitation (IP), immunohistochemistry (IHC), and immunofluorescence (IF) .
Commercial RANGAP1 antibodies exhibit distinct properties:
| Supplier | Product ID | Reactivity | Applications | Molecular Weight |
|---|---|---|---|---|
| Cell Signaling Tech | #36067 | Human, Mouse, Rat, Monkey | WB, IP, eCLIP | 68 kDa, 82 kDa |
| Proteintech | 27405-1-AP | Human, Mouse | WB, IHC, IF, ELISA | 64 kDa, 82 kDa |
| Abcam | ab2081 | Human, Mouse | WB, ICC/IF | 64 kDa |
Notes:
Post-translational modification: RanGAP1 exists in unmodified (~68 kDa) and SUMOylated (~82 kDa) forms, detectable via WB .
Subcellular localization: Predominantly cytoplasmic and nuclear pore-associated, with nuclear accumulation under CRM1 inhibition .
RANGAP1 antibodies are utilized in diverse experimental contexts:
Cancer biology:
Cardiac transplantation:
Vascular biology:
Cross-reactivity: Some antibodies (e.g., Abcam ab2081) show limited validation in non-human species .
SUMOylation detection: Anti-RanGAP1 antibodies must distinguish between unmodified and SUMOylated forms .
Storage: Most antibodies require storage at -20°C with avoidance of freeze-thaw cycles .
When conducting Western blot analysis with RANGAP1 antibodies, researchers typically observe two distinct bands: one at approximately 63-70 kDa representing unmodified RANGAP1 and another at approximately 80-90 kDa corresponding to SUMO-modified RANGAP1 . This dual detection is scientifically significant as the ratio between these two forms provides valuable insights into:
Nuclear-cytoplasmic transport regulation status
Cell cycle phase (particularly in dividing cells)
Protein modification pathway functionality
The SUMO-modified form predominantly localizes to nuclear pore complexes through interaction with RanBP2, while unmodified RANGAP1 is mainly cytoplasmic . Methodologically, researchers should optimize protein extraction buffers to preserve both forms and consider using SUMO-specific antibodies for co-immunoprecipitation studies to confirm modification status.
RANGAP1 exhibits dynamic and highly specific localization patterns during cell division that serve as valuable markers for mitotic progression. In plant cells, RANGAP1 acts as a continuous positive marker of the division plane with distinct temporal-spatial distribution :
| Cell Division Phase | RANGAP1 Localization | Function |
|---|---|---|
| Preprophase | Concentrated at PPB | Division plane marking |
| Metaphase | Remains at cortical division site (CDS); Bright dots on chromosomes (kinetochore regions) | Maintains division plane memory |
| Anaphase | Enriched around spindle midzone while maintaining CDS localization | Spindle orientation regulation |
| Cytokinesis | Present at forming cell plate | Cell wall formation guidance |
To accurately interpret these patterns, researchers should:
Use high-resolution confocal microscopy with appropriate cell cycle markers
Consider the WPP domain's role in targeting (found to be both necessary and sufficient for mitotic targeting)
Correlate localization with functional studies using appropriate mutants
Ensuring RANGAP1 antibody specificity requires rigorous validation through multiple complementary approaches:
Western blot validation: Confirm detection of expected molecular weights (63-70 kDa for unmodified and 80-90 kDa for SUMO-modified forms)
RNAi/knockout controls: Verify signal reduction following RANGAP1 knockdown; several studies reported 75-76% reduction for the 70 kDa band and 31-43% reduction for the 90 kDa band following siRNA treatment
Cross-reactivity testing: Evaluate antibody performance across species based on epitope conservation:
Immunoprecipitation validation: Confirm ability to pull down RANGAP1 and its interaction partners (RanBP2, Ubc9)
Subcellular localization verification: Validate nuclear envelope and mitotic structure labeling patterns through co-localization with established markers
Detecting cell-cycle-dependent phosphorylation of RANGAP1 presents technical challenges requiring specialized approaches:
Synchronization optimization:
Phospho-specific detection methods:
Phospho-specific antibodies targeting known modification sites
Phos-tag™ SDS-PAGE to separate phosphorylated from non-phosphorylated forms
Lambda phosphatase treatment controls to confirm phosphorylation status
Mass spectrometry analysis:
Enrichment of phosphopeptides using titanium dioxide or immobilized metal affinity chromatography
Targeted multiple reaction monitoring (MRM) for specific phosphorylation sites
Functional correlation:
RANGAP1 antibodies provide powerful tools for investigating nucleocytoplasmic transport defects in disease models through multi-dimensional approaches:
Quantitative analysis of SUMO-modified vs. unmodified RANGAP1 ratios:
Altered ratios indicate disruption of the SUMO pathway or nuclear pore complex function
Quantitative Western blotting with dual-fluorescence detection allows precise measurement
Co-localization studies with nuclear pore complex components:
Triple immunofluorescence with RanBP2 and nucleoporins (detected by mAb 414)
Super-resolution microscopy to resolve nanoscale disruptions in complex assembly
Functional transport assays:
Correlation of RANGAP1 localization with import/export reporter protein distribution
