IVNS1ABP Antibody, FITC conjugated consists of an anti-IVNS1ABP immunoglobulin chemically linked to fluorescein isothiocyanate (FITC), a green fluorescent dye (excitation/emission: 495/519 nm). The antibody specifically targets IVNS1ABP, a 642-amino-acid protein involved in actin cytoskeleton stabilization, pre-mRNA splicing, and viral replication processes . The FITC conjugation allows fluorescence-based detection in techniques like immunofluorescence (IF) and flow cytometry.
Immunofluorescence (IF): Localizes IVNS1ABP in cellular compartments, particularly actin-rich structures .
Western Blotting (WB): Detects IVNS1ABP at ~70 kDa in human, mouse, and rat samples .
Actin Cytoskeleton Studies: Visualizes IVNS1ABP's role in stabilizing F-actin networks .
Viral Research: Investigates IVNS1ABP's interaction with influenza A virus NS1 protein during infection .
IVNS1ABP binds F-actin via Kelch repeats, preventing depolymerization and protecting cells from actin destabilization-induced apoptosis . The FITC conjugate has been used to map IVNS1ABP's colocalization with actin filaments in stress fibers .
Studies using this antibody revealed IVNS1ABP's role in influenza A virus mRNA splicing by interacting with HNRNPK, facilitating viral M2 protein synthesis .
FITC-labeled IVNS1ABP antibodies helped identify its modulation of ERK pathway activity, influencing cell proliferation and differentiation .
Vendor | Catalog # | Host | Clonality | Price |
---|---|---|---|---|
Santa Cruz Biotechnology | sc-373909 FITC | Mouse | Monoclonal | $330.00 |
antibodies-online.com | ABIN7156223 | Rabbit | Polyclonal | $389.00 |
Note: Prices reflect 2025 listings .
Fix cells with 4% paraformaldehyde.
Permeabilize with 0.1% Triton X-100.
Block with 5% BSA.
Image using a fluorescence microscope with FITC filters.
IVNS1ABP is a 642 amino acid protein that contains one BACK domain, one BTB (POZ) domain, and six Kelch repeats. It functions as a homodimer connected via its BTB domain and plays critical roles in multiple cellular processes. Research significance stems from its involvement in:
Pre-mRNA splicing and RNA processing
Actin cytoskeleton organization and stabilization through F-actin association
Aryl hydrocarbon receptor (AHR) pathway modulation
Protein ubiquitination regulation
Viral response mechanisms, particularly with influenza virus
The protein localizes to both nucleus and cytoplasm, making it relevant for studies of multiple cellular compartments and processes .
FITC conjugation provides several methodological advantages in IVNS1ABP research:
Direct visualization without secondary antibody requirements, reducing experimental complexity and potential cross-reactivity
Excitation maximum at approximately 495 nm and emission maximum at 519 nm, compatible with standard filter sets in most fluorescence microscopes
Enables multicolor immunofluorescence when combined with other fluorophores (e.g., PE, Alexa Fluor conjugates)
Facilitates flow cytometry (FCM) analysis for quantitative assessment of IVNS1ABP expression across cell populations
For optimal FITC-conjugated antibody performance, samples should be protected from light during incubation and storage to prevent photobleaching.
Based on manufacturer validation data and published research, FITC-conjugated IVNS1ABP antibodies can be reliably used for:
Application | Validated Parameters | Typical Dilution Range |
---|---|---|
Immunofluorescence (IF) | Cytoplasmic and nuclear localization visualization | 1:50-1:500 |
Flow Cytometry (FCM) | Cell population analysis | 1:50-1:200 |
Immunohistochemistry (IHC-P) | Paraffin-embedded tissue sections | 1:20-1:200 |
For each application, antigen retrieval methods and blocking protocols should be optimized based on tissue type and fixation method .
For robust IVNS1ABP localization using FITC-conjugated antibodies:
Fixation optimization: Compare 4% paraformaldehyde (10-15 minutes) versus methanol fixation (5 minutes at -20°C). Methanol fixation has been shown to better preserve cytoskeletal elements for IVNS1ABP visualization .
Permeabilization: Use 0.1-0.3% Triton X-100 for 5-10 minutes for nuclear IVNS1ABP detection. For cytoplasmic focus, reduce permeabilization time to 3-5 minutes.
Blocking strategy: Implement a dual blocking approach with 5% BSA and 5% normal serum (matching secondary antibody host if used in multiplex staining).
Co-localization studies:
F-actin co-staining: Use rhodamine-phalloidin (1:200) to visualize IVNS1ABP association with actin filaments
Nuclear co-localization: DAPI counterstaining (1:1000) for 5 minutes to assess nuclear versus cytoplasmic distribution ratios
Controls: Include unstained, secondary-only, and isotype controls alongside positive controls using cell lines with known IVNS1ABP expression (HeLa, MCF-7, U87-MG are validated positive controls) .
