RAN Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Production

RAN recombinant monoclonal antibodies are genetically engineered antibodies produced using recombinant DNA technology. Unlike traditional hybridoma-derived antibodies, they are synthesized in vitro by cloning antibody genes into expression vectors (e.g., plasmids) and expressing them in mammalian host cells (e.g., CHO or HEK293) .

Key Production Steps:

  1. Gene Cloning: Light and heavy chain genes of anti-RAN antibodies are inserted into plasmids.

  2. Host Cell Transfection: Plasmids are transfected into mammalian cells for antibody expression.

  3. Purification: Antibodies are purified using Protein A/G chromatography .

Immunogen Sequence:
The immunogen for RAN recombinant monoclonal antibodies (e.g., clone ARC0986) includes the peptide sequence:
IVLCGNKVDI KDRKVKAKSI VFHRKKNLQY YDISAKSNYN FEKPFLWLAR KLIGDPNLEF VAMPALAPPE VVMDPALAAQ YEHDLEVAQT TALPDEDDDL .

Target Specificity:

  • Binds to the COOH-terminal acidic domain of RAN (residues 207–216: -DEDDDL), a region critical for interactions with nuclear transport factors like importin β .

  • Recognizes RAN only when complexed with transport factors (e.g., importin β, transportin) but not free RAN-GTP/GDP .

Applications:

ApplicationReactivityHost SpeciesIsotype
Western Blot (WB)Human, Mouse, RatMouseIgG2b
Immunohistochemistry (IHC)HumanMouseIgG2b
Immunofluorescence (IF)HumanMouseIgG2b
Data sourced from commercial antibody specifications .

Critical Studies:

  • Nuclear Transport Regulation:

    • ARAN1, a RAN monoclonal antibody, inhibits RanBP1 binding to the RAN–importin β complex, disrupting nuclear protein import and causing cytoplasmic accumulation of RAN .

    • Injected ARAN1 blocks nuclear import of SV-40 T-antigen substrates, demonstrating its utility in studying nuclear transport dynamics .

  • Structural Insights:

    • The COOH-terminal domain of RAN undergoes conformational changes upon binding to importin β, exposing the epitope recognized by recombinant antibodies .

  • Therapeutic Potential:

    • Recombinant RAN antibodies enable high-throughput screening for cancer therapeutics targeting cell cycle dysregulation .

Key Quality Metrics:

ParameterSpecification
Purity>98% (Protein A-purified)
Batch Consistency>95% (via SEC-HPLC)
Storage-20°C in PBS with 50% glycerol
Stability1 year post-shipment
Adapted from commercial and WHO guidelines .

Validation Standards:

  • EuroMAbNet Guidelines: Ensure specificity via immunofluorescence and immunoblotting .

  • WHO Compliance: Adheres to recombinant antibody production standards to minimize viral contaminants .

Advantages Over Traditional Antibodies

  • Reproducibility: No genetic drift due to defined DNA sequences .

  • Customization: Engineered for species switching (e.g., mouse to human IgG) and functional modifications (e.g., Fab fragments) .

  • Scalability: Suitable for large-scale therapeutic production .

Limitations and Considerations

  • Epitope Masking: The COOH-terminal domain is inaccessible in free RAN-GTP/GDP, limiting antibody utility in non-complexed states .

  • Functional Interference: ARAN1 alters endogenous RAN localization, necessitating controlled experimental conditions .

Future Directions

  • Diagnostic Development: Integration with RNAscope™ ISH Technology for spatial gene expression analysis .

  • High-Resolution Imaging: Engineered variants (e.g., rabbit or humanized IgG) for multispecies compatibility .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

The RAN recombinant monoclonal antibody is produced through a process involving the acquisition of RAN antibody genes, their introduction into suitable host cells, and subsequent culturing for antibody synthesis. This method yields highly pure, stable, and specific RAN recombinant monoclonal antibodies, enhancing their affinity. Following synthesis, the antibody undergoes purification via affinity chromatography and rigorous testing through various assays, including ELISA, IHC, IF, and FC. This antibody specifically targets the human RAN protein.

The GTP-binding nuclear protein Ran is a crucial regulator of nucleocytoplasmic transport, facilitating the movement of molecules between the nucleus and cytoplasm. Its vital role in maintaining proper cellular compartmentalization, coordinating mitotic processes, and regulating the integrity of nuclear pore complexes underscores its importance in fundamental cellular functions and cell cycle progression.

Form
Liquid
Lead Time
Typically, we can dispatch the products within 1-3 working days after receiving your orders. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
GTP-binding nuclear protein Ran (Androgen receptor-associated protein 24) (GTPase Ran) (Ras-like protein TC4) (Ras-related nuclear protein), RAN, ARA24
Target Names
RAN
Uniprot No.

