ARL16 antibodies are polyclonal or monoclonal immunoglobulins designed to detect ARL16, a divergent member of the ADP-ribosylation factor (ARF) family of GTPases. ARL16 plays dual roles in cellular biology:
Ciliary function: Regulates Golgi-to-cilium trafficking of IFT140 and INPP5E, critical for cilium formation and signaling .
Immune regulation: Inhibits RIG-I (Retinoic Acid-Inducible Gene I) signaling in antiviral responses via GTP-dependent interactions with its C-terminal domain .
The antibody’s specificity is determined by its immunogen sequence, which targets conserved regions of ARL16 across species.
ARL16 antibodies are pivotal in elucidating ARL16’s cellular and molecular roles.
Localization studies: Immunofluorescence revealed ARL16’s punctate localization along ciliary axonemes in human RPE1 cells and photoreceptor cells, co-localizing with mitochondrial markers (HSP60) .
Trafficking defects: In Arl16 knockout (KO) mouse embryonic fibroblasts (MEFs), ARL16 antibodies confirmed Golgi accumulation of IFT140 and INPP5E, linking ARL16 to Golgi export pathways .
RIG-I interaction: Western blotting and co-immunoprecipitation demonstrated that ARL16 binds RIG-I’s CTD, suppressing RNA virus-induced interferon-β production .
GTP-dependent inhibition: Mutant ARL16 (T37N or Δ45–54) lacking GTP-binding capacity failed to interact with RIG-I, confirming functional dependency on nucleotide status .
Commercially available ARL16 antibodies vary in specificity and validation. Below is a comparative analysis:
| Parameter | Sigma-Aldrich (HPA043711) | SAB (39855) | Atlas Antibodies (Hpa043711) | Thermo Fisher (PA5-60479) |
|---|---|---|---|---|
| Host | Rabbit | Rabbit | Rabbit | Rabbit |
| Immunogen | Human ARL16 (TQLSASCVQL...) | Human ARL16 (19–197aa) | Human ARL16 (TQLSASCVQL...) | Human ARL16 (TQLSASCVQL...) |
| Applications | IHC, ICC-IF, WB | ELISA, WB | IHC, ICC-IF, WB | WB, ELISA |
| Species Reactivity | Human | Human, Mouse, Rat | Human | Human, Mouse (80%), Rat (79%) |
| Validation | IHC tissue arrays, protein arrays | N/A | IHC, ICC-IF, WB | WB, ELISA |
| Cross-Reactivity | Minimal (via Prestige protocols) | N/A | Low (rigorous selection) | <80% with mouse/rat orthologs |
Data sourced from product specifications .
Arl16 KO phenotypes: ARL16 antibodies revealed reduced ciliation (~25% decrease) and elongated cilia in KO MEFs, reversible by ARL16-myc expression .
Golgi retention: IFT140 and INPP5E accumulate at the Golgi in Arl16 KO cells, while other IFT proteins (e.g., IFT88) remain unaffected, indicating pathway-specific defects .
Antiviral suppression: ARL16 overexpression inhibited Sendai virus-induced interferon-β production, while RNAi knockdown enhanced RIG-I signaling .
Structural basis: ARL16’s interaction with RIG-I CTD is GTP-dependent, as GDP-bound mutants (T37N) lost binding capacity .
Therapeutic potential: ARL16 antibodies may serve as tools to study ARL16’s role in ciliopathies (e.g., Bardet-Biedl syndrome) or viral immunopathology.
Mechanistic studies: Elucidating ARL16’s GTP hydrolysis mechanism (altered G-3 motif) and its interaction with PDE6D or GM130 .
Cancer research: Exploring ARL16’s role in Hedgehog signaling, implicated in a CRISPR screen .
ARL16 belongs to the ADP-ribosylation factor-like family of small GTPases involved in regulating vesicular trafficking processes within cells. Unlike its homologous proteins ARL1 and ARF1, ARL16 shows unique structural and functional properties. Sequence analysis reveals that human ARL16 shares only 25% identity and 43.4% similarity with ARF1, and 28.2% identity and 48.1% similarity with ARL1 . A key distinguishing feature is ARL16's altered G-3 motif (RELGGC), which differs significantly from the highly conserved WDXGGQ motif found in most GTPases. This structural variation suggests ARL16 employs an alternative mechanism for GTP hydrolysis and makes traditional methods of generating dominant active mutants (commonly targeting the Q71/Q61 residue) challenging for researchers . When designing experiments to study ARL16 function, researchers should consider these unique structural properties rather than applying approaches standardized for other ARF family members.
