RANGAP1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to RANGAP1 Antibody

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) .

Key Features of RANGAP1 Antibodies

Commercial RANGAP1 antibodies exhibit distinct properties:

SupplierProduct IDReactivityApplicationsMolecular Weight
Cell Signaling Tech#36067Human, Mouse, Rat, MonkeyWB, IP, eCLIP68 kDa, 82 kDa
Proteintech27405-1-APHuman, MouseWB, IHC, IF, ELISA64 kDa, 82 kDa
Abcamab2081Human, MouseWB, ICC/IF64 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 .

Applications in Research

RANGAP1 antibodies are utilized in diverse experimental contexts:

  • Cancer biology:

    • RanGAP1 is upregulated in colorectal cancer (CRC) and promotes metastasis via m6A methylation and MAPK signaling .

    • Mutant KRAS binds RanGAP1 to enhance XPO1-dependent nuclear export of oncoproteins like EZH2 .

  • Cardiac transplantation:

    • Elevated serum RanGAP1 levels predict acute cellular rejection (ACR) in heart transplant patients (AUC = 0.70, cutoff ≥90 ng/mL) .

  • Vascular biology:

    • RanGAP1 depletion reduces smooth muscle cell proliferation and migration, implicating it in neointima formation .

Table 1: Key Studies Using RANGAP1 Antibodies

StudyKey FindingsAntibody Used
Colorectal cancer progression METTL3/YTHDF1-mediated m6A modification upregulates RanGAP1, driving CRC metastasis.Not specified
Nuclear transport regulation CRM1 inhibition redistributes RanGAP1 from cytoplasm/nuclear pores to nucleoplasm.Anti-Myc, anti-RanGAP1
Cardiac rejection Serum RanGAP1 ≥90 ng/mL predicts ACR (OR = 6.8, p = 0.006).ELISA-based detection
Vascular injury RanGAP1 silencing reduces smooth muscle proliferation by 57% and migration by 48%.Custom siRNA + IHC

Challenges and Considerations

  • 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 .

Future Directions

  • Therapeutic targeting: RanGAP1 inhibition may synergize with KRAS or XPO1 inhibitors in cancers .

  • Biomarker development: Serum RanGAP1 quantification could non-invasively monitor cardiac rejection .

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Stored at -20°C. Avoid freeze-thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days of receiving them. Delivery times may vary depending on the purchase method and location. Please consult your local distributor for specific delivery times.
Synonyms
Fug 1 antibody; Fug1 antibody; GTPase-activating protein, RAN, 1 antibody; KIAA1835 antibody; MGC20266 antibody; OTTHUMP00000028918 antibody; OTTHUMP00000198755 antibody; OTTHUMP00000198756 antibody; OTTHUMP00000198757 antibody; OTTHUMP00000198758 antibody; RAGP1_HUMAN antibody; Ran 1 antibody; RAN GTPase activating protein 1 antibody; Ran GTPase-activating protein 1 antibody; Ran1 antibody; RANGAP 1 antibody; RANGAP antibody; RanGAP1 antibody; SD antibody; Segregation distorter homolog antibody; Segregation distortion antibody
Target Names
RANGAP1
Uniprot No.