Target Background

Function

RAN, a GTPase involved in nucleocytoplasmic transport, plays a pivotal role in both import and export processes of proteins and RNAs between the nucleus and cytoplasm. Its state transitions between a cytoplasmic GDP-bound and a nuclear GTP-bound form through nucleotide exchange and GTP hydrolysis. Nuclear import receptors, such as importin beta, bind their substrates only in the absence of GTP-bound RAN and release them upon direct interaction with GTP-bound RAN. Conversely, export receptors exhibit the opposite behavior. This mechanism ensures the controlled loading and release of cargo by transport receptors within the appropriate compartment, dictating the directionality of transport. Interaction with RANBP1 induces a conformational change in the complex formed by XPO1 and RAN, triggering the release of the nuclear export signal of cargo proteins.

Furthermore, RAN (in its GTP-bound form) initiates microtubule assembly at mitotic chromosomes, playing a critical role in normal mitotic spindle assembly and chromosome segregation. Its presence is essential for proper progression through mitosis. The complex formed with BIRC5/survivin contributes to mitotic spindle formation by acting as a physical scaffold to facilitate the delivery of the RAN effector molecule TPX2 to microtubules. Additionally, RAN serves as a negative regulator of the kinase activity of VRK1 and VRK2. It enhances AR-mediated transactivation, which decreases as the poly-Gln length within AR increases.

