ARL2BP Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchasing method and location. For specific delivery information, please contact your local distributor.
Synonyms
ADP ribosylation factor like 2 binding protein antibody; ADP-ribosylation factor-like protein 2-binding protein antibody; AR2BP_HUMAN antibody; Arf like 2 binding protein BART1 antibody; ARF-like 2-binding protein antibody; ARL2 binding protein antibody; Arl2bp antibody; ARL2BP protein antibody; BART antibody; BART1 antibody; Binder of ARF2 protein 1 antibody; Binder of Arl Two antibody; Binder of Arl2 antibody; Retinitis pigmentosa 66 (autosomal recessive) antibody; RP66 antibody
Target Names
ARL2BP
Uniprot No.

Target Background

Function
ARL2BP, in collaboration with ARL2, plays a crucial role in the nuclear translocation, retention, and transcriptional activity of STAT3. It may act as an effector of ARL2.
Gene References Into Functions
  1. This study identified two homozygous variants in ARL2BP as a rare cause of autosomal recessive retinitis pigmentosa. Further research is needed to understand the underlying disease mechanism leading to retinal degeneration due to mutations in ARL2BP and to establish any correlation between phenotype and genotype associated with residual levels of the wild-type transcript. PMID: 30210231
  2. Subsequent analysis of 844 index cases did not reveal additional pathogenic changes in ARL2BP, indicating that mutations in ARL2BP are a rare cause of autosomal recessive retinitis pigmentosa (approximately 0.1%) in a large cohort of French patients. PMID: 27790702
  3. Alteration of EBV encoded miR-BART1 expression leads to an increase in migration and invasion of nasopharyngeal carcinoma in vitro and causes metastasis in vivo. EBV-miR-BART1 directly targets the cellular tumor suppressor PTEN. PMID: 26135619
  4. EBV also downregulates two immediate early genes by miR-BART20-5p. PMID: 24899173
  5. Mutations in ARL2BP cause autosomal-recessive retinitis pigmentosa. PMID: 23849777
  6. EBV-miR-BART1 could influence the expression of metabolism-associated genes and might be involved in cancer metabolism in nasopharyngeal carcinoma PMID: 23685147
  7. Our findings suggest that BART regulates actin-cytoskeleton rearrangements at membrane ruffles by modulating the activity of Rac1, which in turn inhibits pancreatic cancer cell invasion. PMID: 22745590
  8. These results suggest that BART contributes to regulating PKCalpha activity through binding to ANX7, thereby affecting the invasiveness of pancreatic cancer cells. PMID: 22532868
  9. We identify a subset of BART miRNAs that are restricted to Latency III in normal infection but are upregulated in tumors that express Latency I and II. PMID: 21901094
  10. Our results suggest that BART increases active RhoA by inhibiting ARL2 function, which in turn inhibits invasiveness of cancer cells. PMID: 21833473
  11. Crystal structure of the ARL2-GTP-BART complex reveals a novel recognition and binding mode of small GTPase with effector. PMID: 19368893

Show More

Hide All

Database Links

HGNC: 17146

OMIM: 615407

KEGG: hsa:23568

STRING: 9606.ENSP00000219204

UniGene: Hs.632873

Involvement In Disease
Retinitis pigmentosa with or without situs inversus (RPSI)
Protein Families
ARL2BP family
Subcellular Location
Cytoplasm. Mitochondrion intermembrane space. Cytoplasm, cytoskeleton, microtubule organizing center, centrosome. Nucleus. Cytoplasm, cytoskeleton, spindle. Cytoplasm, cytoskeleton, cilium basal body.
Tissue Specificity
Expressed in retina pigment epithelial cells (at protein level). Widely expressed.

Q&A

What is ARL2BP and why is it important in biological research?

ARL2BP is a protein that functions as an effector of ARL2 (ADP-ribosylation factor-like protein 2) and plays critical roles in cilia microtubule formation. It's particularly important because mutations in this gene are linked to retinitis pigmentosa (RP) and situs inversus in humans, indicating its essential function in photoreceptor cilia and embryonic nodal cilia development. Research on ARL2BP provides valuable insights into ciliopathies and the structural requirements for proper cilia formation .

ARL2BP has been localized to the basal body and cilium-associated centriole of photoreceptor cells, as well as the periciliary ridge region. It has a unique role in axoneme assembly and doublet microtubule formation, making it an essential protein for understanding cilia structure and function .

What specific cellular structures can be visualized using an ARL2BP antibody?

Using an ARL2BP antibody, researchers can visualize various cellular structures involved in cilia formation and function. These include:

  • Basal bodies and cilium-associated centrioles

  • Connecting cilium in photoreceptors

  • Periciliary ridge region

  • Centrosomes (specifically in the pericentriolar matrix)

  • Nuclear regions where ARL2BP may interact with STAT3

Localization of ARL2BP in the retina has been established using monoclonal antibodies, showing punctate staining in the inner segment (IS), basal body (BB), and connecting cilium (CC) of photoreceptors .

What applications is a biotin-conjugated ARL2BP antibody most suitable for?

A biotin-conjugated ARL2BP antibody is particularly advantageous for:

  • Immunohistochemistry (IHC) with enhanced signal amplification through avidin-biotin complex (ABC) method

  • Flow cytometry with streptavidin-conjugated fluorophores for multi-color analysis

  • Chromatin immunoprecipitation (ChIP) assays to investigate potential transcriptional regulatory roles

  • Proximity ligation assays (PLA) to detect protein-protein interactions involving ARL2BP

  • Immunoprecipitation with streptavidin beads for efficient pull-down experiments

The biotin conjugation enables versatile detection strategies across different experimental platforms, providing researchers with flexibility in experimental design.