ARL16 expression appears to be ubiquitous across human cell lines tested, including HeLa, U937, HUVEC, HT1080, K562, 293T, and Huh7 cells, as demonstrated by Western blot analysis using specific antisera . This broad expression pattern suggests ARL16 performs fundamental cellular functions rather than tissue-specific roles. For researchers investigating ARL16 regulation, immunoblotting with validated antibodies provides the most reliable detection method at the protein level. When analyzing expression patterns, researchers should consider that endogenous ARL16 migrates as a slightly smaller protein (approximately 30 kDa) compared to Flag-tagged overexpressed versions . Additionally, researchers should be aware of the existence of different ARL16 isoforms - studies have identified both 173 and 197 residue forms of human ARL16 . When designing experiments to study ARL16 expression regulation, researchers should utilize primers or antibodies that can distinguish between these isoforms and validate findings across multiple cell types to establish general versus context-specific expression patterns.
Selecting the appropriate ARL16 antibody requires careful consideration of several key factors that will impact experimental outcomes. First, researchers must identify which applications they need the antibody for - currently available ARL16 antibodies are validated for different techniques including ELISA (typically at dilutions of 1:2000-1:80000), Western blotting (1:500-1:1000), and immunohistochemistry (1:20-1:200) . Second, species reactivity is crucial - while most ARL16 antibodies recognize human ARL16, only some cross-react with mouse and rat orthologs . The immunogen used to generate the antibody is another important consideration - most commercial ARL16 antibodies are raised against recombinant human ARL16 protein fragments (common immunogens include amino acids 1-197 or 19-197) . The host species and clonality affect applications and potential cross-reactivity issues - currently available options are predominantly rabbit polyclonal antibodies . For quantitative applications, researchers should select antibodies that have been affinity-purified rather than those purified by protein G alone, as they typically offer higher specificity . Finally, researchers should review validation data provided by manufacturers and, ideally, independent validation studies before selecting an antibody for their specific research questions.
Proper validation of ARL16 antibodies is essential for generating reliable research data. A comprehensive validation approach includes several complementary methods. First, Western blot analysis should confirm the antibody detects a single band of appropriate molecular weight (approximately 30 kDa for endogenous ARL16) . This validation should include positive controls (cell lines known to express ARL16, such as HeLa or 293T) and negative controls (either ARL16 knockout cells or siRNA-treated cells with confirmed ARL16 knockdown) . For immunofluorescence applications, researchers should compare staining patterns using different antibodies targeting distinct regions of ARL16 and verify localization through co-expression of tagged ARL16 constructs . For functional studies, researchers should confirm the ability of the antibody to immunoprecipitate native ARL16 and its known interacting partners, such as RIG-I . When validating antibodies for quantitative applications like ELISA, standard curves using recombinant ARL16 protein should be established to determine sensitivity and dynamic range. Finally, researchers should cross-validate findings using orthogonal methods that don't rely on antibodies, such as mass spectrometry or RNA sequencing, to confirm the specificity and sensitivity of antibody-based detection methods.
Commercial ARL16 antibodies exhibit significant variations in their properties and optimal applications, necessitating careful selection based on experimental requirements. The table below compares key characteristics of several commercially available ARL16 antibodies:
These antibodies differ in their immunogen specificities (full-length vs. partial protein), validated applications, and recommended working dilutions. Researchers should select the appropriate antibody based on their specific application needs, desired species reactivity, and experimental conditions. For studies requiring detection of specific ARL16 isoforms or functional domains, attention should be paid to the exact immunogen sequence used to generate the antibody. Additionally, researchers planning long-term studies should consider antibody stability under different storage conditions, with most manufacturers recommending aliquoting and storing at -20°C or below to avoid freeze-thaw cycles .
