AGO3 Antibody, FITC conjugated is a fluorescently labeled immunological reagent targeting the Argonaute 3 (AGO3) protein, a key component of the RNA-induced silencing complex (RISC) involved in RNA interference (RNAi) pathways . FITC (fluorescein isothiocyanate) conjugation enables visualization of AGO3 in techniques requiring fluorescence detection, such as flow cytometry, immunofluorescence (IF), and immunohistochemistry (IHC) .
FITC-conjugated AGO3 antibodies are widely used for:
Intracellular localization studies (e.g., detecting AGO3 in U2OS cells via IF) .
Western blotting and ELISA, where high specificity and minimal background are critical .
Parameter | Value/Description | Source |
---|---|---|
Host Species | Rabbit | |
Reactive Species | Human, Mouse, Rat | |
Excitation/Emission | 499/515 nm | |
Observed MW | 97 kDa | |
Immunogen | Recombinant human AGO3 (M1–S181 or 251–400 AA) |
Staining Protocol: Fixed cells incubated with AGO3-FITC (1 μg/10⁶ cells) and DyLight®488 secondary antibody .
Result: Clear separation between stained (blue line) and isotype control (green line) .
FITC Labeling Impact: Higher FITC-to-antibody molar ratios (>6:1) correlate with reduced antigen-binding affinity and increased non-specific staining .
Recommended Molar Ratios:
Dilution Optimization: Test concentrations between 1–5 µg/mL for flow cytometry .
Fixation/Permeabilization: Required for intracellular staining (4% paraformaldehyde) .
Controls: Include isotype-matched IgG and unstained samples to validate specificity .
AGO3 (Argonaute 3, also known as eIF2C3) is a member of the Argonaute family of proteins involved in RNA interference (RNAi) mediated gene silencing through siRNA and miRNA effectors. It functions as a component of the RNA-Induced Silencing Complex (RISC) and microRNA (miRNA)-containing ribonucleoprotein particles (miRNP). Unlike its family member AGO2, AGO3 lacks endonuclease activity and does not appear to cleave target mRNA molecules. AGO3 is primarily involved in translational repression rather than direct mRNA cleavage. The protein contains both PAZ and PIWI domains, which are characteristic of the Argonaute family . AGO3 is primarily localized in the cytoplasm and participates in the stabilization of small RNA derivatives (riRNA) and subsequent riRNA-dependent degradation of mRNAs by recruiting decapping complexes involving EDC4 .
AGO3 is a protein with a molecular weight of approximately 97 kDa and consists of 860 amino acids in humans. Its structure includes the characteristic PAZ domain (which binds to the 3' ends of small RNAs) and the PIWI domain (which structurally resembles RNase H). AGO3 is predominantly localized in the cytoplasm , where it associates with processing bodies (P-bodies) that are involved in mRNA degradation and storage. The protein functions by binding to short RNAs such as microRNAs and repressing the translation of complementary mRNAs. AGO3 is encoded by the EIF2C3 gene (Gene ID: 192669) .
FITC (Fluorescein Isothiocyanate) conjugation provides direct fluorescent labeling of the AGO3 antibody, eliminating the need for secondary antibody detection in immunofluorescence applications. The conjugation process attaches the FITC molecule to the antibody while preserving the antibody's ability to recognize and bind to AGO3 protein epitopes. When working with FITC-conjugated antibodies, researchers should be aware that the fluorophore is sensitive to photobleaching and has an excitation maximum at approximately 495 nm and an emission maximum at approximately 519 nm. For optimal results, the FITC-conjugated AGO3 antibody should be stored protected from light at -20°C and aliquoted into multiple vials to avoid repeated freeze-thaw cycles, which can damage both the antibody and the fluorophore .
FITC-conjugated AGO3 antibodies are particularly useful for fluorescence-based applications. According to product information, the main applications include:
Immunofluorescence (IF) on paraffin-embedded tissues (IHC-P) with recommended dilutions of 1:50-200
Immunofluorescence on frozen sections (IHC-F)
Immunocytochemistry (ICC) for cultured cells
The direct fluorescent labeling makes these antibodies especially valuable for multi-color immunofluorescence staining, co-localization studies, and flow cytometry applications. When designing experiments, researchers should consider the specific reactivity of the antibody, which typically includes human, mouse, and rat samples, with some products showing broader cross-reactivity with other species such as dog, cow, pig, horse, chicken, and zebrafish .