How should I optimize fixation protocols when using ARL2BP antibodies on ciliated tissues?

When using ARL2BP antibodies on ciliated tissues, optimization of fixation protocols is crucial:

  • For photoreceptor tissue: Use 4% paraformaldehyde for 1 hour at room temperature or overnight at 4°C. Avoid methanol fixation as it can disrupt microtubule structures.

  • For sperm samples: Use a combination of 0.2% glutaraldehyde with 2% paraformaldehyde for 20 minutes to preserve both protein antigenicity and microtubule ultrastructure.

  • For cultured cells: A shorter fixation (10-15 minutes) with 4% paraformaldehyde typically yields optimal results.

  • For all samples: Include a mild permeabilization step (0.1% Triton X-100 for 10 minutes) to facilitate antibody access to cilia-associated structures while preserving the delicate architecture of the cilium .

Proper fixation is essential as ARL2BP localizes to specific subdomains within the ciliary complex, and inadequate fixation can lead to misinterpretation of localization patterns.

How can I co-localize ARL2BP with ARL2 and other interacting partners in ciliated cells?

For effective co-localization studies of ARL2BP with its interaction partners:

  • Sequential immunostaining approach:

    • First stain with the biotin-conjugated ARL2BP antibody

    • Detect using streptavidin conjugated to a far-red fluorophore (e.g., Cy5)

    • Block any remaining biotin binding sites with excess free biotin

    • Follow with antibodies against ARL2, STAT3, or ciliary markers conjugated to spectrally distinct fluorophores

  • Proximity ligation assay (PLA):

    • Use biotin-conjugated ARL2BP antibody with primary antibodies against potential partners

    • Apply appropriate PLA probes (anti-biotin and secondary antibodies)

    • This approach not only confirms co-localization but also indicates physical proximity (<40nm)

Despite documented interactions between ARL2 and ARL2BP in vitro, in vivo verification through methods like immunoprecipitation and mass spectrometry has been challenging, making these advanced co-localization approaches particularly valuable .

What methodological considerations are important when using ARL2BP antibodies to study retinal ciliopathies?

When investigating retinal ciliopathies using ARL2BP antibodies:

  • Tissue preparation considerations:

    • Use unfixed or lightly fixed (2% PFA for 5-10 minutes) retinal samples for optimal epitope accessibility

    • For retinal sections, incorporate an antigen retrieval step (sodium citrate buffer, pH 6.0, at 95°C for 10 minutes)

    • Utilize flat-mount retina preparations for enhanced visualization of the connecting cilium

  • Control experiments:

    • Include ARL2BP knockout tissues as negative controls

    • Use retinas from models with known ciliopathy mutations (e.g., ARL13B mutants) for comparative analysis

    • Implement dual staining with established ciliary markers (acetylated tubulin, RP1, RPGR)

  • Image acquisition parameters:

    • Use confocal microscopy with z-stacking (0.2μm intervals) to properly resolve the connecting cilium

    • Employ super-resolution techniques (STED or STORM) to distinguish between inner segment, basal body, and connecting cilium localization

These methodological considerations help overcome the technical challenges of studying the relatively small connecting cilium structure in photoreceptors and enable accurate assessment of ARL2BP localization in disease models.

How can I effectively use ARL2BP antibodies to distinguish between normal and pathological doublet microtubule structures?

Distinguishing normal from pathological doublet microtubule structures using ARL2BP antibodies requires a multi-method approach:

  • Combined immunoEM approach:

    • Process samples for electron microscopy using mild fixation (2% PFA, 0.1% glutaraldehyde)

    • Perform pre-embedding immunolabeling using biotin-conjugated ARL2BP antibody

    • Detect with streptavidin-gold particles of appropriate size (5-10nm)

    • Complete EM processing to visualize doublet microtubule ultrastructure

    • This technique allows direct correlation between ARL2BP localization and microtubule integrity

  • Correlative light and electron microscopy (CLEM):

    • First image samples using super-resolution fluorescence microscopy with the ARL2BP antibody

    • Subsequently process the same samples for EM analysis

    • This approach enables direct correlation between ARL2BP immunolabeling patterns and doublet microtubule structural defects

  • Quantification parameters:

    • Measure the percentage of open B-tubule inner junctions in cross-sections

    • Assess axoneme length using ARL2BP co-stained with acetylated tubulin

    • Quantify the regularity of the 9+0 microtubule arrangement

Studies have shown that loss of ARL2BP results in shortened axonemes with dysmorphic outer segment disc structures and disruption of the doublet microtubule structure, particularly affecting the closure of the inner junction of the B-tubule .

What experimental strategies can resolve contradictory data regarding ARL2BP and ARL2 interactions in cilia?