Gene References Into Functions
  1. The DICER rs3742330 AG+GG genotype has been associated with more advanced T stage compared to the AA genotype (P=0.009). Additionally, a higher number of patients with XPO5 rs2257082 CC genotype exhibited poorly differentiated tumors compared to carriers of the CT+TT genotype. Furthermore, carriers of RAN rs14035 CC genotype demonstrated a higher three-year OS rate than carriers of the CT+TT genotype (adjusted HR 3.174; 95% CI 1.010, 9.973; P=0.048). PMID: 29683064
  2. Collectively, the findings provide strong in vitro evidence for the involvement of Ran in breast cancer progression and suggest its potential as a therapeutic target and/or marker of disease. PMID: 29750309
  3. The molecular structure of the exportin Xpo4 in complex with RanGTP and the hypusine-containing translation factor eIF5A has been reported. PMID: 27306458
  4. Emerin mobility is dependent on RanGTP. In cells overexpressing YFP-Emerin, the mobility of YFP-Emerin was elevated in Samp1 knockout cells and reduced in cells overexpressing Samp1, suggesting that Samp1 significantly attenuates the mobility of Emerin in the nuclear envelope. The affinity between Samp1 and Emerin is diminished in the presence of Ran, indicating that Ran weakens the interaction between Samp1 and Emerin. PMID: 29510091
  5. KPNB1 and Ran protein collectively mediated the nuclear import of NDV M protein, demonstrating that KPNB1 protein interacts with NDV M protein to form a binary complex that subsequently enters the nucleus with the assistance of Ran protein. PMID: 29746765
  6. Ran GTPase promotes cancer progression through Met receptor-mediated downstream signaling. PMID: 27716616
  7. Data indicate that the distribution of components of the ras-related nuclear protein (Ran) pathway, influencing microtubule behaviors, is determined by their interactions with microtubules, resulting in microtubule nucleators being localized by the microtubules whose formation they stimulate. PMID: 27439876
  8. The RAS-related nuclear protein ((P) ran), breast cancer metastasis suppressor 1 ((P) brms1), and minichromosome maintenance complex component 5 ((P) mcm5) promoters possess the specificity and strength required for cancer-specific expression-targeted gene therapy. PMID: 27140445
  9. Research has shown that decreasing Ran-GTP levels or tethering active Ran to the equatorial membrane affects anillin's localization and causes cytokinesis phenotypes. PMID: 28931593
  10. Lysine acetylation regulates nearly all aspects of Ran function, including RCC1 catalyzed nucleotide exchange, intrinsic nucleotide hydrolysis, its interaction with NTF2, and the formation of import- and export-complexes. PMID: 26507377
  11. These results indicate that BLM enters the nucleus via the importin beta1, RanGDP, and NTF2 dependent pathway, demonstrating for the first time the nuclear trafficking mechanism of a DNA helicase. PMID: 29017749
  12. RAN translation is regulated by Muscleblind Proteins in Myotonic Dystrophy Type 2. PMID: 28910618
  13. Studies have shown that DICER (rs3742330) and RAN (rs14035) are associated with the survival of HCC patients. PMID: 27611467
  14. RAN translation from antisense CCG repeats generates novel proteins that accumulate in ubiquitinated inclusions in Fragile X-associated tremor/ataxia syndrome patients. PMID: 27761921
  15. Interaction of Samp1 and Ran in the inner nuclear membrane. PMID: 27541860
  16. RAN plays a crucial role in cell cycle, DNA repair, and DNA damage-induced cell senescence. PMID: 26864624
  17. Binding of Ran to NTF2 is essential for NTF2 to inhibit nuclear expansion and import of large cargo molecules. PMID: 26823604
  18. RAN is pivotal in linking spatiotemporal control of centrosome regulators to the cell cycle machinery. PMID: 26725228
  19. FGF2 nuclear translocation is regulated by Karyopherin-beta2 and Ran GTPase in human glioblastoma cells. PMID: 26056081
  20. Enterovirus 71-induced downregulation of miR-197 expression increased the expression of RAN, supporting the nuclear transport of essential viral proteins 3D/3CD and host protein hnRNP K for viral replication. PMID: 26581983
  21. Inter-cellular transport of Ran GTPase. PMID: 25894517
  22. Variations in RAN rs14035 of Korean patients are significantly associated with their risk of colorectal cancer. PMID: 26147304
  23. Data show that RNA-binding protein LIN28B coordinates the expression of the oncogene RAN protein and aurora A kinase (AURKA) in neuroblastoma. PMID: 26481147
  24. Elevated metaphase RanGTP levels utilize Ubr5 to couple overall chromosome congression to SAC silencing. PMID: 26438829
  25. Data indicate that importin-beta (impbeta) alters the nuclear pore's permeability in a Ran-dependent manner, suggesting that impbeta is a functional component of the nuclear pore complex (NPC). PMID: 25748139
  26. RAN nucleo-cytoplasmic transport and mitotic spindle assembly partners XPO7 and TPX2 have roles in serous epithelial ovarian cancer. PMID: 24625450
  27. A comparison of the GTPase reaction of the slower hydrolyzing GTPase Ran with Ras. PMID: 26272610
  28. Ran silencing did not affect the A375 invasive capability, while it was enhanced in 526 cells, suggesting that Ran knockdown, by AurkA downregulation, resulted in a Ran-independent enhanced melanoma cell invasion. PMID: 25444926
  29. Stress can disrupt the Ran gradients through RCC1-dependent and RCC1-independent mechanisms. PMID: 25452301
  30. Based on new structures, it is suggested that LM phosphorylation status may mediate Ran's selection of exportin(s) and cargo(s), diverting these native trafficking elements into the lethal antihost Nup phosphorylation pathways. PMID: 25331866
  31. PI3Kbeta regulates the nuclear envelope through upstream regulation of RCC1 and Ran. PMID: 25348717
  32. To better understand its molecular mechanism, a 2.9 A resolution crystal structure of human TRN-SR2 complexed with the small GTPase Ran has been determined. PMID: 24915079
  33. An association between recurrent pregnancy loss development and the polymorphism of the miRNA machinery gene RAN and combined genotype of DROSHA/DICER has been observed. PMID: 24769857
  34. Reducing the nuclear concentration of Ran is sufficient to induce reactive oxygen species irrespective of progerin. PMID: 24523287
  35. Our findings emphasize the significant role of Ran in differentiation, disease stage, and metastasis in human colorectal cancer. PMID: 22956174
  36. Our data suggest a model for mitotic spindle positioning in which RanGTP and CLASP1 collaborate to align the spindle along the long axis of the dividing cell. PMID: 23783028
  37. Thioredoxin-like protein 2 is overexpressed in colon cancer and promotes cancer cell metastasis by interacting with Ran. PMID: 23311631
  38. MiR-203 may act as a novel tumor suppressor in esophageal cancer through down-regulating the expression of Ran and miR-21. PMID: 24001611
  39. Polymorphism in the RAN gene is associated with HBV-related hepatocellular carcinoma. PMID: 23868705
  40. A model wherein one monomer of TRN-SR2 is bound to one monomer of RanGTP. PMID: 23878195
  41. Overexpressed Ran in pancreatic cancer tissues has been found to be highly correlated with the histological grade. Downregulation of Ran led to significant suppression of cell proliferation, cell cycle arrest at the G1/S phase, and induction of apoptosis. PMID: 24076388
  42. Data show that five genes, CKAP5, KPNB1, RAN, TPX2, and KIF11, were shown to be essential for tumor cell survival in both head and neck squamous cell carcinoma (HNSCC) and non-small cell lung cancer (NSCLC), but most particularly in HNSCC. PMID: 23444224
  43. Therefore, the Encephalomyocarditis virus leader protein:Ran GTPase binding, mediated by the linker-hinge, is a required step in the leader protein-induced nuclear transport inhibition. PMID: 23711384
  44. RCC1 facilitates a tight binding between the encephalomyocarditis virus leader and cellular Ran GTPase. PMID: 23536659
  45. Reduced RAN expression and disrupted transport between cytoplasm and nucleus are key events in Alzheimer's disease pathophysiology. PMID: 23308199
  46. Reduction in Ran levels causes cytoplasmic decrease and nuclear accumulation of importin alpha, leading to cellular senescence in normal cells. PMID: 23160023
  47. Results suggest that Ran is required for and is a potential therapeutic target of Myc-driven cancer progression in both breast and lung cancers. PMID: 23468463
  48. RanBPM loss of expression results in constitutive activation of the ERK pathway and promotes cellular events leading to cellular transformation and tumorigenesis. PMID: 23118896
  49. The increased expression of Ran in lung squamous cell carcinoma might be correlated with carcinogenesis. PMID: 21033436
  50. A novel connection between the hyper-activation of the small GTPase Ran and the matricellular protein SMOC-2 has been identified, with significant consequences for oncogenic transformation. PMID: 22679017
Database Links