FRAP (Fluorescence Recovery After Photobleaching) to measure transport kinetics
Disease-specific applications:
Therapeutic response monitoring:
Evaluate RANGAP1 distribution and modification state as biomarkers for treatments targeting nuclear transport or SUMO pathways
Investigating the interplay between sumoylation and nucleocytoplasmic transport requires sophisticated experimental strategies:
Sequential immunoprecipitation approach:
SUMO isopeptidase inhibition/depletion studies:
Proximity ligation assays (PLA):
Detection of in situ RANGAP1-SUMO interactions with single-molecule sensitivity
Spatial mapping of modification relative to nuclear pore complexes
SUMO paralogue specificity analysis:
Functional correlation with transport dynamics:
Measure Ran GTPase activity in relation to RANGAP1 sumoylation status
Quantitative transport assays with cargo-specific readouts
Comprehensive experimental design for studying RANGAP1 in cell division requires:
Temporal resolution optimization:
Time-lapse imaging with stably expressed fluorescent cell cycle markers
Synchronization followed by fixed-time-point analysis using indirect immunofluorescence
For plant cells, both methanol fixation (-20°C/6 mins) and paraformaldehyde fixation (3.7% PFA/10 min) have proven effective for RANGAP1 detection
Multi-protein co-localization analysis:
In plant cells, track RANGAP1 relative to PPB, CDS, kinetochores, and phragmoplast markers
In mammalian cells, focus on nuclear envelope breakdown and reformation
Domain-specific functional analysis:
Quantitative localization measurements:
Rigorous controls for studying RANGAP1 post-translational modifications include:
Expression system controls:
Recombinant RANGAP1 without modifications as baseline reference
Site-directed mutants eliminating specific modification sites
Pathway inhibition/activation controls:
SUMO pathway inhibitors (ginkgolic acid, 2-D08)
Proteasome inhibitors to prevent degradation of modified forms
Phosphatase inhibitors to preserve phosphorylation status
Subcellular fractionation controls:
Nuclear and cytoplasmic fractions with markers verifying separation purity
Nuclear envelope isolation for enrichment of SUMO-modified RANGAP1
Technical validation controls:
Alternative antibody clones recognizing different RANGAP1 epitopes
Peptide competition experiments to confirm specificity
Stably expressed tagged RANGAP1 as internal standard
Modification-specific controls:
Optimizing RANGAP1 immunofluorescence across different cell types requires consideration of several key parameters:
Fixation method optimization:
For plant cells: Both methanol (-20°C/6 mins) and paraformaldehyde (3.7% PFA/10 min at room temperature, 0.1% Triton/PBS 5 min, glycine 5 min) have been validated
For mammalian cells: 4% PFA followed by 0.1-0.5% Triton X-100 permeabilization
Cold methanol fixation may better preserve nuclear envelope structures
Antibody selection and dilution optimization:
Signal amplification strategies:
Tyramide signal amplification for low abundance detection
Fluorophore selection to avoid autofluorescence in specific tissues
Background reduction techniques:
Extended blocking (3-5% BSA, 5% normal serum from secondary antibody species)
Pre-adsorption of secondary antibodies with cellular proteins
Inclusion of detergents and carrier proteins to reduce non-specific binding
Imaging parameter optimization:
Z-stack acquisition with deconvolution for 3D structural analysis
For nuclear envelope imaging, super-resolution techniques provide superior resolution
Resolving contradictory RANGAP1 localization patterns requires systematic investigation:
Epitope mapping analysis:
Compare epitope locations of different antibodies relative to functional domains:
N-terminal WPP domain (plant RanGAP1)
LRR domain (Ran interaction site)
C-terminal acidic domain
Cross-validation with tagged constructs:
Compare antibody staining patterns with GFP-tagged RANGAP1 expressed at near-endogenous levels
Use multiple tag positions (N-terminal vs C-terminal) to identify tag interference effects
Conformational accessibility assessment:
SUMO-modification may mask certain epitopes
Different fixation protocols can affect epitope accessibility
Use multiple antibodies recognizing different regions in the same experiment
Antibody specificity validation:
Western blot confirmation of specificity prior to immunofluorescence
Peptide competition controls to verify specific binding
RANGAP1 knockdown/knockout controls to demonstrate specificity
Cell-type and condition-specific factors:
| Application | Recommended Dilution Range | Expected Molecular Weight | Cell/Tissue Types Successfully Used | Key Controls |
|---|---|---|---|---|
| Western Blot | 1:500-1:50,000 | 63-70 kDa (unmodified), 80-90 kDa (SUMO-modified) | HeLa, HEK-293, MCF-7, Jurkat, NIH/3T3, U2OS | siRNA knockdown, recombinant protein |
| Immunofluorescence | 1:50-1:200 | N/A | HeLa, NIH-3T3, HepG2, Plant root tips | WPP domain mutants, mitotic phase markers |
| Immunohistochemistry | 1:50-1:500 | N/A | Human stomach tissue, rat carotid tissue | Non-injured control tissue, isotype control |
| Immunoprecipitation | 0.5-1 μg per IP reaction | N/A | HeLa | IgG control, non-specific antibody control |