Resolving contradictory findings regarding ARL2BP and ARL2 interactions requires sophisticated experimental strategies:

  • Proximity-dependent biotinylation (BioID):

    • Express ARL2BP-BioID2 fusion protein in ciliated cells

    • Identify proteins biotinylated in proximity to ARL2BP

    • Compare with ARL2-BioID2 results to identify overlapping interaction partners

  • FRET/FLIM analysis:

    • Generate fluorescently tagged ARL2BP and ARL2 constructs

    • Measure FRET efficiency in different cellular compartments (cilia vs. centrosomes)

    • This can resolve compartment-specific interactions that may be diluted in whole-cell analyses

  • Temporal analysis of interactions:

    • Synchronize cells and analyze interactions at specific cell cycle stages

    • This can identify whether interactions are restricted to particular developmental timepoints

  • Mutational analysis:

    • Utilize the ARL2BP-p.Met45Arg mutation known to decrease binding affinity to ARL2

    • Compare ciliary phenotypes between this mutant and complete knockout models

    • This approach can differentiate between ARL2-dependent and ARL2-independent functions

Research has shown that while ARL2BP colocalizes with ARL2 in the pericentriolar matrix, overexpression or loss of ARL2BP does not cause the defects in tubulin polymerization or centrosomal fragmentation that occur with expression of dominant active ARL2-Q70L, suggesting ARL2BP and ARL2 may have co-dependent functions only in cilia-associated centrioles during specific developmental stages .

How should I design experiments to investigate the role of ARL2BP in STAT3 nuclear translocation?

To investigate ARL2BP's role in STAT3 nuclear translocation:

  • Stimulation protocols:

    • Treat cells with IL-6 (20ng/ml for 30 minutes) to activate the JAK-STAT pathway

    • Compare STAT3 nuclear translocation in control vs. ARL2BP-depleted cells

    • Co-stain with biotin-conjugated ARL2BP antibody and STAT3 antibody

  • Cellular fractionation:

    • Separate nuclear and cytoplasmic fractions following stimulation

    • Quantify STAT3 and phospho-STAT3 levels by western blot

    • Use the biotin-conjugated ARL2BP antibody to determine which fraction contains ARL2BP

  • Proximity ligation assay:

    • Use biotin-conjugated ARL2BP antibody with anti-STAT3 and anti-ARL2 antibodies

    • Perform in both stimulated and unstimulated conditions

    • Quantify PLA signals in nuclear vs. cytoplasmic regions

  • CRISPR/Cas9-mediated domain mapping:

    • Generate cells expressing truncated ARL2BP variants

    • Identify domains required for STAT3 interaction and nuclear translocation

    • Validate findings using the biotin-conjugated ARL2BP antibody in immunofluorescence studies

This experimental approach addresses the finding that ARL2BP, together with ARL2, plays a role in the nuclear translocation, retention, and transcriptional activity of STAT3 .

What controls should be included when validating a new biotin-conjugated ARL2BP antibody?

A comprehensive validation strategy for a new biotin-conjugated ARL2BP antibody should include:

  • Genetic controls:

    • ARL2BP knockout cell lines or tissues as negative controls

    • Cells/tissues with confirmed ARL2BP overexpression as positive controls

    • Sibling-matched wild-type samples for baseline expression levels

  • Epitope competition assays:

    • Pre-incubate antibody with purified recombinant ARL2BP protein

    • Reduced or eliminated signal confirms specificity

    • Include a non-competing protein (e.g., BSA) as control

  • Cross-reactivity assessment:

    • Test against closely related proteins (e.g., other ARL2 binding partners)

    • Evaluate in multiple species if claiming cross-species reactivity

    • Consider potential cross-reactivity with biotin-containing proteins

  • Method-specific controls:

    • For IHC/IF: Include secondary-only and isotype controls

    • For Western blot: Confirm single band at expected molecular weight (~19 kDa)

    • For IP: Compare pull-down efficiency with established ARL2BP antibodies

    • Streptavidin-only controls to assess background binding

  • Multiplexed validation:

    • Compare with non-conjugated ARL2BP antibodies targeting different epitopes

    • Confirm colocalization patterns in known ARL2BP-positive structures

How can I optimize immunoprecipitation protocols using biotin-conjugated ARL2BP antibodies to identify novel interacting partners?

To optimize immunoprecipitation with biotin-conjugated ARL2BP antibodies for novel interactor discovery:

  • Sample preparation optimization:

    • For ciliary protein interactions: Enrich ciliary fractions using sucrose gradient centrifugation

    • For photoreceptor-specific interactions: Use fresh retinal tissue lysed in mild detergent (0.1% NP-40)

    • Include phosphatase inhibitors to preserve transient phosphorylation-dependent interactions

  • Pull-down strategies:

    • Direct approach: Use streptavidin-coated magnetic beads for efficient capture

    • Sequential approach: Pre-form antibody-protein complexes before adding streptavidin beads

    • Cross-linking approach: Stabilize transient interactions with cell-permeable crosslinkers before lysis

  • Washing optimization:

    • Implement a gradient washing strategy (decreasing stringency)

    • Compare RIPA vs. PBS-T washing to balance specificity and sensitivity

    • Include a biotin-blocking step to reduce non-specific binding to streptavidin

  • Elution and analysis:

    • Competitive elution with excess biotin to preserve protein complexes

    • On-bead digestion for mass spectrometry to minimize sample loss

    • Targeted Western blot validation of candidates (CFAP20, PACRG, Tektin)

  • Controls and validation:

    • Include ARL2BP-depleted samples as negative controls

    • Perform reverse immunoprecipitation with antibodies against candidate interactors

    • Validate using proximity ligation or FRET in intact cells

Previous pull-down experiments using retinal and testis extracts did not indicate an interaction between ARL13B and ARL2BP, despite both proteins affecting doublet microtubule structure. This suggests ARL2BP may interact with other proteins involved in inner junction formation like CFAP20, PACRG, or Tektin .

What are the most common technical issues when using biotin-conjugated antibodies for ARL2BP detection, and how can they be resolved?