For optimal Western blot detection of IVNS1ABP:
Sample preparation:
Nuclear fractionation is recommended for enrichment as IVNS1ABP distributes between cytoplasmic and nuclear compartments
Use RIPA buffer with protease inhibitors, supplemented with 10mM N-ethylmaleimide to preserve ubiquitination status
Electrophoresis parameters:
Transfer and detection:
Semi-dry transfer: 15V for 60 minutes provides efficient transfer
Blocking: 5% non-fat milk in TBST for 1 hour at room temperature
Primary antibody dilution: 1:500-1:3000 determined through titration experiments
For fluorescent detection systems, secondary antibodies conjugated with compatible fluorophores (avoiding spectral overlap with FITC) should be considered
Validation approach: The expected 70-72 kDa band should be validated through siRNA knockdown experiments to confirm specificity .
For flow cytometry applications using FITC-conjugated IVNS1ABP antibodies:
Cell preparation considerations:
Fixation: 2% paraformaldehyde for 10-15 minutes at room temperature
Permeabilization: 0.1% saponin in PBS for intracellular staining
Cell concentration: 1×10^6 cells/mL is optimal for detection
Staining protocol optimization:
Antibody dilution: Start at 1:100 and titrate to determine optimal signal-to-noise ratio
Incubation conditions: 30-45 minutes at room temperature in the dark
Washing buffer: PBS with 1% FBS and 0.1% sodium azide
Instrument setup:
Use FITC channel (typically FL1 on many cytometers)
Compensation is critical when multiplexing with other fluorophores
Include single-color controls for each fluorophore
Analytical approaches:
IVNS1ABP interacts with influenza virus NS1 protein, playing a critical role in viral replication. Advanced research approaches include:
Co-immunoprecipitation studies:
Use FITC-conjugated IVNS1ABP antibodies to visualize co-localization with viral NS1 protein
Immunoprecipitate with anti-IVNS1ABP followed by Western blot detection of NS1
Validate interactions with proximity ligation assay (PLA) for in situ detection
Functional analysis during infection:
Monitor IVNS1ABP relocalization during viral infection timepoints (0, 2, 4, 8, 12, 24 hours post-infection)
Conduct FRAP (Fluorescence Recovery After Photobleaching) experiments to assess dynamics
Implement IVNS1ABP knockdown/knockout to evaluate effects on viral replication
Alternative splicing investigation:
These approaches help delineate the mechanistic role of IVNS1ABP in viral replication and host-pathogen interactions.
For investigating IVNS1ABP's cytoskeletal functions:
Live-cell imaging approaches:
Transfect cells with IVNS1ABP-GFP fusion constructs alongside LifeAct-RFP for real-time visualization
Implement photo-activatable IVNS1ABP constructs for spatiotemporal control
Use TIRF microscopy to visualize membrane-proximal cytoskeletal rearrangements
Biochemical fractionation:
Separate G-actin and F-actin fractions using ultracentrifugation
Quantify IVNS1ABP distribution between fractions under various treatments
Perform in vitro F-actin binding/bundling assays with purified components
Cytoskeletal perturbation experiments:
Structure-function analysis:
Generate domain deletion mutants (ΔBTB, ΔKelch) to assess domain-specific functions
Point mutations at key residues within Kelch repeats to disrupt F-actin binding
Investigate binding partner interactions using proximity-dependent biotinylation (BioID)
Recent studies highlight IVNS1ABP's role in modulating macrophage responses to inflammatory stimuli. Advanced methodologies include:
Macrophage polarization studies:
Monitor IVNS1ABP expression during M1 (pro-inflammatory) vs. M2 (anti-inflammatory) polarization
Assess cytoskeletal changes using FITC-conjugated IVNS1ABP antibodies alongside phalloidin staining
Quantify phagocytic capacity under IVNS1ABP overexpression/knockdown conditions
Transcriptional regulation investigation:
ChIP assays to study c-myc binding to the Ivns1abp promoter under inflammatory conditions
Luciferase reporter assays with wild-type and mutant promoter constructs
Evaluate histone modifications at the Ivns1abp locus during inflammation
Functional consequences assessment:
Phagocytosis assays using fluorescent beads or labeled bacteria
Migration/chemotaxis analysis using Transwell or Ibidi μ-slide systems
Cytokine production profiling via multiplex ELISA or Cytometric Bead Array
In vivo validation approaches:
These methodologies provide comprehensive insights into IVNS1ABP's role in regulating macrophage function during inflammation.
Issue | Possible Causes | Solution Approach |
---|---|---|
Weak or absent signal | Photobleaching | Protect samples from light; add anti-fade reagents; reduce exposure time |
Low antigen abundance | Implement signal amplification (TSA); optimize antigen retrieval methods | |
Improper storage | Store at -20°C, avoid freeze-thaw cycles; aliquot antibody | |
High background | Insufficient blocking | Increase blocking time; use combination of BSA and serum |
Non-specific binding | Titrate antibody; pre-adsorb with acetone powder from non-relevant tissues | |
Autofluorescence | Include unstained control; use Sudan Black B (0.1% in 70% ethanol) to quench | |
Cross-reactivity | Similar epitopes | Validate specificity with knockdown/knockout controls; pre-adsorb antibody |
Variability between experiments | Inconsistent protocol | Standardize fixation duration, antibody dilution, and incubation times |
For troubleshooting FITC-specific issues, use a spectral detector to confirm emission profile and rule out photoconversion or unexpected spectrum shifts .