HGNC: 9846

OMIM: 601179

KEGG: hsa:5901

STRING: 9606.ENSP00000376176

UniGene: Hs.10842

Protein Families
Small GTPase superfamily, Ran family
Subcellular Location
Nucleus. Nucleus envelope. Cytoplasm, cytosol. Cytoplasm. Melanosome.
Tissue Specificity
Expressed in a variety of tissues.

Q&A

What is Ran protein and why are monoclonal antibodies against it valuable for research?

Ran is a small GTPase (approximately 25 kDa) that functions as a key regulator for active nuclear transport. It cycles between GTP-bound and GDP-bound states, creating a concentration gradient across the nuclear envelope that drives directional transport. Recombinant monoclonal antibodies against Ran are particularly valuable because they allow researchers to investigate the protein's conformational changes, localization patterns, and interactions with transport factors.

The value of these antibodies lies in their ability to recognize specific epitopes or domains of Ran that may be exposed or masked depending on its nucleotide-bound state or interaction with partner proteins. For example, the monoclonal antibody ARAN1 recognizes an epitope in the COOH-terminal domain of Ran that becomes exposed only when Ran interacts with importin β or related transport factors, but not when Ran-GTP or Ran-GDP is alone . This property makes such antibodies powerful tools for studying the structural dynamics of Ran during nuclear transport processes.

How can researchers validate the specificity of a Ran monoclonal antibody?

Validating antibody specificity is crucial before using it in experimental applications. For Ran monoclonal antibodies, a multi-step validation process should include:

  • Immunoblotting analysis: The antibody should detect a single band of approximately 25 kDa in cell extracts from various species. ARAN1, for example, was validated by detecting a single 25-kDa band in cell extracts from mouse Ehrlich ascites tumor cells, BHK21 cells, human embryonic lung cells, and other cell types .

  • Two-dimensional electrophoresis: The antibody should recognize a single spot corresponding to Ran when total cell extracts are separated by two-dimensional electrophoresis .

  • Immunofluorescence microscopy: The staining pattern should match that of validated polyclonal anti-Ran antibodies. Ran typically shows predominantly nuclear localization with some cytoplasmic distribution .

  • Epitope mapping: Using truncated forms of recombinant Ran (e.g., GST-fusion proteins), researchers can determine the specific region recognized by the antibody. This is particularly important for understanding whether the antibody recognizes a conformational or linear epitope .

  • Cross-reactivity testing: Examine whether the antibody cross-reacts with other GTPases or related proteins.

What approaches are used to generate recombinant monoclonal antibodies against Ran?

Several approaches can be employed to generate recombinant monoclonal antibodies against Ran:

  • Hybridoma technology: This traditional approach involves immunizing mice with recombinant human Ran protein followed by fusion of spleen cells with myeloma cells. ARAN1, for instance, was produced by immunizing a BDF1 mouse with 50 μg of denatured recombinant human Ran, followed by three subsequent injections at 3-week intervals. Spleen cells were then fused with mouse myeloma cell line P3U1, and hybridomas were screened by ELISA and immunoblotting .

  • Phage display: This technique allows for the selection of antibody fragments that bind to Ran from large libraries of antibody genes displayed on the surface of bacteriophage.

  • Single B cell cloning: This more recent approach involves isolating antigen-specific B cells and cloning their antibody genes.

  • Transcriptionally active PCR (TAP): This method uses PCR to produce transcriptionally active linear DNA fragments (minigenes) for both heavy and light antibody chains, allowing for rapid generation of recombinant antibodies from single antigen-specific antibody-secreting cells .

How can Ran monoclonal antibodies be used to investigate conformational changes during protein-protein interactions?

Monoclonal antibodies that recognize specific conformational states of Ran are powerful tools for investigating structural changes during protein-protein interactions. The ARAN1 antibody provides an excellent example of this application:

  • Solution binding assays: These revealed that ARAN1 recognizes Ran only when it is complexed with importin β, transportin, or CAS, but not Ran-GTP or Ran-GDP alone. This indicates that the COOH-terminal domain of Ran is exposed only upon interaction with these transport factors .

  • Competition assays: ARAN1 was found to suppress the binding of RanBP1 to the Ran-importin β complex, suggesting that the epitope recognized by ARAN1 overlaps with or affects the binding site for RanBP1 .

