ARF13 Antibody

Shipped with Ice Packs
In Stock

Description

ARF Protein Family Overview

ARFs are small GTPases critical for intracellular vesicular trafficking, lipid metabolism, and cellular signaling. The human ARF family includes six members (ARF1–6), with ARF1 being the most extensively characterized .

ARF Family MemberKey FunctionsAntibody Availability
ARF1Golgi transport, lipid droplet regulationMultiple clones (e.g., ab183576, MA3-060)
ARF3Membrane traffickingOften co-targeted with ARF1 (e.g., ab129003)
ARF5/6Endosomal recycling, cytoskeletal dynamicsLimited commercial availability

ARF1 Antibodies: Key Research Tools

ARF1 antibodies are pivotal in studying cancer biology and metabolic pathways. Notable findings include:

Role in Cancer and Immune Regulation

  • Cancer Stem Cells (CSCs): ARF1 ablation disrupts lipid metabolism, inducing mitochondrial stress and necrosis in CSCs. This triggers dendritic cell activation and antitumor immunity .

  • Therapeutic Potential: ARF1 inhibition enhances chemotherapy efficacy by blocking tumor invasion pathways .

Mechanistic Studies

  • Lipid Metabolism: ARF1 sustains CSCs by regulating lipid droplets. Knockdown induces ER stress and DAMPs release, activating dendritic cells and T-cell responses .

  • Immunogenicity: ARF1-targeting antibodies (e.g., DFRF4539A) showed limited clinical efficacy in myeloma despite FcRH5 binding, highlighting challenges in ADC design .

Technical Validation

  • Western Blot: ARF1 antibodies consistently detect bands at ~18–21 kDa, with knockout controls confirming specificity .

  • Immunohistochemistry: Robust staining in human colon, breast, and lung carcinomas .

Gaps and Future Directions

  • ARF13 Ambiguity: The term "ARF13" may stem from nomenclature errors or emerging isoforms not yet cataloged in major databases.

  • Therapeutic Development: Next-gen ARF1 inhibitors (e.g., afucosylated antibodies) show promise in preclinical models but require clinical validation .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Composition: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
ARF13 antibody; At1g34170 antibody; F12G12.1 antibody; F23M19.15 antibody; Auxin response factor 13 antibody
Target Names
ARF13
Uniprot No.

Target Background

Function
Auxin response factors (ARFs) are transcription factors that bind specifically to the DNA sequence 5'-TGTCTC-3' found in the auxin-responsive promoter elements (AuxREs). These factors can act as either transcriptional activators or repressors. Formation of heterodimers with Aux/IAA proteins may influence their ability to regulate the expression of early auxin response genes.
Database Links

KEGG: ath:AT1G34170

UniGene: At.51184

Protein Families
ARF family
Subcellular Location
Nucleus.

Q&A

What are the key differences between ARF1 and ARL13B antibodies in terms of research applications?

ARF1 and ARL13B antibodies target distinct members of the ADP-ribosylation factor family but serve different research purposes. ARF1 antibodies primarily detect ADP-ribosylation factor 1, a small GTPase involved in protein trafficking between cellular compartments with critical roles in the Golgi complex functionality . These antibodies are frequently utilized in studies examining vesicular transport, membrane trafficking, and Golgi structure maintenance.

In contrast, ARL13B antibodies recognize ADP-ribosylation factor-like protein 13B, predominantly expressed in developing brain tissues . ARL13B antibodies find particular utility in neurodevelopmental research, ciliary studies, and investigation of Joubert syndrome (JBTS8), which is associated with ARL13B mutations. The specificity of each antibody determines its application scope in cellular and molecular biology research.

How can researchers verify the specificity of ARF1 antibodies for experimental validation?

Researchers should employ multiple complementary approaches to validate ARF1 antibody specificity:

  • Western blot analysis: Verify a single band of appropriate molecular weight (approximately 18-20 kDa for ARF1)

  • Positive and negative controls: Include known ARF1-expressing tissues/cells alongside knockout or low-expression samples

  • Immunoprecipitation followed by mass spectrometry: Confirm the identity of the precipitated protein

  • Cross-reactivity assessment: Test against related ARF family proteins (ARF3, ARF5, etc.)