When conflicting localization patterns are observed:
Systematic validation approach:
Compare multiple antibody clones targeting different IVNS1ABP epitopes
Validate with genetic tagging (IVNS1ABP-GFP) or CRISPR knock-in of endogenous tags
Perform subcellular fractionation followed by Western blot as biochemical validation
Biological variable consideration:
Cell cycle dependence: Synchronize cells and analyze IVNS1ABP localization at defined cell cycle stages
Cell type differences: Compare expression patterns across multiple relevant cell types
Stimulus-dependent relocalization: Standardize culture conditions and stress inducers
Technical parameter standardization:
Rigorous validation requires multiple controls:
Genetic controls:
siRNA/shRNA knockdown: ≥70% reduction in target expression
CRISPR/Cas9 knockout: Complete elimination of target protein
Rescue experiments: Re-expression of siRNA-resistant constructs
Antibody specificity controls:
Peptide competition: Pre-incubation with immunizing peptide should abolish signal
Multiple antibodies targeting different epitopes
Recombinant protein controls: Overexpression systems with tagged IVNS1ABP
Technical controls:
Secondary antibody-only control
Isotype control at equivalent concentration
Endogenous IVNS1ABP-negative cell line (if available)
Functional validation:
Implementing these comprehensive controls ensures confidence in experimental findings and facilitates detection of genuine biological effects versus technical artifacts.
Recent genetic data has implicated IVNS1ABP in Immunodeficiency 70, opening new research avenues:
Patient-derived cell analysis:
Compare IVNS1ABP expression, localization, and phosphorylation status in patient vs. control cells
Assess cytoskeletal organization using FITC-conjugated IVNS1ABP antibodies
Evaluate immune cell functions (phagocytosis, migration, immune synapse formation)
Variant functional characterization:
Generate disease-associated mutations using CRISPR base editing
Assess protein stability, interaction network, and subcellular localization
Determine impact on downstream signaling pathways
Therapeutic exploration:
Screen for compounds that restore IVNS1ABP function or bypass pathways
Develop gene correction approaches
Identify compensatory mechanisms in resilient patients
Mechanistic investigation:
These approaches may provide insights into disease mechanisms and potential therapeutic targets.
Emerging technologies offer new opportunities to investigate IVNS1ABP's splicing functions:
Transcriptome-wide approaches:
RIP-seq (RNA immunoprecipitation sequencing) to identify direct RNA targets
CLIP-seq (Cross-linking immunoprecipitation) for precise binding site mapping
Nanopore direct RNA sequencing to characterize full-length isoforms
In situ visualization techniques:
FISH-IF co-labeling to visualize target RNAs and IVNS1ABP simultaneously
Live-cell imaging with MS2-tagged RNAs and fluorescently labeled IVNS1ABP
Proximity ligation assays to detect interactions with splicing factors
Functional genomics screening:
CRISPR-Cas13 RNA targeting to modulate specific IVNS1ABP-RNA interactions
Splicing reporter assays with minigene constructs
High-throughput mutagenesis of IVNS1ABP binding sites
Structural approaches:
These cutting-edge approaches will provide mechanistic insights into IVNS1ABP's role in RNA processing.
IVNS1ABP's interaction with influenza virus NS1 protein suggests potential antiviral applications:
Drug discovery approaches:
High-throughput screens for compounds that modulate IVNS1ABP-NS1 interactions
Structure-based design of peptide inhibitors targeting interaction interfaces
Development of degraders (PROTACs) targeting viral proteins through IVNS1ABP machinery
Broad-spectrum antiviral investigation:
Screen for IVNS1ABP interactions with other viral proteins
Compare cytoskeletal responses across multiple viral infections
Identify common mechanisms that could be targeted therapeutically
Host-directed therapy exploration:
Modulate IVNS1ABP expression or activity to enhance intrinsic antiviral responses
Target downstream pathways regulated by IVNS1ABP
Combination approaches with direct-acting antivirals
Predictive modeling:
These research directions may yield novel antiviral strategies targeting host factors rather than viral components, potentially reducing the risk of resistance development.
Multimodal approaches enhance research depth:
Correlative microscopy workflows:
CLEM (Correlative Light and Electron Microscopy): Visualize FITC-labeled IVNS1ABP followed by ultrastructural analysis
Combine immunofluorescence with super-resolution techniques (STORM, PALM) for nanoscale localization
Implement live-cell imaging followed by fixation and immunolabeling of the same cells
Multi-omics integration:
Combine imaging with proteomics by using laser capture microdissection of IVNS1ABP-rich regions
Correlate transcriptomics data with protein localization patterns
Implement spatial transcriptomics in tissues with IVNS1ABP immunofluorescence
Functional association techniques:
These integrated approaches provide multidimensional data for comprehensive mechanistic insights.