For optimal Western blotting results with ARL16 antibodies, researchers should follow a methodology tailored to the unique properties of this protein. Sample preparation is critical - cells should be lysed in a buffer containing appropriate detergents (such as 1% Triton X-100) and protease inhibitors to preserve ARL16 integrity. Given ARL16's expected molecular weight of approximately 30 kDa, researchers should use 12-15% SDS-PAGE gels for optimal resolution in this range . For protein transfer, a semi-dry transfer system with PVDF membranes typically provides better results than nitrocellulose for this protein. Blocking should be performed with 5% non-fat dry milk in TBST, as BSA-based blocking can produce higher background with some ARL16 antibodies. The primary antibody dilution should be optimized based on manufacturer recommendations, with common working dilutions ranging from 1:500 to 1:1000 for Western blotting applications . Overnight incubation at 4°C generally yields better signal-to-noise ratios than shorter incubations at room temperature. For detection, HRP-conjugated secondary antibodies followed by enhanced chemiluminescence provide suitable sensitivity. Researchers should include appropriate controls, including positive controls (cell lines known to express ARL16, such as HeLa or 293T) and negative controls (ARL16 knockdown samples). When analyzing data, researchers should be aware that endogenous ARL16 appears as a band slightly smaller than tagged overexpressed versions .
Successful immunohistochemistry (IHC) and immunofluorescence (IF) with ARL16 antibodies require specific protocol optimizations. For paraffin-embedded tissues, antigen retrieval is critical - a citrate buffer (pH 6.0) with heat-induced epitope retrieval typically provides optimal results, as demonstrated in human adrenal gland tissue studies . For IHC applications, researchers should begin with dilutions between 1:20 and 1:200, with 1:100 serving as a good starting point for optimization . For IF in cultured cells, fixation method significantly impacts results - 4% paraformaldehyde fixation for 15 minutes at room temperature preserves ARL16 epitopes while maintaining cellular architecture. When examining subcellular localization, co-staining with organelle markers is essential for accurate interpretation - HSP60 for mitochondrial localization and acetylated tubulin for ciliary structures have been successfully used in previous studies . Researchers should be aware that ARL16 displays different localization patterns depending on cell type - diffuse cytoplasmic staining in 293T and HeLa cells, but punctate patterns along ciliary axonemes in RPE1 cells . Controls should include primary antibody omission, isotype controls, and ideally, ARL16 knockdown or knockout samples. For quantitative analysis of fluorescence intensity or localization patterns, appropriate image acquisition settings must be established using control samples to avoid saturation and ensure reproducibility across experimental conditions.
Investigating ARL16's protein interactions requires specialized techniques that account for its GTP-binding properties and multiple subcellular localizations. Co-immunoprecipitation (Co-IP) has been successfully used to detect interactions between ARL16 and RIG-I, revealing that ARL16 specifically interacts with the C-terminal domain (CTD) of RIG-I . When performing Co-IP experiments, researchers should use lysis buffers containing 1% NP-40 or Triton X-100 with protease inhibitors, avoiding harsh detergents that might disrupt protein-protein interactions. Given ARL16's GTP-dependent binding properties, adding exogenous GTP (0.1-1 mM) to lysis and wash buffers can help preserve nucleotide-dependent interactions . Proximity ligation assays (PLA) provide an alternative approach for detecting endogenous protein interactions in situ, offering spatial resolution that Co-IP lacks. For studying dynamics of ARL16 interactions, fluorescence resonance energy transfer (FRET) or bimolecular fluorescence complementation (BiFC) with fluorescently tagged constructs can reveal interaction kinetics in live cells. Pull-down assays using recombinant GST-tagged ARL16 can identify direct binding partners, but researchers must consider that ARL16's unique G-3 motif (RELGGC instead of WDXGGQ) affects its GTP hydrolysis mechanism . When designing these experiments, preloading ARL16 with non-hydrolyzable GTP analogs (such as GTPγS) or creating GTP-locked mutants can help stabilize specific interaction states. Researchers should also consider that ARL16's interactions may differ based on its subcellular localization, necessitating compartment-specific interaction studies.