  • Functional validation: Perform ARF1 activation assays using GST-GGA3-GAT pull-down methods to confirm the antibody detects the active form

A properly validated antibody should demonstrate consistent results across these validation methods, with particular attention to the distinction between ARF1 and other ARF family members with high sequence homology.

What are the optimal sample preparation methods when using ARF1 antibodies for studying Golgi complex dynamics?

Optimal sample preparation for ARF1 antibody-based studies of Golgi dynamics requires careful consideration of fixation methods, permeabilization protocols, and subcellular preservation techniques:

TechniqueRecommended ProtocolRationale
Cell Fixation4% paraformaldehyde (10 min) followed by methanol (-20°C, 5 min)Preserves ARF1 localization while maintaining Golgi structure
Permeabilization0.1% Triton X-100 (5 min)Allows antibody access without disrupting membrane integrity
Blocking5% BSA in PBS (1 hour)Reduces non-specific binding
Antibody Dilution1:100-1:500 rangeOptimal for immunocytochemistry/immunofluorescence (ICC/IF)
CounterstainingDAPI (nuclear) and GM130 (Golgi marker)Provides subcellular context for ARF1 localization

For live-cell imaging studies, researchers should consider using fluorescently tagged ARF1 constructs alongside fixed-sample antibody validation to confirm that tagged proteins reflect endogenous ARF1 distribution patterns.

How should researchers design experiments to study ARF1 activation in cancer cell models?

When investigating ARF1 activation in cancer models, particularly head and neck squamous cell carcinoma (HNSCC), researchers should implement the following experimental design:

  • Baseline activation measurement: Establish baseline ARF1 activation levels using GST-GGA3-GAT pull-down assays followed by Western blotting with anti-ARF1 antibodies

  • Comparative analysis: Compare ARF1 activation between metastatic and non-metastatic paired cell lines (e.g., metastatic HN12/HN31 versus non-metastatic HN4/HN30)

  • Pathway inhibition studies: Systematically inhibit upstream regulators (particularly EGFR phosphorylation) to assess impact on ARF1 activity

  • Small molecule intervention: Test ARF1-targeting compounds such as γ-dipeptides based on 4-amino-(methyl)-1,3-thiazole-5-carboxylic acid (ATC) scaffolds

  • Functional readouts: Measure cell viability, invasion capacity, and metastatic potential in both 2D and 3D culture systems following ARF1 modulation

This comprehensive approach enables researchers to establish causality between ARF1 activation status and cancer cell behaviors while identifying potential therapeutic intervention points.

What methodological approaches can distinguish between different activation states of ARF1 in complex tissue samples?

Distinguishing between GDP-bound (inactive) and GTP-bound (active) forms of ARF1 in complex tissue samples requires specialized techniques:

  • GST-GGA3-GAT pull-down assay: This gold-standard technique selectively captures GTP-bound ARF1 by leveraging the binding specificity of the GAT domain from GGA3, which only interacts with active ARF1 . The subsequent detection by Western blotting with ARF1-specific antibodies reveals the proportion of active ARF1.

  • Proximity ligation assay (PLA): Allows in situ detection of active ARF1 by visualizing interactions between ARF1 and its effector proteins that only bind the GTP-bound form.

  • Conformational-specific antibodies: Some specialized antibodies can distinguish between GDP-bound and GTP-bound conformations, though these require extensive validation.

  • FRET-based biosensors: For living tissue analysis, genetically encoded biosensors can report ARF1 activation in real-time by measuring changes in FRET efficiency upon ARF1 activation.

  • Mass spectrometry with crosslinking: Can identify ARF1 binding partners specifically associated with active conformations in tissue lysates.

When analyzing complex tissues, researchers should employ tissue-clearing techniques compatible with immunohistochemistry to preserve ARF1 activation state while enabling deep tissue imaging.

How can researchers correctly interpret ARF1 antibody data in the context of cholera toxin studies?