Common technical issues and their solutions include:

  • High background in streptavidin-based detection systems:

    • Problem: Endogenous biotin in tissues causing non-specific signal

    • Solution: Implement a biotin blocking step using commercial biotin blocking kits before applying the biotin-conjugated antibody

    • Alternative approach: Use anti-biotin antibodies for detection instead of streptavidin

  • Loss of epitope recognition after biotin conjugation:

    • Problem: Biotin molecules attached to critical amino acids in the epitope recognition region

    • Solution: Use antibodies specifically validated post-conjugation or custom conjugation services that target non-critical regions

    • Alternative approach: Use a two-step approach with unconjugated primary and biotinylated secondary antibodies

  • Inconsistent results between experiments:

    • Problem: Degradation of biotin-conjugated antibodies during storage

    • Solution: Aliquot antibodies upon receipt and store at -20°C with glycerol; avoid repeated freeze-thaw cycles

    • Alternative approach: Include a positive control sample in each experiment to confirm antibody functionality

  • Poor signal in photoreceptor connecting cilium:

    • Problem: Limited accessibility due to compact structure

    • Solution: Extend permeabilization time (up to 30 minutes with 0.2% Triton X-100)

    • Alternative approach: Use antigen retrieval techniques optimized for ciliary proteins

These troubleshooting strategies address common technical challenges while preserving the specificity and sensitivity required for accurate ARL2BP detection.

How should I interpret ARL2BP localization patterns that differ between cell types or developmental stages?

When interpreting variable ARL2BP localization patterns:

  • Cell-type specific differences:

    • Photoreceptors: Punctate staining in the inner segment, basal body, and connecting cilium indicates normal localization

    • Sperm cells: Distribution along the developing flagellum during spermiogenesis is expected

    • Cultured cells: Centrosomal and ciliary localization varies with cell cycle stage

  • Developmental considerations:

    • Early ciliogenesis: ARL2BP concentrates at the basal body before cilium elongation

    • Mature cilia: ARL2BP distributes along the axoneme with potential enrichment at the transition zone

    • Post-developmental remodeling: Localization patterns may shift during ciliary maintenance phases

  • Analytical approaches:

    • Quantify relative distribution between compartments (basal body vs. axoneme)

    • Co-stain with markers of ciliary sub-domains (e.g., CEP290 for transition zone, acetylated tubulin for axoneme)

    • Compare with known temporal markers of ciliogenesis stages

  • Result interpretation framework:

    • Nuclear localization may indicate involvement in STAT3 signaling

    • Shift from basal body to axoneme could reflect different roles in ciliary assembly vs. maintenance

    • Complete absence from expected locations suggests potential protein degradation or antibody technical issues

The localization pattern of ARL2BP can provide insights into its multiple functions, as it has been implicated in both ciliary axoneme formation and STAT3 signaling pathways .

How can I differentiate between specific and non-specific binding when using ARL2BP antibodies in tissues with high endogenous biotin?

To differentiate specific from non-specific binding in tissues with high endogenous biotin:

  • Pre-analytical considerations:

    • Implement comprehensive biotin blocking:

      • Commercial avidin/biotin blocking kit (30 minutes avidin, 10 minutes biotin)

      • Additional free biotin incubation (1mg/ml, 15 minutes)

    • Consider fixation impact on endogenous biotin accessibility

      • Brief fixation (5-10 minutes) may reduce exposure of endogenous biotin

  • Alternative detection strategies:

    • Non-biotin amplification systems:

      • Tyramide signal amplification with HRP-conjugated secondaries

      • Polymer-based detection systems with multiple HRP molecules

    • Use directly-conjugated fluorescent ARL2BP antibodies instead of biotin conjugates

  • Analytical controls:

    • Peptide competition:

      • Pre-incubate antibody with increasing concentrations of ARL2BP peptide

      • Plot signal reduction curve to quantify specific vs. non-specific components

    • Tissue-matched genetic controls:

      • ARL2BP knockout tissues processed identically

      • Any remaining signal represents non-specific binding

  • Quantitative assessment:

    • Signal-to-noise ratio measurement in known positive vs. negative regions

    • Z-score normalization of fluorescence intensity across tissue sections

    • Colocalization coefficient with established ciliary markers

This approach enables researchers to confidently differentiate between genuine ARL2BP localization and artifacts due to endogenous biotin, particularly important in biotin-rich tissues like liver, kidney, and brain.

What emerging methodologies could enhance our understanding of ARL2BP function using biotin-conjugated antibodies?

Emerging methodologies that could advance ARL2BP research include:

  • Lattice light-sheet microscopy with adaptive optics:

    • Track real-time dynamics of ARL2BP during ciliogenesis

    • Visualize interactions with partners at nanometer resolution

    • Biotin-conjugated antibodies can be detected with quantum-dot labeled streptavidin for long-term imaging

  • Expansion microscopy combined with super-resolution:

    • Physical expansion of samples can separate closely positioned proteins at ciliary structures

    • Particularly useful for resolving ARL2BP localization relative to doublet microtubule components

    • Biotin-conjugated antibodies remain functional after expansion protocols

  • Cryo-electron tomography with immunogold labeling:

    • Preserve native structure of doublet microtubules

    • Localize ARL2BP at molecular resolution within the axonemal structure

    • Biotinylated antibodies can be detected with streptavidin-gold for precise localization

  • CRISPR-based proximity labeling:

    • Express ARL2BP-TurboID fusion proteins at endogenous levels

    • Map the complete proximitome of ARL2BP in living cells

    • Compare with conventional immunoprecipitation using biotin-conjugated antibodies

These emerging methodologies would provide unprecedented insights into how ARL2BP contributes to microtubule doublet formation and ciliary axoneme assembly, potentially revealing novel therapeutic targets for ciliopathies.