ARL16's function as a negative regulator of RIG-I signaling has significant implications for innate immunity research design. Studies have demonstrated that ARL16 binds to the C-terminal domain (CTD) of RIG-I, suppressing the association between RIG-I and RNA, thereby inhibiting downstream type I interferon production . When investigating innate immune responses to RNA viruses, researchers should consider ARL16 expression levels as a potential variable affecting experimental outcomes. Overexpression of ARL16 inhibits RIG-I-mediated signaling and antiviral activity, while knockdown of endogenous ARL16 potentiates Sendai virus-induced IFN-β expression and affects vesicular stomatitis virus replication . For gain-of-function experiments, researchers should note that ARL16's homologous proteins (ARL1 and ARF1) do not exhibit the same inhibitory effect on RIG-I signaling, making ARL16 specifically relevant to this pathway . When designing loss-of-function studies, RNA interference approaches targeting ARL16 have successfully demonstrated enhanced IFN-β production in response to viral infection . For mechanistic investigations, researchers should consider that ARL16's inhibitory function is GTP-dependent - mutants restricted to the GTP-disassociated form (ARL16 T37N and ARL16Δ45–54) fail to interact with RIG-I and lose their inhibitory function . Time-course experiments are critical, as endogenous ARL16 changes its GTP-binding status upon viral infection, dynamically regulating its interaction with RIG-I. These considerations necessitate careful experimental design, including appropriate controls for ARL16 expression and activity when studying innate immune responses to viral infection.
Investigating ARL16's GTP-dependent functions requires specialized approaches that address its unique structural and biochemical properties. Unlike most GTPases, ARL16 contains an altered G-3 motif (RELGGC instead of the conserved WDXGGQ), suggesting an alternative GTP hydrolysis mechanism and complicating traditional mutational approaches . Researchers can employ several complementary techniques to study these functions. Biochemical assays using purified recombinant ARL16 can measure GTP binding (using fluorescent GTP analogs like mant-GTP) and hydrolysis rates (using [γ-32P]GTP and thin-layer chromatography). For cellular studies, researchers can generate mutations in the GTP-binding pocket - the T37N mutation has been shown to restrict ARL16 to the GTP-disassociated form . Pull-down assays using GTP-agarose can isolate the active GTP-bound form of ARL16 from cell lysates. To study dynamic changes in ARL16's GTP-binding status upon viral infection or other stimuli, researchers can use conformation-specific antibodies that recognize the GTP-bound versus GDP-bound states, though these may need to be custom-developed given ARL16's unique properties. Fluorescence-based sensors incorporating ARL16 between fluorescent proteins can detect conformational changes associated with nucleotide binding in live cells. When interpreting results, researchers should consider that GTP-dependent functions of ARL16 may vary across subcellular compartments, necessitating spatial resolution in experimental readouts. Additionally, the finding that ARL16 changes its GTP-binding status upon Sendai virus infection highlights the importance of studying these functions under physiologically relevant conditions .
Understanding ARL16's evolutionary history provides valuable insights into its fundamental biological functions. The ARF family of regulatory GTPases is ancient, with 16 members predicted to have been present in the last eukaryotic common ancestor . To investigate ARL16's phylogenetic conservation, researchers should employ comparative genomics approaches analyzing sequence data across diverse taxonomic groups. Current data indicate human ARL16 shares 86.5% sequence identity at the amino acid level with its mouse ortholog and 61.1% with its zebrafish ortholog, suggesting strong evolutionary conservation across vertebrates . For comprehensive phylogenetic analysis, researchers should use multiple sequence alignment tools (such as MUSCLE or CLUSTAL) followed by maximum likelihood or Bayesian inference methods to construct phylogenetic trees. When analyzing sequence conservation, special attention should be paid to functional domains, particularly the GTP-binding regions and the unique G-3 motif (RELGGC), which differs from the canonical WDXGGQ motif found in most GTPases . Structural modeling using homology-based approaches can predict three-dimensional conservation despite sequence divergence. Functional conservation can be assessed through complementation studies, expressing orthologs from different species in model systems with ARL16 depletion to test functional rescue. Researchers should also analyze synteny (conservation of gene order on chromosomes) across species, which can provide insights into evolutionary constraints and gene duplication events. Additionally, selection analysis using dN/dS ratios can identify regions under purifying or positive selection, highlighting functionally critical domains. These evolutionary approaches can reveal the core ancestral functions of ARL16 versus more recently evolved roles, informing experimental design and interpretation across model systems.