For optimal immunofluorescence results with FITC-conjugated AGO3 antibodies, follow this methodology:
For paraffin-embedded tissue sections (IHC-P):
Deparaffinize and rehydrate sections through a graded ethanol series
Perform antigen retrieval (typically heat-induced epitope retrieval in citrate buffer pH 6.0)
Block endogenous peroxidase activity with 3% H₂O₂ (if applicable)
Block non-specific binding with 5-10% normal serum in PBS for 1 hour
Incubate with FITC-conjugated AGO3 antibody at 1:50-200 dilution in blocking buffer overnight at 4°C
Wash thoroughly with PBS (3 × 5 minutes)
Counterstain nuclei with DAPI
Mount with anti-fade mounting medium
Visualize using a fluorescence microscope with appropriate filter sets (excitation ~495 nm, emission ~519 nm)
For cultured cells (ICC):
Fix cells with 4% paraformaldehyde for 15 minutes
Permeabilize with 0.1-0.25% Triton X-100 in PBS for 10 minutes
Block with 1-5% BSA or 10% normal serum in PBS for 1 hour
Incubate with FITC-conjugated AGO3 antibody at appropriate dilution in blocking buffer for 1-2 hours at room temperature or overnight at 4°C
Wash 3 × 5 minutes with PBS
Counterstain nuclei with DAPI
Mount and visualize
Store the antibody at -20°C and protect from light to maintain fluorescence intensity. Aliquot the antibody to avoid repeated freeze-thaw cycles .
To validate the specificity of FITC-conjugated AGO3 antibodies, implement these methodological approaches:
Positive and negative controls:
Use cell lines or tissues known to express AGO3 as positive controls
Use AGO3 knockout cells or tissues as negative controls
Compare with known expression patterns from literature or databases
Peptide competition assay:
Pre-incubate the antibody with the immunizing peptide before application
If the antibody is specific, the signal should be significantly reduced or eliminated
Cross-validation with different antibodies:
Compare staining patterns with other validated AGO3 antibodies targeting different epitopes
Consider using non-conjugated primary antibodies with secondary detection methods as reference
Western blot verification:
Confirm that the antibody recognizes a protein of the expected size (~97 kDa for AGO3)
Look for a single specific band at the correct molecular weight
siRNA knockdown:
Perform RNA interference to reduce AGO3 expression
Verify reduced signal intensity in knockdown samples compared to controls
Remember that the FITC-conjugated Argonaute 3/eIF2C3 polyclonal antibody is derived from a KLH-conjugated synthetic peptide corresponding to amino acid region 251-350 of the 860 amino acid human AGO3 protein , so expression patterns should match this epitope specificity.
When troubleshooting, remember that AGO3 is primarily cytoplasmic , so nuclear staining might indicate non-specific binding. Additionally, ensure that imaging is performed with appropriate filter sets for FITC (excitation ~495 nm, emission ~519 nm).
Differentiating between AGO3 and other Argonaute family proteins (AGO1, AGO2, AGO4) requires careful experimental design:
Antibody selection: Choose antibodies specifically targeting unique regions of AGO3 not conserved in other Argonaute proteins. The epitope information is crucial - the polyclonal antibody described in the search results targets amino acids 251-350 of human AGO3 .
Co-localization studies: Perform dual immunofluorescence with antibodies against different Argonaute proteins labeled with distinct fluorophores to observe differential localization patterns.
Functional assays: AGO3 lacks endonuclease activity, unlike AGO2 . In functional assays, AGO3 will not cleave perfectly complementary targets, whereas AGO2 will. This functional difference can help distinguish between these proteins.
Expression analysis: Use quantitative PCR to determine relative expression levels of different Argonaute transcripts in your experimental system.
RNAi approaches: Perform selective knockdown of AGO3 using siRNAs that specifically target AGO3 but not other family members, then confirm specificity with the antibody.
Immunoprecipitation followed by mass spectrometry: This approach can definitively identify the protein being recognized by the antibody based on peptide sequences.
Binding partners: AGO3 interacts with specific miRNAs and mRNAs that may differ from those interacting with other Argonaute proteins .
When interpreting results, note that AGO3 primarily functions in translational repression rather than mRNA cleavage, which is the main function of AGO2 .
When interpreting AGO3 subcellular localization patterns using FITC-conjugated antibodies, consider these key points:
Normal distribution: AGO3 is primarily localized in the cytoplasm , often concentrated in processing bodies (P-bodies) that appear as cytoplasmic foci. Any substantial nuclear staining should be carefully validated.
P-body association: AGO3 typically co-localizes with other P-body markers (like GW182, DCP1, DCP2). Consider co-staining experiments to confirm P-body localization.
Stress conditions: Under cellular stress (oxidative stress, heat shock, etc.), AGO3 distribution may change, potentially showing increased accumulation in stress granules. Compare stressed vs. unstressed cells to document these changes.
Cell cycle dependence: AGO3 distribution may vary throughout the cell cycle. Note cell morphology and potentially co-stain with cell cycle markers to correlate localization patterns with cell cycle phases.
Experimental artifacts: Overexpression of AGO3 can lead to artifactual localization patterns. Compare endogenous staining with overexpression systems carefully.