Interpreting ARF1 antibody data in cholera toxin research requires understanding of the specific functional relationship between ARF1 and cholera toxin components:

  • Mechanistic context: ARF1 functions as an allosteric activator of the cholera toxin catalytic subunit, specifically enhancing its ADP-ribosyltransferase activity . Therefore, detection of ARF1-cholera toxin complexes indicates potential toxin activation.

  • Subcellular localization analysis: Co-localization studies using ARF1 antibodies and labeled cholera toxin components should focus on endosomal compartments where the interaction typically occurs.

  • Activation state considerations: Only the GTP-bound form of ARF1 interacts productively with cholera toxin components, so researchers must distinguish between total ARF1 (detected by standard antibodies) and active ARF1 (detected by pull-down assays).

  • Quantitative assessment: The degree of ARF1-cholera toxin interaction correlates with toxin efficacy, making quantitative analysis of co-immunoprecipitation results particularly informative.

  • Technical controls: Include ARF1 inhibitor treatments (such as Brefeldin A) to establish specificity of observed interactions.

How does ARF1 activity correlation with HNSCC progression, and what antibody-based approaches best characterize this relationship?

Research demonstrates a clear correlation between ARF1 activity and HNSCC progression, with specific antibody-based approaches providing critical insights:

Studies have shown higher levels of active GTP-bound ARF1 in metastatic HNSCC cell lines (HN12, HN31) compared to their paired non-metastatic counterparts (HN4, HN30), suggesting a direct correlation between ARF1 activation status and cancer cell aggressiveness . This relationship appears mediated through interactions with phosphorylated EGFR on the plasma membrane of HNSCC cells.

The most effective antibody-based approaches for characterizing this relationship include:

  • Quantitative immunohistochemistry: Using validated ARF1 antibodies on patient tissue microarrays to correlate expression levels with clinical outcomes

  • Activation-specific analysis: Employing GST-GGA3-GAT pull-down assays with ARF1 antibodies to assess the proportion of active ARF1 in patient-derived samples

  • Proximity detection methods: Using in situ PLA to visualize ARF1-EGFR interactions in preserved tissue specimens

  • Phospho-EGFR correlation: Dual staining for ARF1 and phosphorylated EGFR to establish their spatial relationship in tumor samples

These approaches, particularly when combined with patient outcome data, provide robust characterization of how ARF1 activity contributes to HNSCC progression and metastatic potential.

What role does ARL13B play in neurodevelopmental disorders, and how can antibodies help elucidate disease mechanisms?

ARL13B plays crucial roles in neurodevelopmental processes, with mutations associated with Joubert syndrome (JBTS8), a rare brain malformation disorder . ARL13B antibodies serve as essential tools for investigating the underlying disease mechanisms:

  • Expression pattern analysis: ARL13B antibodies enable detailed mapping of protein expression throughout brain development, revealing spatiotemporal patterns critical for normal neurogenesis. The protein is primarily expressed in developing brain tissues .

  • Primary cilia investigation: As ARL13B localizes to primary cilia, antibody-based imaging allows visualization of ciliary morphology and function in neural progenitors and mature neurons.

  • Mutation impact assessment: By comparing ARL13B localization and abundance between wild-type and JBTS patient-derived cells, researchers can determine how specific mutations affect protein function.

  • Pathway interaction studies: Co-immunoprecipitation using ARL13B antibodies followed by mass spectrometry identifies interaction partners altered in disease states.

  • In vivo developmental tracking: Immunohistochemical analysis of brain sections throughout development using ARL13B antibodies enables correlation of protein expression with critical neurodevelopmental milestones.

These approaches have revealed that ARL13B dysfunction primarily affects ciliary signaling pathways essential for cerebellum and brainstem development, particularly those involving Sonic Hedgehog signaling.

What are the most common technical pitfalls when using ARF1 antibodies for immunoprecipitation, and how can researchers overcome them?