Antibody specificity issues present significant challenges in ARL16 research and require systematic troubleshooting approaches. When Western blotting reveals multiple bands, researchers should first determine whether these represent genuine isoforms (such as the 173 and 197 residue forms of human ARL16) or non-specific binding. Validation strategies include comparing patterns across different antibodies targeting distinct ARL16 epitopes and performing knockdown/knockout experiments to confirm band disappearance. For antibodies showing high background in immunofluorescence, optimization of blocking conditions is essential - BSA-based blocking solutions (3-5%) often yield cleaner results than milk-based blockers for this application. Titrating primary antibody concentrations and extending wash steps can further improve signal-to-noise ratios. Cross-reactivity with related ARF family proteins is a particular concern given sequence similarities (ARL16 shares 25-28% identity with ARF1 and ARL1) . To address this, researchers can perform parallel experiments in cells overexpressing these related proteins to identify potential cross-reactivity. Peptide competition assays using the specific immunogen peptide can confirm binding specificity - signal elimination after antibody pre-incubation with the immunizing peptide indicates specific binding. For quantitative applications, researchers should establish standard curves using recombinant ARL16 protein to determine detection limits and linear range. Additionally, researchers should be aware that fixation methods significantly impact epitope accessibility - comparing results across multiple fixation protocols (paraformaldehyde, methanol, or acetone) can help identify optimal conditions for specific antibodies.
Rigorous experimental controls are critical for generating reliable data in ARL16 research. For expression studies, positive controls should include cell lines with confirmed ARL16 expression (HeLa, U937, HUVEC, HT1080, K562, 293T, and Huh7 cells have all demonstrated detectable ARL16 levels) . Negative controls should include ARL16 knockdown (using validated siRNAs) or knockout samples. When overexpressing ARL16, comparing wild-type to functionally relevant mutants provides important controls - the T37N mutation and ARL16Δ45–54 deletion, which prevent GTP binding, serve as functional negative controls . For interaction studies, researchers should include controls for non-specific binding (such as IgG or unrelated proteins of similar size and charge) and validate interactions using reciprocal co-immunoprecipitation approaches. When studying ARL16's inhibitory effect on RIG-I signaling, parallel experiments with ARL1 and ARF1 provide important negative controls, as these homologous proteins do not inhibit RIG-I-mediated downstream signaling . For localization studies, co-expression of fluorescently tagged ARL16 alongside immunofluorescence detection provides validation of antibody specificity. When examining ARL16's role in viral infection responses, time-course experiments with appropriate viral and non-viral stimuli are essential to distinguish specific effects from general cellular stress responses. Additionally, researchers should implement loading controls for Western blotting (such as GAPDH or β-actin) and housekeeping gene controls for qRT-PCR to ensure equal loading and consistent reference for quantification. These comprehensive controls enable reliable interpretation of experimental results and minimize the risk of artifacts or misinterpretation.
Conflicting reports on ARL16 subcellular localization require careful methodological approaches to resolve. Published studies have variously reported ARL16 in the cytoplasm, along ciliary axonemes, and in mitochondria , suggesting either context-dependent localization or methodological variables affecting detection. To address these discrepancies, researchers should implement multiple complementary techniques. First, comparing subcellular fractionation results with imaging-based localization provides orthogonal validation - biochemical isolation of organelles followed by Western blotting for ARL16 can confirm microscopy findings. Super-resolution microscopy techniques (STED, STORM, or PALM) offer improved spatial resolution over conventional confocal microscopy, potentially clarifying ambiguous localization patterns. Live-cell imaging with fluorescently tagged ARL16 can reveal dynamic localization changes under different physiological conditions that may be missed in fixed-cell analyses. When interpreting localization data, researchers should consider cell type-specific differences - ARL16 shows diffuse cytoplasmic staining in 293T and HeLa cells, but punctate patterns along ciliary axonemes in RPE1 cells . Cell cycle phase and differentiation state may also influence localization patterns and should be controlled for in experimental design. Co-localization studies with multiple markers for each suspected compartment (e.g., different mitochondrial proteins beyond HSP60) provide more robust evidence of specific localization. Additionally, researchers should examine whether different ARL16 isoforms (173 vs. 197 residue forms) show distinct localization patterns . Functional approaches, such as proximity-based labeling techniques (BioID or APEX), can identify proteins in close proximity to ARL16 in living cells, providing functional validation of localization. By systematically implementing these approaches, researchers can determine whether conflicting localization data reflect biological complexity or methodological limitations.