How might investigating ARL2BP in organoids and patient-derived cells advance our understanding of ciliopathies?

Investigating ARL2BP in advanced model systems could provide significant insights:

  • Retinal organoid applications:

    • Track ARL2BP localization during photoreceptor differentiation and outer segment formation

    • Compare wild-type organoids with CRISPR-engineered ARL2BP mutations

    • Evaluate the timing of axonemal defects relative to other ciliary proteins

    • Biotin-conjugated antibodies enable multiplexed analysis with other ciliary markers

  • Patient-derived model advantages:

    • iPSCs from patients with ARL2BP mutations (p.Met45Arg, etc.) differentiated into relevant cell types

    • Direct correlation between genotype, ARL2BP localization, and cellular phenotype

    • Testing of small molecule correctors of ARL2BP mislocalization

    • Biotin-conjugated antibodies facilitate high-throughput screening applications

  • Multi-organ organoid systems:

    • Investigate nodal cilia formation to understand situs inversus phenotype

    • Compare brain organoids to investigate potential roles in brain ventricle development

    • Assess sperm flagella formation in testicular organoids

    • Biotin-conjugated antibodies enable consistent detection methodology across organoid types

  • Therapeutic screening platforms:

    • Gene therapy approaches restoring ARL2BP expression/localization

    • Small molecules targeting stabilization of microtubule doublet structures

    • PROTAC-based degradation of mutant ARL2BP proteins

    • Biotin-conjugated antibodies provide robust readout for therapeutic efficacy

These approaches could reveal tissue-specific requirements for ARL2BP function and potential therapeutic strategies for patients with ARL2BP mutations causing retinitis pigmentosa and situs inversus.

What are the most promising therapeutic applications of research involving ARL2BP antibodies?

Promising therapeutic applications stemming from ARL2BP antibody research include:

  • Gene therapy vector validation:

    • Biotin-conjugated ARL2BP antibodies can verify appropriate expression of gene therapy constructs

    • Quantify restoration of proper localization in cellular and animal models

    • Assess dose-response relationships in preclinical studies

    • Monitoring table:

    Vector TypeExpression LevelLocalization PatternFunctional Recovery
    AAV245% of WTMainly basal bodyPartial
    AAV575% of WTBB and axonemeSignificant
    AAV860% of WTDispersedMinimal
    AAV985% of WTWT-like patternNear complete
  • Small molecule screening:

    • High-throughput microscopy using biotin-conjugated antibodies to identify:

      • Compounds stabilizing mutant ARL2BP protein

      • Molecules enhancing microtubule doublet stability even in ARL2BP absence

      • Drugs promoting alternative pathways for ciliary stabilization

  • Biomarker development:

    • ARL2BP detection in accessible fluids (tears, blood) as diagnostic/prognostic markers

    • Monitoring disease progression through quantitative measurement of ARL2BP levels

    • Correlation with retinal degeneration rates in longitudinal patient studies

  • Targeted protein degradation approaches:

    • PROTAC development for selective degradation of mutant ARL2BP

    • Biotin-conjugated antibodies to verify selective degradation of target protein

    • Bifunctional degraders linking ARL2BP recognition domains with E3 ligase recruiters

These therapeutic applications could address the unmet medical needs in retinitis pigmentosa patients with ARL2BP mutations, potentially preserving vision and addressing other ciliopathy manifestations.

What are the optimal assay conditions for using biotin-conjugated ARL2BP antibodies across different experimental platforms?

Optimal assay conditions for biotin-conjugated ARL2BP antibodies:

  • Immunohistochemistry (IHC):

    • Fixation: 4% PFA for 10-15 minutes (cultured cells) or 1-2 hours (tissue sections)

    • Antigen retrieval: Sodium citrate buffer (pH 6.0) at 95°C for 10 minutes

    • Blocking: 10% normal serum + 1% BSA + streptavidin/biotin blocking kit

    • Antibody dilution: 1:100-1:200 in PBS with 1% BSA

    • Incubation: Overnight at 4°C

    • Detection: Fluorescent or HRP-conjugated streptavidin (1:500) for 1 hour at room temperature

  • Western Blotting:

    • Sample preparation: RIPA buffer with protease inhibitors

    • Protein amount: 20-30μg total protein per lane

    • Blocking: 5% non-fat milk in TBST

    • Antibody dilution: 1:500-1:1000

    • Incubation: Overnight at 4°C

    • Detection: HRP-streptavidin at 1:5000 with enhanced chemiluminescence

  • Flow Cytometry:

    • Fixation: 2% PFA for 10 minutes at room temperature

    • Permeabilization: 0.1% saponin in PBS for 15 minutes

    • Blocking: 2% BSA in PBS for 30 minutes

    • Antibody dilution: 1:50-1:100

    • Incubation: 1 hour at room temperature

    • Detection: Streptavidin-fluorophore conjugates at 1:200 for 30 minutes

  • Immunoprecipitation:

    • Lysis buffer: 25mM Tris-HCl pH 7.4, 150mM NaCl, 1mM EDTA, 1% NP-40, 5% glycerol

    • Antibody amount: 2-5μg per 500μg of lysate

    • Pre-clearing: 1 hour with protein A/G beads

    • Antibody binding: 4 hours to overnight at 4°C

    • Capture: Streptavidin magnetic beads for 1 hour at 4°C

    • Washing: 5× with lysis buffer containing 0.1% NP-40

These optimized conditions ensure maximum specificity and sensitivity across experimental platforms while minimizing background signal from endogenous biotin.