Researchers frequently encounter several technical challenges when using ARF1 antibodies for immunoprecipitation:

ChallengeCauseSolution
Low IP efficiencySmall size of ARF1 protein (18-20 kDa)Use high-affinity antibodies; optimize antibody-to-lysate ratios
Conformation-dependent epitope maskingGDP/GTP binding alters ARF1 structureInclude both native and denaturing conditions in protocol optimization
High background signalCross-reactivity with other ARF family membersPre-clear lysates; use highly specific monoclonal antibodies
Weak detection post-IPLow abundance of endogenous ARF1Scale up starting material; use sensitive detection methods like chemiluminescence
Loss of GTP-bound state during lysisGTP hydrolysis during sample preparationInclude GTPγS (non-hydrolyzable GTP analog) in lysis buffers

Additionally, researchers should consider using recombinant protein standards to establish IP efficiency and covalent crosslinking of antibodies to beads to prevent heavy/light chain interference during Western blot detection. When studying ARF1-interacting proteins, gentle elution conditions help maintain weak or transient interactions that might be biologically significant.

How should researchers address contradictory results between ARF1 antibody-based assays and functional studies?

When faced with discrepancies between antibody-based detection of ARF1 and functional observations, researchers should systematically investigate potential explanations:

  • Antibody validation reassessment: Verify antibody specificity using multiple approaches including Western blotting against recombinant ARF1, ARF3, and ARF5 to rule out cross-reactivity.

  • Post-translational modification effects: Consider whether specific modifications affect antibody recognition but not function (or vice versa). Phosphorylation, ubiquitination, or membrane interactions may mask epitopes.

  • Subcellular compartmentalization: The relevant ARF1 pool may be sequestered in specific cellular locations. Use fractionation approaches with activity assays to identify functionally relevant populations.

  • Temporal dynamics: ARF1 activation is often transient. Time-course experiments with both antibody detection and functional readouts can reconcile apparently contradictory snapshots.

  • Compensatory mechanisms: In knockdown/knockout studies, other ARF family members may compensate functionally while antibody-based detection correctly shows ARF1 reduction.

  • Technical approach complementarity: Employ orthogonal techniques like CRISPR-Cas9 genome editing, rescue experiments with mutant variants, and pharmacological intervention to build a consistent mechanistic model.

When reporting contradictory results, researchers should clearly document all experimental conditions, antibody validation efforts, and potential confounding factors to facilitate interpretation by the scientific community.

How can ARF1 antibodies be utilized to study the interplay between vesicular trafficking and cancer cell metastasis?

ARF1 antibodies enable sophisticated investigations into the mechanistic connections between vesicular trafficking dysregulation and cancer metastasis:

  • Invasive front analysis: Immunohistochemistry using ARF1 antibodies can reveal accumulation patterns at invasive fronts of tumors, particularly in HNSCC samples . This localization often coincides with invadopodia formation where regulated secretion drives matrix degradation.

  • Trafficking pathway dissection: Combined immunofluorescence with markers for different endocytic and exocytic compartments allows mapping of altered trafficking routes in metastatic versus non-metastatic cells.

  • Secretome regulation: Pulse-chase experiments with trafficking inhibitors alongside ARF1 activation monitoring can determine how ARF1 controls the release of metastasis-promoting factors.

  • Real-time invasion imaging: Live-cell imaging using antibody-based biosensors for ARF1 activation can visualize dynamic activation patterns during directed cell migration through matrices.

  • Therapeutic target validation: ARF1-targeting γ-dipeptides demonstrate potent anti-cancer effects by disrupting ARF1-dependent trafficking essential for maintaining the invasive phenotype .

These approaches have revealed that metastatic HNSCC cells maintain abnormally high levels of active GTP-bound ARF1, which supports the specialized secretory machinery required for invasive behavior. The association between ARF1 and phosphorylated EGFR on the plasma membrane appears to sustain this hyperactivation, making it a promising therapeutic intervention point.

What emerging technologies are enhancing the resolution and information content of ARL13B antibody-based imaging in neurodevelopmental research?

Revolutionary imaging technologies are transforming the utility of ARL13B antibodies in neurodevelopmental research:

  • Super-resolution microscopy: Techniques like STED, PALM, and STORM enable visualization of ARL13B distribution within primary cilia at nanometer-scale resolution, revealing previously unobservable organizational patterns.