  • Structural analysis: By determining the exact epitope recognized by the antibody (in the case of ARAN1, the highly negatively charged COOH-terminal portion, -DEDDDL), researchers can identify domains that undergo conformational changes during protein interactions .

  • Immunoprecipitation analysis: When performed with crude cell extracts in the presence of Q69L Ran-GTP (a mutant that stabilizes the complexes of importin β-related transport factors and Ran-GTP), importin β, CAS, and transportin were found to co-precipitate with Ran using ARAN1, confirming the antibody's specificity for the complexed form of Ran .

These approaches allow researchers to use monoclonal antibodies as conformational sensors, providing insights into how Ran's structure changes during its functional cycle.

What methodological strategies can be employed to study Ran's role in nucleocytoplasmic transport using monoclonal antibodies?

Several methodological strategies utilizing monoclonal antibodies can elucidate Ran's role in nucleocytoplasmic transport:

  • Microinjection experiments: Injecting antibodies like ARAN1 into specific cellular compartments (nucleus or cytoplasm) can help track Ran's movement and interactions. ARAN1, when injected into the nucleus of BHK cells, was rapidly exported to the cytoplasm, indicating that the Ran-importin β-related protein complex is exported as a complex from the nucleus to the cytoplasm in living cells .

  • Transport inhibition studies: Antibodies that disrupt specific Ran interactions can be used to determine the functional significance of these interactions. ARAN1, when injected into cultured cells, prevented the nuclear import of SV-40 T-antigen nuclear localization signal substrates, demonstrating the importance of Ran recycling for nuclear protein transport .

  • Localization studies: By examining the distribution of endogenous Ran after antibody injection, researchers can identify key regulatory steps. Injection of ARAN1 caused the accumulation of endogenous Ran in the cytoplasm, suggesting that RanBP1 binding to the Ran-importin β complex is required for the dissociation of the complex in the cytoplasm .

  • Confocal microscopy time-lapse imaging: Combined with fluorescently labeled antibodies, this technique can provide real-time visualization of Ran dynamics.

  • FRET-based approaches: Using antibody fragments conjugated with fluorophores to detect conformational changes in Ran during transport cycles.

The table below summarizes the observed effects of ARAN1 injection on cellular processes:

Injection SiteTime Post-InjectionARAN1 LocalizationEffect on Ran DistributionEffect on Nuclear Import
Nucleus30 minutesCytoplasmAccumulation in cytoplasmInhibition
Cytoplasm30 minutesCytoplasmAccumulation in cytoplasmInhibition

How can epitope mapping of Ran monoclonal antibodies reveal functional domains of the protein?

Epitope mapping of monoclonal antibodies against Ran can reveal functionally important domains of the protein through several approaches:

  • Truncation analysis: By creating a series of truncated forms of recombinant Ran (e.g., GST-fusion proteins) and analyzing their interaction with antibodies by immunoblotting, researchers can identify the region containing the epitope. For ARAN1, removal of the COOH-terminal seven amino acids (210-216) of Ran completely abolished reactivity, indicating that this region contains the epitope .

  • Peptide mapping: Synthetic peptides corresponding to different regions of Ran can be tested for antibody binding. A 10-mer peptide of the COOH-terminal domain (residues 207-216) of Ran fused to GST was recognized by ARAN1 in both immunoblotting and solution binding assays, confirming the epitope location .

  • Mutational analysis: Point mutations in specific residues can identify critical amino acids within the epitope that are essential for antibody binding.

  • Structural correlation: By correlating epitope locations with known structural elements of Ran, researchers can gain insights into domain functions. The COOH-terminal domain recognized by ARAN1 is highly negatively charged (-DEDDDL) and conserved among species, suggesting functional importance .

  • Functional domain exposure analysis: The differential recognition of Ran by ARAN1 depending on its interaction state (recognizing Ran-importin β complex but not Ran alone) revealed that the COOH-terminal domain undergoes a conformational change when Ran interacts with transport factors .

This approach led to the discovery that the COOH-terminal domain of Ran is not exposed to the surface of the molecule until Ran interacts with importin β or related transport factors, suggesting a regulatory role for this domain in transport complex formation and disassembly .

What are the key methodological differences between generating antibodies against Ran-GTP versus Ran-GDP conformations?

Generating conformation-specific antibodies against Ran-GTP versus Ran-GDP requires careful consideration of several methodological factors:

  • Antigen preparation:

    • For Ran-GTP-specific antibodies: Recombinant Ran must be stably loaded with non-hydrolyzable GTP analogs (such as GTPγS or GMPPNP) or use the GTPase-deficient mutant RanQ69L charged with GTP .

    • For Ran-GDP-specific antibodies: Recombinant Ran can be loaded with GDP through nucleotide exchange protocols.

  • Stabilization strategies:

    • The GTP-bound form can be stabilized by using the Q69L mutation, which has reduced GTPase activity .