Target Background

Function
RAN GTPase Activator. Catalyzes the conversion of cytoplasmic GTP-bound RAN to GDP-bound RAN, a crucial step in RAN-mediated nuclear import and export. Facilitates the dissociation of cargo from nuclear export complexes containing XPO1, RAN, and RANBP2 following nuclear export.
Gene References Into Functions
  • Increased RanGAP1 expression has been linked to drug resistance in Chronic Myeloid Leukemia. PMID: 27228340
  • Aberrant RanGAP1 localization has been observed in the cortex of Huntington's disease patients. PMID: 28384474
  • Our research demonstrates that RanGAP1 is actively transported between the nucleus and cytoplasm. Moreover, cytoplasmic and nuclear pore complex (NPC) localization of RanGAP1 depends on CRM1-mediated nuclear export. PMID: 26506250
  • The immune cell adaptor SLP-76 directly binds to SUMO-RanGAP1 of cytoplasmic fibrils at the nuclear pore complex. This interaction is essential for optimal NFATc1 and NF-kappaB p65 nuclear entry in T cells. PMID: 26321253
  • Differentiation of human coronary artery smooth muscle cells to a contractile phenotype through stepwise serum depletion results in a significant reduction of RanGAP1 protein levels. PMID: 24988324
  • Analysis of the dynamics of E2(Ubc9)-SUMO-Target(RanGAP1) in the presence and absence of E3(RanBP2) revealed that two distinct allosteric sites regulate the ligase activity. PMID: 21216249
  • The RanGAP1 consensus sumoylation site and SUMO-1 C terminus exhibit conformational flexibility. PMID: 15355965
  • The 3.0-A crystal structure of a four-protein complex comprising Ubc9, a Nup358/RanBP2 E3 ligase domain (IR1-M), and SUMO-1 conjugated to the carboxy-terminal domain of RanGAP1 has been determined. PMID: 15931224
  • RanGAP1 undergoes phosphorylation on Ser-358 both in vivo and in vitro. Phosphorylated RanGAP1, but not a mutant at 358S, forms a stable ternary complex with Ran and RanBP1 in vivo, suggesting that phosphorylation at 358S affects the Ran system. PMID: 16428860
  • Our findings reinforce the conclusion that mel-18 functions as an anti-SUMO E3 factor and extend its targets to include regulation of the sumoylation of the critical cellular protein RanGAP1. PMID: 18706886
Database Links

HGNC: 9854

OMIM: 602362

KEGG: hsa:5905

STRING: 9606.ENSP00000348577

UniGene: Hs.183800

Protein Families
RNA1 family
Subcellular Location
Cytoplasm. Nucleus, nucleoplasm. Nucleus envelope. Chromosome, centromere, kinetochore. Cytoplasm, cytoskeleton, spindle.
Tissue Specificity
Highly expressed in brain, thymus and testis.

Q&A

What is the functional significance of detecting both unmodified and SUMO-modified RANGAP1 in Western blots?

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.

How should researchers interpret RANGAP1 localization patterns during cell division?

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 PhaseRANGAP1 LocalizationFunction
PreprophaseConcentrated at PPBDivision plane marking
MetaphaseRemains at cortical division site (CDS); Bright dots on chromosomes (kinetochore regions)Maintains division plane memory
AnaphaseEnriched around spindle midzone while maintaining CDS localizationSpindle orientation regulation
CytokinesisPresent at forming cell plateCell 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

What are the critical validation steps for ensuring RANGAP1 antibody specificity in experimental applications?

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:

    • Human, mouse, and rat reactivity is common for many commercial antibodies

    • Xenopus laevis epitopes have been successfully targeted with synthetic peptide mixtures

  • 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

What methodological approaches overcome challenges in detecting cell-cycle-dependent RANGAP1 phosphorylation?

Detecting cell-cycle-dependent phosphorylation of RANGAP1 presents technical challenges requiring specialized approaches:

  • Synchronization optimization:

    • For smooth muscle cells, serum depletion for 48-72 hours has demonstrated effective synchronization with progressive RANGAP1 downregulation (-43.8±19.4% for 90 kD band; -76.2±8% for 70 kD band)

    • For dividing cells, thymidine-nocodazole blocks are preferable over single-method synchronization

  • 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:

    • Correlation with mitotic markers (cyclins, phospho-histone H3)

    • Measurement of catalytic activity using in vitro GTPase assays with recombinant Ran

How can researchers effectively use RANGAP1 antibodies to investigate nucleocytoplasmic transport defects in disease models?