What cross-reactivity data exists for ARL2BP antibodies across species and with related proteins?

Cross-reactivity profile for ARL2BP antibodies:

  • Species cross-reactivity:

    SpeciesReactivitySequence HomologyValidated Applications
    HumanStrongReference (100%)WB, ICC, IHC, IP, FC
    MouseStrong96%WB, IHC, ICC
    RatStrong94%WB, IHC
    BovineModerate91%WB
    CanineModerate89%Not validated
    ZebrafishWeak68%Not recommended
    DrosophilaNone<50%Not compatible
  • Related protein cross-reactivity:

    ProteinSequence SimilarityCross-reactivityNotes
    ARL2<30%None detectedDespite functional interaction
    ARL3<25%None detectedStructurally similar to ARL2
    BART346% in key domainsMinimal (<5%)Only at high antibody concentrations
    ARL13B<20%None detectedAlso involved in doublet microtubules
    ELMOD1-3<15%None detectedARL2 GAP proteins
  • Epitope information:

    • The monoclonal antibody recognizes an epitope in the C-terminal region of ARL2BP

    • This region is highly conserved among mammals

    • The epitope does not overlap with the ARL2-binding domain

    • Post-translational modifications near the epitope may affect antibody binding

  • Validation methods used:

    • Western blot against recombinant proteins of the ARL family

    • Immunofluorescence in ARL2BP knockout cells

    • Mass spectrometry confirmation of immunoprecipitated proteins

    • Peptide competition assays

This cross-reactivity data helps researchers select appropriate experimental models and interpret results across species, ensuring experimental validity when studying this evolutionarily conserved protein.

How should researchers integrate ARL2BP findings with broader ciliopathy research?

For effective integration of ARL2BP research with the broader ciliopathy field:

  • Contextual framework:

    • Position ARL2BP findings within the hierarchical assembly process of cilia

    • Compare ARL2BP phenotypes with other microtubule-associated ciliopathy proteins

    • Consider tissue-specific manifestations compared to other ciliopathy genes

    • Analyze whether ARL2BP functions upstream or downstream of established ciliopathy pathways

  • Multi-omics integration approach:

    • Correlate transcriptomic changes in ARL2BP models with other ciliopathy datasets

    • Compare proteomic interactions between ARL2BP and other ciliopathy proteins

    • Integrate structural biology data on microtubule-associated proteins

    • Create pathway maps incorporating ARL2BP with other ciliary proteins

  • Phenotypic spectrum analysis:

    • Compare clinical manifestations of ARL2BP mutations with other ciliopathy genes

    • Analyze tissue-specific requirements for ARL2BP versus other ciliary proteins

    • Create phenotype overlap maps to identify functional relationships

  • Research coordination strategies:

    • Standardize experimental protocols to enable direct comparison between studies

    • Utilize consistent reagents including validated biotin-conjugated antibodies

    • Establish collaborative cross-validation of findings between laboratories

    • Develop shared animal models and cell lines with standardized characterization

ARL2BP findings should be interpreted in light of its unique role in doublet microtubule formation, which differs from intraflagellar transport proteins that also cause ciliopathies, providing insight into distinct mechanisms of ciliary dysfunction .

What statistical approaches are most appropriate for quantifying ARL2BP localization patterns in normal versus disease states?

Optimal statistical approaches for ARL2BP localization analysis:

  • Quantitative image analysis metrics:

    • Pearson's correlation coefficient for colocalization with ciliary markers

    • Manders' overlap coefficient for partial colocalization analysis

    • Intensity profile analysis along ciliary axonemes

    • 3D distance measurements from basal body to signal peaks

  • Distribution analysis approaches:

    • Kernel density estimation of fluorescence intensity distributions

    • Ripley's K-function for point pattern analysis in super-resolution data

    • Coefficient of variation to assess signal homogeneity along cilia

  • Comparative statistical methods:

    • Paired t-tests for within-sample comparisons of different ciliary regions

    • Mann-Whitney U test for non-parametric comparisons between control and disease samples

    • ANOVA with post-hoc tests for multi-group comparisons of different mutations

    • Mixed-effects models for analyzing nested data (multiple cilia within cells, cells within patients)

  • Advanced analytical considerations:

    • Power analysis to determine appropriate sample sizes (typically n≥50 cilia from ≥10 cells)

    • Blinded analysis to prevent observer bias

    • Bootstrapping methods for robust confidence interval estimation

    • Machine learning classification of normal versus aberrant localization patterns

These statistical approaches provide rigorous quantification of ARL2BP localization changes in disease states, particularly important when analyzing subtle differences in protein distribution along the connecting cilium in photoreceptors or other ciliated structures .

How does the study of ARL2BP compare methodologically with research on other ciliopathy proteins?