    • Both forms require purification under conditions that maintain the nucleotide-bound state, typically including the appropriate nucleotide in all buffers.

  • Screening approaches:

    • Differential screening protocols must be employed to identify clones that specifically recognize one conformation but not the other.

    • ELISA and immunoblotting with both Ran-GTP and Ran-GDP forms are used to identify conformation-specific antibodies.

  • Validation methods:

    • Solution binding assays comparing antibody recognition of Ran-GTP versus Ran-GDP.

    • Structural studies to confirm the conformational state of the antigen.

    • Functional assays to verify that the antibody recognizes the intended conformational state in cellular contexts.

It's worth noting that antibodies like ARAN1 provide insights into how such conformational differences might be detected. While ARAN1 itself is not strictly conformation-specific (it recognizes both Ran-GTP-importin β and Ran-GDP-importin β complexes), it demonstrates how antibodies can detect structural changes that occur upon protein-protein interactions .

What controls should be included when using Ran monoclonal antibodies in immunoprecipitation experiments?

When using Ran monoclonal antibodies in immunoprecipitation experiments, several critical controls should be included:

  • Isotype control: Use a matched isotype control antibody (e.g., normal mouse IgG for a mouse monoclonal) to assess non-specific binding. This was employed as a control for ARAN1 (IgG2b) experiments .

  • Input sample control: Include an aliquot of the starting material before immunoprecipitation to verify the presence of target proteins.

  • Negative control samples: Immunoprecipitate from cells or extracts lacking Ran expression, if available.

  • Competition controls: Pre-incubate the antibody with purified antigen (e.g., recombinant Ran) to block specific binding sites before immunoprecipitation.

  • Nucleotide state controls: When studying Ran-GTP versus Ran-GDP interactions, include controls with non-hydrolyzable GTP analogs or specific mutations like Q69L Ran-GTP that stabilize the GTP-bound form .

  • Interaction partner controls: When studying Ran complexes, verify the presence of known interaction partners (e.g., importin β, transportin, CAS) in the immunoprecipitates using specific antibodies .

  • Validation with multiple antibodies: If possible, perform parallel immunoprecipitations with different antibodies against Ran to confirm results.

For example, in the study with ARAN1, immunoprecipitation analysis was performed with mouse Ehrlich ascites tumor cell cytosolic extract in the presence of Q69L Ran-GTP to stabilize the complexes of importin β-related transport factors and Ran-GTP. Western blotting confirmed that importin β, CAS, and transportin were co-precipitated with Ran by ARAN1, validating the antibody's specificity for the Ran-transport factor complexes .

How can researchers optimize conditions for using Ran monoclonal antibodies in live-cell imaging studies?

Optimizing conditions for using Ran monoclonal antibodies in live-cell imaging studies requires addressing several technical challenges:

  • Antibody format selection:

    • Full IgG molecules are large (150 kDa) and may interfere with protein function

    • Consider using smaller formats such as Fab fragments (~50 kDa) or single-chain variable fragments (scFv, ~25 kDa)

    • For ARAN1-type studies, the IgG format was suitable for microinjection experiments, but smaller formats might provide less interference

  • Fluorescent labeling strategies:

    • Direct labeling with small fluorophores (e.g., Alexa Fluor dyes) minimizes interference

    • Maintain a suitable dye-to-antibody ratio (typically 2-4 dyes per antibody) to avoid quenching

    • Verify that labeling does not affect antibody binding properties

  • Delivery methods:

    • Microinjection: As demonstrated with ARAN1, allows precise delivery into specific cellular compartments

    • Cell-penetrating peptides: Can facilitate antibody entry without microinjection

    • Electroporation: Suitable for difficult-to-transfect cell types

  • Imaging parameters:

    • Use minimal laser power and exposure times to reduce phototoxicity

    • Consider spinning disk or light-sheet microscopy for extended imaging sessions

    • Employ deconvolution algorithms to improve signal-to-noise ratio

  • Controls and validation:

    • Include non-binding antibody controls labeled with the same fluorophore

    • Validate antibody specificity in fixed cells before live-cell experiments

    • Confirm that antibody binding does not significantly alter Ran function

  • Physiological considerations:

    • Maintain physiological temperature, pH, and CO₂ levels during imaging

    • Determine the optimal antibody concentration that provides sufficient signal without disrupting function

    • Consider the timing of experiments relative to the cell cycle, as Ran functions may vary

The study with ARAN1 demonstrated successful use of antibody microinjection followed by fixation and immunofluorescence , but adapting such approaches for real-time live-cell imaging would require these additional optimizations.

How can researchers address potential artifacts when using Ran monoclonal antibodies to study nuclear transport?