  • Expansion microscopy: Physical expansion of specimens labeled with ARL13B antibodies allows conventional microscopes to achieve super-resolution imaging of ciliary structures in intact brain tissue.

  • Lattice light-sheet microscopy: Enables long-term, low-phototoxicity imaging of ARL13B-labeled cilia in developing organoids, capturing dynamic morphological changes during neurogenesis.

  • Correlative light and electron microscopy (CLEM): Combines the molecular specificity of ARL13B immunolabeling with ultrastructural context from electron microscopy, providing multi-scale characterization of ciliary defects.

  • Multiplexed epitope detection: Sequential antibody labeling and elution techniques allow simultaneous visualization of ARL13B alongside dozens of other proteins in the same specimen, creating rich datasets for network analysis.

  • Tissue clearing methods: Advanced clearing protocols like CLARITY and iDISCO maintain ARL13B antibody labeling while rendering entire developing brains transparent, enabling whole-organ analysis of ciliary patterns.

These technologies have revealed that ARL13B distribution within cilia is not uniform but exhibits nanoscale organizational patterns that correlate with functional specialization in different neural cell types during development.

What are the most promising research avenues for utilizing ARF1 antibodies in developing targeted cancer therapeutics?

Several high-potential research directions leverage ARF1 antibodies in the development of targeted cancer therapeutics:

  • Therapeutic antibody development: Converting research-grade ARF1 antibodies into therapeutic formats (like bispecific antibodies linking ARF1-expressing cells to immune effectors) represents an emerging approach for cancers with ARF1 hyperactivation.

  • Antibody-drug conjugate (ADC) targeting: For cancers with cell-surface ARF1 exposure, ADCs utilizing ARF1 antibodies could deliver cytotoxic payloads specifically to malignant cells.

  • Intrabody engineering: Developing cell-penetrating ARF1 antibody fragments that disrupt ARF1-EGFR interactions could specifically inhibit cancer cell invasion without affecting normal ARF1 functions .

  • Rational drug design guidance: ARF1 antibodies in structural biology applications (cryo-EM, X-ray crystallography) can reveal binding pockets for structure-based design of small molecule inhibitors, as demonstrated with ATC-based γ-dipeptides .

  • Precision medicine patient selection: Developing companion diagnostic assays using ARF1 antibodies could identify patients most likely to benefit from ARF1-targeting therapies based on activation levels in biopsy specimens.

These approaches are particularly promising for aggressive cancers like HNSCC, where ARF1 hyperactivation correlates with metastatic potential and poor patient outcomes .

How might comparative studies of ARF1 and ARL13B antibodies in different biological systems advance our understanding of GTPase evolution and functional adaptation?

Comparative studies using antibodies against ARF1 and ARL13B across diverse biological systems offer unique insights into GTPase evolution and functional specialization:

  • Evolutionary conservation mapping: Detecting these proteins across phylogenetically diverse organisms using cross-reactive antibodies can reveal structural elements preserved through evolutionary time, indicating core functional domains.

  • Tissue-specific expression patterns: Systematic immunohistochemical surveys across tissues and organs can identify specialization patterns that suggest functional adaptation to tissue-specific requirements.

  • Developmental trajectory analysis: Comparing expression timing and localization during embryogenesis across species may reveal how these GTPases were recruited for specialized functions during evolutionary innovation.

  • Interaction partner diversification: Immunoprecipitation studies across model organisms can identify both conserved and species-specific binding partners, illuminating how interaction networks evolved.

  • Structure-function relationships: Correlation of antibody epitope accessibility with functional readouts across species can identify regulatory mechanisms that emerged during functional adaptation.

This comparative approach has already revealed intriguing divergence: while ARF1 maintains highly conserved functions in membrane trafficking across eukaryotes, ARL13B has evolved specialized roles in ciliary biology particularly critical to vertebrate neurodevelopment , suggesting different evolutionary constraints on these related GTPases.

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.