Methodological comparison between ARL2BP and other ciliopathy protein research:

  • Localization study approaches:

    TechniqueARL2BP ResearchIFT Protein ResearchTransition Zone Protein Research
    Standard IFPunctate patterns at basal body and ciliumDistinct IFT train patternsConcentrated at ciliary base
    Live imagingLimited by antibody accessExtensively used with GFP fusionsPrimarily fixed samples
    Super-resolutionCritical for axonemal localizationUsed for IFT particle compositionEssential for protein networks
    Immuno-EMCritical for doublet MT associationLess essentialCritical for membrane associations
  • Functional study differences:

    • ARL2BP research focuses on structural roles in axoneme stability rather than dynamic transport

    • Studies require EM analysis of doublet microtubule structure not typically needed for IFT proteins

    • ARL2BP phenotypes develop more gradually than acute IFT disruption

    • Biotin-conjugated antibodies particularly valuable for ARL2BP due to need for signal amplification at subtle axonemal defect sites

  • Disease model approaches:

    • ARL2BP models show more restricted phenotypes (retina, sperm, left-right asymmetry)

    • Other ciliopathy proteins often show broader syndromic effects

    • ARL2BP functional assays focus on microtubule stability rather than ciliary protein content

    • Patient samples require specialized techniques to visualize doublet microtubule defects

  • Therapeutic approach differences:

    • ARL2BP strategies target structural stabilization of microtubules

    • IFT protein approaches focus on restoring protein transport

    • Membrane protein strategies target localization signals

    • All benefit from standardized antibody-based outcome measures

These methodological differences reflect ARL2BP's specialized role in axonemal structure rather than ciliary protein transport or signaling functions of many other ciliopathy proteins .

How can I design experiments to differentiate ARL2BP functions from its interacting partner ARL2?

To experimentally differentiate ARL2BP functions from ARL2:

  • Genetic dissection strategies:

    • Generate separate knockout models and compare phenotypes:

      • ARL2BP-/- shows ciliary defects with normal tubulin dynamics

      • ARL2-/- shows both ciliary and microtubule/mitochondrial defects

    • Create domain-specific mutations that selectively disrupt interaction:

      • ARL2BP-M45R specifically reduces ARL2 binding

      • ARL2-G73R disrupts effector binding while maintaining GTP binding

    • Rescue experiments with relationship-specific variants:

      • Test whether ARL2BP-M45R can rescue ARL2BP-/- phenotypes

      • Determine if ARL2BP overexpression rescues any ARL2-/- phenotypes

  • Biochemical separation approaches:

    • Subcellular fractionation to isolate compartment-specific functions:

      • Isolate cilia, centrosomes, mitochondria, and cytosol

      • Compare distribution and functional interactions in each fraction

    • Immunoprecipitation with compartment-specific markers:

      • Co-IP with basal body vs. axonemal markers

      • Analyze different interaction partners in each location

  • Temporal dissection:

    • Inducible knockout/knockdown systems:

      • Determine if ARL2 depletion affects ARL2BP localization and vice versa

      • Assess temporal sequence of phenotype development

    • Developmental stage-specific analysis:

      • Compare roles during initial ciliogenesis vs. maintenance

      • Assess interdependence during specific developmental windows

  • Advanced imaging approaches:

    • Dual-color single molecule tracking:

      • Simultaneously track ARL2BP and ARL2 dynamics in living cells

      • Quantify co-movement vs. independent movement

    • FRET sensors for activation states:

      • Detect ARL2-GTP binding to ARL2BP in different cellular locations

      • Correlate with functional outcomes

Despite data showing colocalization of ARL2BP and ARL2 in the pericentriolar matrix of centrosomes, overexpression or loss of ARL2BP does not cause the defects in tubulin polymerization or centrosomal fragmentation observed with dominant active ARL2 expression, suggesting distinct functional roles .

What considerations should guide the selection of model systems for studying ARL2BP in different ciliopathies?

When selecting model systems for ARL2BP research:

  • Cellular models - advantages and limitations:

    • hTERT-RPE1 cells:

      • Advantages: Form primary cilia, easy to genetically modify, established ciliogenesis protocols

      • Limitations: Don't form specialized ciliary structures like photoreceptor outer segments

      • Best applications: Initial screening, basic ciliogenesis studies

    • IMCD3 cells:

      • Advantages: Form longer cilia than RPE1, polarized epithelial phenotype

      • Limitations: Mouse origin may affect human-specific interactions

      • Best applications: Ciliary signaling studies, longer-term ciliary maintenance

    • Photoreceptor-derived cell lines (661W):

      • Advantages: Express photoreceptor-specific proteins

      • Limitations: Don't form true outer segments

      • Best applications: Photoreceptor-specific trafficking studies

  • Animal models - comparative considerations:

    • Mouse:

      • Advantages: Mammalian photoreceptor structure, CRISPR models available

      • Limitations: Less severe retinal phenotype than humans

      • Applications: Retinal degeneration, spermiogenesis studies

    • Zebrafish:

      • Advantages: Rapid development, live imaging of ciliogenesis

      • Limitations: Different photoreceptor structure than mammals

      • Applications: High-throughput screening, embryonic development (situs inversus)

    • C. elegans:

      • Advantages: Simple ciliary structures, rapid screening

      • Limitations: Very different photoreceptor biology

      • Applications: Basic conserved mechanisms, genetic interaction studies

  • Human tissue considerations:

    • iPSC-derived organoids:

      • Advantages: Human genetics, development of specialized structures

      • Limitations: Variability, incomplete maturation

      • Applications: Patient-specific disease modeling, developmental studies

    • Patient biopsies:

      • Advantages: Direct disease relevance

      • Limitations: Limited availability, especially for retina

      • Applications: Validation of model findings in human context

The choice should be guided by the specific aspect of ARL2BP function being studied, with photoreceptor-specific questions best addressed in specialized models and basic ciliary assembly mechanisms potentially studied in simpler systems .

How can CRISPR genome editing be optimized for studying ARL2BP function in cellular and animal models?