When using Ran monoclonal antibodies to study nuclear transport, several potential artifacts may arise. Here are strategies to address them:

  • Antibody-induced conformational changes:

    • Problem: Antibody binding may alter Ran's conformation or interaction capabilities

    • Solution: Use multiple antibodies targeting different epitopes to corroborate findings

    • Validation: Compare results with alternative approaches such as FRET-based sensors or proximity ligation assays

  • Interference with Ran's functional cycle:

    • Problem: Antibodies like ARAN1 can intentionally or unintentionally block specific interactions (e.g., RanBP1 binding)

    • Solution: Use antibodies at titrated concentrations and as tools to specifically inhibit distinct steps

    • Control: Always include time-course experiments to distinguish between primary effects and secondary consequences

  • Fixation artifacts in immunofluorescence:

    • Problem: Different fixation methods may affect epitope accessibility or Ran localization

    • Solution: Compare multiple fixation protocols (paraformaldehyde, methanol, etc.)

    • Validation: Correlate findings with live-cell experiments when possible

  • Epitope masking in complexes:

    • Problem: Some antibodies may not recognize Ran when it's in certain complexes

    • Solution: Use complementary antibodies like ARAN1 that specifically recognize complexed forms

    • Control: Include biochemical assays to verify complex formation independent of antibody detection

  • Off-target effects of high antibody concentrations:

    • Problem: High concentrations of injected antibodies may cause non-specific effects

    • Solution: Establish dose-response relationships and use the minimum effective concentration

    • Control: Include non-binding antibody controls at equivalent concentrations

The study with ARAN1 addressed several of these concerns by using both nuclear and cytoplasmic injection approaches, comparing results with control IgG injections, and correlating antibody localization with effects on Ran distribution and nuclear import .

What are the most effective strategies for distinguishing between direct and indirect effects when using inhibitory Ran antibodies?

Distinguishing between direct and indirect effects when using inhibitory Ran antibodies like ARAN1 requires a systematic approach:

  • Temporal analysis:

    • Perform time-course experiments to determine the sequence of events following antibody introduction

    • Rapid effects (within minutes) are more likely to be direct consequences

    • For example, ARAN1 localized to the cytoplasm within 30 minutes after injection, coinciding with changes in Ran distribution

  • Concentration dependence:

    • Establish dose-response relationships to identify threshold concentrations

    • Direct effects typically show clearer concentration dependence

    • Compare with known direct inhibitors of Ran function

  • Rescue experiments:

    • Attempt to rescue the antibody-induced phenotype by co-injecting excess recombinant Ran protein

    • Successful rescue suggests the antibody effect is specific to Ran

    • Alternatively, introduce recombinant interaction partners to compete with antibody binding

  • Domain-specific mutants:

    • Use cells expressing Ran mutants lacking specific functional domains (e.g., COOH-terminal truncations)

    • Compare antibody effects on wild-type versus mutant cells

    • This approach can reveal whether the antibody's effect depends on specific Ran domains

  • Targeted validation experiments:

    • Design experiments to test specific hypotheses about the mechanism of inhibition

    • For ARAN1, the hypothesis that it prevents RanBP1 binding to the Ran-importin β complex could be directly tested in vitro

    • In vitro competition assays can verify direct molecular interactions

  • Comparison with genetic approaches:

    • Compare antibody effects with those of Ran knockdown/knockout or expression of dominant-negative Ran mutants

    • Similar phenotypes support the specificity of antibody effects

The study with ARAN1 employed several of these strategies, including comparing the effects of nuclear versus cytoplasmic injection, using control antibodies, and correlating the findings with biochemical assays of Ran-importin β-RanBP1 interactions .

How can researchers interpret conflicting data between antibody-based and genetic approaches to studying Ran function?

When researchers encounter conflicts between antibody-based and genetic approaches to studying Ran function, a systematic interpretative framework becomes essential:

  • Temporal differences in intervention:

    • Antibodies (like ARAN1) typically cause acute inhibition, while genetic approaches (knockdown/knockout) lead to chronic depletion

    • Acute inhibition may reveal immediate functions without compensatory mechanisms

    • Example: ARAN1 injection showed rapid effects on Ran localization within 30 minutes, which might differ from long-term genetic interventions

  • Domain-specific versus complete protein effects:

    • Antibodies often target specific domains (ARAN1 targets the COOH-terminal domain), while genetic approaches affect the entire protein

    • Compare domain-specific genetic mutations with domain-specific antibodies

    • The COOH-terminal domain recognized by ARAN1 has specific functions that may not be apparent in complete Ran depletion

  • Gain-of-function versus loss-of-function:

    • Some antibodies may stabilize specific conformations (gain-of-function) rather than simply blocking function

    • ARAN1 appears to stabilize the Ran-importin β complex, preventing its dissociation by RanBP1

  • Resolution of apparent conflicts:

    • Analyze the exact molecular step affected by each approach

    • Design experiments to test specific hypotheses explaining the discrepancies

    • Use alternative approaches (e.g., optogenetic tools, small molecule inhibitors) as tiebreakers