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:

    • In vascular pathology models, correlate RANGAP1 expression with smooth muscle cell dedifferentiation markers (increased on day 3 post-injury, decreased by day 14)

    • In cancer models, analyze kinetochore-associated RANGAP1 in relation to chromosomal instability

  • Therapeutic response monitoring:

    • Evaluate RANGAP1 distribution and modification state as biomarkers for treatments targeting nuclear transport or SUMO pathways

What are the optimal strategies for using RANGAP1 antibodies to investigate the interplay between sumoylation and nucleocytoplasmic transport?

Investigating the interplay between sumoylation and nucleocytoplasmic transport requires sophisticated experimental strategies:

  • Sequential immunoprecipitation approach:

    • First IP with SUMO-specific antibodies followed by RANGAP1 Western blot

    • Alternative: RANGAP1 IP followed by SUMO detection

    • This approach has revealed significant SUMO-2/3 modification of RANGAP1 in SENP1/SENP2-depleted cells

  • SUMO isopeptidase inhibition/depletion studies:

    • RNAi targeting SENP1 and SENP2 (>90% reduction) reveals differential effects on SUMO-1 vs. SUMO-2/3 modification of RANGAP1

    • Acute chemical inhibition compared to genetic depletion helps distinguish direct from compensatory effects

  • 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:

    • Compare SUMO-1 vs. SUMO-2/3 modification patterns of RANGAP1

    • In SUMO-1 depleted cells, SUMO-2/3 modified RANGAP1 accumulates and localizes to nuclear pore complexes

  • Functional correlation with transport dynamics:

    • Measure Ran GTPase activity in relation to RANGAP1 sumoylation status

    • Quantitative transport assays with cargo-specific readouts

How should researchers design experiments to study the role of RANGAP1 in cell division using available antibodies?

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:

    • In plants, compare wildtype RANGAP1 antibody staining with WPP/AAP-GFP and RanGAP1ΔC-GFP mutants to distinguish targeting mechanisms

    • Correlate with point mutations that affect GAP activity but not localization (N219A, D330A)

  • Quantitative localization measurements:

    • For statistically robust analysis, examine multiple cells (25-69 cells per condition)

    • Use standardized scoring criteria for mitotic structure association

What controls are essential when using RANGAP1 antibodies to study its post-translational modifications?

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:

    • For SUMO studies: Compare SUMO-1 vs SUMO-2/3 specific antibodies

    • For phosphorylation: Lambda phosphatase treatment to remove phosphate groups

    • Use of cells depleted of specific modifying enzymes (e.g., SENP1/2 knockdown increased SUMO-2/3 modified RANGAP1)

What are the most effective strategies for optimizing RANGAP1 immunofluorescence protocols in different cell types?

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:

    • Optimal dilutions vary significantly between antibodies: 1:50-1:200 for ICC/IF applications is common

    • Cell-type specific validation is essential (see positive detection in specific cell lines)

  • 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

How can researchers resolve contradictory results when different RANGAP1 antibodies show varying localization patterns?

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:

    • RANGAP1 localization is cell-cycle dependent

    • Expression levels vary with differentiation state (e.g., decreased in quiescent smooth muscle cells)

    • Species-specific differences in localization patterns should be considered

Reference Table: RANGAP1 Antibody Applications and Performance

ApplicationRecommended Dilution RangeExpected Molecular WeightCell/Tissue Types Successfully UsedKey Controls
Western Blot1:500-1:50,00063-70 kDa (unmodified), 80-90 kDa (SUMO-modified)HeLa, HEK-293, MCF-7, Jurkat, NIH/3T3, U2OSsiRNA knockdown, recombinant protein
Immunofluorescence1:50-1:200N/AHeLa, NIH-3T3, HepG2, Plant root tipsWPP domain mutants, mitotic phase markers
Immunohistochemistry1:50-1:500N/AHuman stomach tissue, rat carotid tissueNon-injured control tissue, isotype control
Immunoprecipitation0.5-1 μg per IP reactionN/AHeLaIgG control, non-specific antibody control

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.