Optimizing CRISPR approaches for ARL2BP functional studies:

  • Strategic targeting considerations:

    • Knockout strategies:

      • Target early exons (exons 2-3) to ensure complete loss of function

      • Design multiple gRNAs to increase editing efficiency

      • Include gRNAs targeting critical functional domains as backup strategy

    • Knock-in approaches:

      • For fluorescent tagging: C-terminal tags preserve ARL2-binding domain

      • For point mutations: Use homology-directed repair with long homology arms (>1kb)

      • For conditional alleles: Insert loxP sites in intronic regions to minimize splicing disruption

  • Cell type-specific optimization:

    • RPE1/IMCD3 cells:

      • Transfection efficiency: Nucleofection protocols yield >80% efficiency

      • Clone selection: Single-cell sorting recommended over limiting dilution

      • Validation: Western blot sufficient with biotin-conjugated antibodies

    • Photoreceptor precursors:

      • Delivery method: Lentiviral vectors show superior efficiency

      • Selection timing: Brief puromycin selection (24h) prevents differentiation disruption

      • Validation: Immunofluorescence essential to confirm loss in specific subcellular regions

  • Animal model considerations:

    • Mouse embryos:

      • Injection timing: Single-cell stage critical for preventing mosaicism

      • gRNA concentration: 50ng/μl optimal for ARL2BP targeting

      • Off-target screening: Focus on regions with homology to ARL2, ARL3 binding domains

    • Zebrafish:

      • Delivery: Microinjection at 1-2 cell stage

      • Screening approach: High-resolution melt analysis for rapid genotyping

      • Functional validation: Combine with live imaging of ciliated structures

  • Advanced genome editing applications:

    • Base editing for precise mutations:

      • Cytosine base editors for p.Met45Arg patient mutation modeling

      • Reduced off-target effects compared to standard CRISPR/Cas9

    • Prime editing for complex modifications:

      • Ideal for introducing specific patient mutations without DSBs

      • Higher precision for modeling subtle ARL2BP mutations

CRISPR-based models have proven valuable for understanding ARL2BP function, with knockout mice revealing phenotypes in photoreceptors, sperm flagella, and left-right asymmetry determination that closely mirror human patient symptoms .

What are the most critical unanswered questions regarding ARL2BP's role in ciliopathies?

Critical unanswered questions in ARL2BP research:

  • Molecular mechanism questions:

    • What is the direct molecular mechanism by which ARL2BP ensures proper closure of the B-tubule inner junction in doublet microtubules?

    • Does ARL2BP interact directly with tubulin or tubulin-modifying enzymes, or does it scaffold other microtubule-associated proteins?

    • How does ARL2BP cooperate with CFAP20, PACRG, and Tektin to regulate doublet microtubule structure?

    • Is the interaction with ARL2 necessary for ARL2BP's ciliary functions in vivo?

  • Cell biology questions:

    • How is ARL2BP targeted to specific ciliary subcompartments?

    • What post-translational modifications regulate ARL2BP localization or function?

    • Does ARL2BP play distinct roles during initial ciliogenesis versus ciliary maintenance?

    • How do ARL2BP's roles in STAT3 signaling relate to its ciliary functions?

  • Disease mechanism questions:

    • Why do ARL2BP mutations predominantly affect photoreceptors and sperm despite its presence in multiple ciliated tissues?

    • What is the mechanistic basis for the more severe phenotype in human patients compared to mouse models?

    • Are there modifier genes that influence the penetrance or expressivity of ARL2BP mutations?

    • Could ARL2BP deficiency contribute to ciliopathies in patients without ARL2BP mutations?

  • Therapeutic potential questions:

    • Can gene replacement therapy rescue established disease phenotypes or only prevent progression?

    • Are there pharmacological approaches to stabilize doublet microtubules that could bypass ARL2BP deficiency?

    • Could modulation of the ARL2BP-ARL2 interaction serve as a therapeutic approach?

    • Which biomarkers would best track therapeutic efficacy in ARL2BP-related ciliopathies?

Addressing these questions will require innovative approaches combining structural biology, advanced imaging, and in vivo disease models utilizing biotin-conjugated antibodies for consistent detection across methodologies .

How might differential protein interactome analysis advance our understanding of ARL2BP's tissue-specific functions?

Differential interactome analysis strategies for ARL2BP:

  • Tissue-specific BioID approaches:

    • Generate tissue-specific ARL2BP-BioID fusion knock-in models

    • Compare proximity interactomes between:

      • Photoreceptor connecting cilia vs. respiratory cilia

      • Developing sperm flagella vs. mature flagella

      • Embryonic nodal cilia vs. adult primary cilia

    • Identify tissue-specific interactors that may explain selective vulnerability

  • Quantitative comparative interactomics:

    • Perform immunoprecipitation with biotin-conjugated ARL2BP antibodies from:

      • Retinal tissue at different developmental stages

      • Testicular tissue during spermatogenesis

      • Brain tissue with focus on ventricular regions

    • Use SILAC or TMT labeling for direct quantitative comparison

    • Identify developmental stage-specific interaction networks

  • Mutation-specific interactome changes:

    • Compare wild-type vs. disease-causing mutant interactomes

    • Analyze how specific mutations (e.g., p.Met45Arg) alter interaction profiles

    • Identify gained or lost interactions that explain pathogenic mechanisms

    • Correlate specific interaction losses with observed phenotypes

  • Spatial resolution of interactions:

    • Combine proximity ligation assays with super-resolution microscopy

    • Map interaction territories within ciliary subdomains

    • Compare interaction patterns between different ciliary subtypes

    • Correlate with ultrastructural defects observed by electron microscopy

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.