  • Experimental context considerations:

    • Cell type differences may contribute to conflicting results

    • Cell cycle stage can affect Ran function and should be controlled

    • Subcellular targeting of interventions (e.g., nuclear vs. cytoplasmic injection of ARAN1) may produce different outcomes

  • Integrated model development:

    • Develop models that incorporate all data, explaining apparent conflicts

    • For example, if antibody studies suggest the COOH-terminal domain is essential for RanBP1 binding but genetic studies with COOH-terminal mutations show mild phenotypes, this might indicate redundant mechanisms or thresholds effects

    • The ARAN1 study suggested a specific model where RanBP1 binding to the Ran-importin β complex requires the exposed COOH-terminal domain of Ran

This integrative approach allows researchers to extract maximum insight from seemingly conflicting data about Ran function.

What emerging technologies might enhance the development of next-generation Ran monoclonal antibodies?

Several emerging technologies hold promise for developing next-generation Ran monoclonal antibodies with enhanced properties:

  • Single B cell cloning and sequencing technologies:

    • Rapid isolation of antigen-specific B cells followed by sequencing of antibody genes

    • Enables direct cloning of naturally paired heavy and light chains

    • Potentially faster than traditional hybridoma approaches used for antibodies like ARAN1

  • Transcriptionally active PCR (TAP) approaches:

    • Production of transcriptionally active linear DNA fragments (minigenes) for both heavy and light chains

    • Allows rapid generation of recombinant antibodies from single antigen-specific antibody-secreting cells

    • Could accelerate development of diverse anti-Ran antibodies targeting different epitopes

  • Structural biology-guided antibody engineering:

    • Using cryo-EM or X-ray crystallography data of Ran in various conformational states to design antibodies against specific structural features

    • Computational modeling to predict epitopes that become exposed during specific Ran interactions

    • May yield antibodies with enhanced specificity for distinct Ran conformations

  • Nanobody/single-domain antibody development:

    • Much smaller than conventional antibodies (~15 kDa vs. ~150 kDa)

    • Can access epitopes in protein complexes that might be inaccessible to larger antibodies

    • Potentially less disruptive for intracellular applications compared to larger antibodies like ARAN1

  • Antibody-fluorescent protein fusions:

    • Direct genetic fusion of fluorescent proteins to antibody fragments

    • Enables live-cell imaging without chemical labeling

    • Could enhance monitoring of Ran dynamics in real-time

  • Switchable antibody technologies:

    • Light- or small molecule-controllable antibodies that can be activated or inactivated on demand

    • Would allow temporal control over antibody function for studying dynamic Ran processes

    • Could minimize adaptation responses observed with constitutively active antibodies

These technologies could substantially expand the toolkit available for studying Ran's diverse functions in nuclear transport and other cellular processes.

How might structural biology approaches complement antibody-based studies of Ran conformational changes?

Structural biology approaches can powerfully complement antibody-based studies of Ran conformational changes through several synergistic strategies:

  • Co-crystallization of antibody-Ran complexes:

    • Determining atomic resolution structures of antibodies like ARAN1 bound to Ran complexes

    • Would provide direct visualization of the exposed COOH-terminal domain in the importin β-bound state

    • Could reveal the structural basis for the conformational change that exposes this domain

  • Cryo-electron microscopy (cryo-EM) of transport complexes:

    • Visualizing larger assemblies of Ran with transport factors and antibodies

    • Could capture dynamic intermediates in the transport cycle

    • May reveal how antibodies like ARAN1 prevent RanBP1 binding to the Ran-importin β complex

  • Hydrogen-deuterium exchange mass spectrometry (HDX-MS):

    • Mapping regions of Ran that undergo conformational changes upon binding to transport factors

    • Can identify protected regions when antibodies bind

    • Would complement the epitope mapping studies performed with ARAN1

  • Single-molecule FRET combined with antibody binding:

    • Monitoring real-time conformational changes in Ran using fluorescent donors and acceptors

    • Observing how antibody binding affects these conformational dynamics

    • Could provide kinetic information about the structural transitions

  • Nuclear magnetic resonance (NMR) spectroscopy:

    • Studying the dynamics of specific domains (like the COOH-terminal region) in solution

    • Characterizing how these dynamics change upon antibody binding

    • Particularly useful for flexible regions like the acidic COOH-terminal domain recognized by ARAN1

  • Integrative structural biology approaches:

    • Combining multiple structural techniques with computational modeling

    • Building comprehensive models of the conformational changes throughout the Ran cycle

    • Using antibody binding data as constraints for these models

The information from these structural studies would provide a molecular foundation for understanding the conformational switches in Ran that were initially revealed by antibodies like ARAN1, which showed that the COOH-terminal domain becomes exposed only upon interaction with importin β-related transport factors .

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.