Fixation artifacts: Different fixation methods can affect AGO3 localization patterns. Cross-validate with multiple fixation protocols.
Comparison with functional data: Correlate localization patterns with functional readouts of miRNA activity to establish biological relevance.
In disease states or experimental manipulations, changes in AGO3 localization (such as nuclear accumulation or dispersion from P-bodies) may indicate alterations in miRNA pathway functionality that correlate with changes in target gene expression .
FITC-conjugated AGO3 antibodies offer powerful approaches for investigating RNA-protein interactions:
Immunofluorescence-FISH (IF-FISH) co-localization:
Combine FITC-AGO3 immunofluorescence with fluorescence in situ hybridization (FISH) for specific miRNAs or target mRNAs
This allows visualization of spatial relationships between AGO3 and its RNA partners
Use spectral unmixing if needed to distinguish FITC signal from other fluorophores
Proximity Ligation Assay (PLA) with RNA modification:
Adapt standard PLA protocols to detect AGO3-RNA interactions
Use FITC-conjugated AGO3 antibody with a complementary antibody against RNA-binding proteins
Alternatively, use biotinylated RNA probes with streptavidin-conjugated PLA probes
Live-cell imaging with complementary techniques:
While FITC-conjugated antibodies can't be used directly for live cells, they can validate findings from live-cell systems
Correlate fixed-cell FITC-AGO3 staining with data from live-cell experiments using fluorescently tagged AGO3
FRET-based approaches:
Use FITC as a donor fluorophore in Förster Resonance Energy Transfer (FRET) studies
Pair with acceptor fluorophores conjugated to antibodies against RNA-binding proteins or in situ hybridization probes
Immuno-electron microscopy:
Convert FITC signals to electron-dense materials for ultrastructural visualization
Map AGO3-RNA interactions at nanometer resolution
These advanced techniques can reveal mechanistic insights into how AGO3 participates in RNA silencing through interaction with miRNAs and target mRNAs, particularly its role in translational repression independent of endonucleolytic cleavage .
AGO3 has been implicated in various disease mechanisms, and FITC-conjugated antibodies offer valuable tools for investigation:
Cancer biology:
Altered AGO3 expression and localization have been observed in various cancers
FITC-conjugated AGO3 antibodies can be used to:
Quantify expression levels in tumor vs. normal tissues
Track subcellular redistributions associated with malignant transformation
Correlate with miRNA dysregulation patterns specific to cancer types
Neurodegenerative diseases:
miRNA dysregulation is implicated in conditions like Alzheimer's and Parkinson's
AGO3-FITC immunostaining can:
Reveal altered distribution in affected neurons
Detect co-localization with disease-specific protein aggregates
Monitor stress granule associations in neurodegeneration models
Viral infections:
Some viruses manipulate host miRNA machinery for replication
FITC-AGO3 antibodies enable:
Visualization of AGO3 redistribution during viral infection
Co-localization studies with viral components
Assessment of virus-induced alterations in miRNA-mediated silencing
Inflammatory disorders:
AGO3 may regulate inflammatory gene expression through miRNAs
Fluorescence-based quantification can:
Measure changes in AGO3 levels during inflammation
Track AGO3 in immune cell activation and differentiation
Developmental disorders:
When investigating disease mechanisms, combining FITC-AGO3 immunostaining with patient-derived samples and disease models can provide insights into how alterations in AGO3 function contribute to pathogenesis and potential therapeutic targets in the RNA interference pathway.
Integrating FITC-conjugated AGO3 antibodies with complementary molecular techniques creates powerful research paradigms:
Integration with sequencing technologies:
Fluorescence-activated cell sorting (FACS) of AGO3-FITC positive cells followed by RNA-seq or small RNA-seq
Correlation of AGO3 expression/localization patterns with transcriptome profiles
CLIP-seq validation using AGO3 antibodies to confirm binding partners identified in imaging
Combination with live-cell dynamics:
Fixed-cell FITC-AGO3 imaging to validate GFP-AGO3 live-cell experiments
Pulse-chase experiments to track AGO3 dynamics over time
Photoconversion studies paired with subsequent FITC-AGO3 immunofluorescence
Multi-omics approaches:
AGO3-FITC immunostaining of cells prepared for spatial transcriptomics
Integration of proteomics data with AGO3 localization patterns
Correlation of AGO3 distribution with epigenomic features
High-throughput screening applications:
FITC-AGO3 immunofluorescence in cell-based screens for miRNA pathway modulators
Automated image analysis of AGO3 localization changes in response to compound libraries
Validation of screen hits with biochemical assays using non-conjugated AGO3 antibodies
Super-resolution microscopy:
FITC-AGO3 antibodies can be used with techniques like STORM or STED
Nanoscale visualization of AGO3 within P-bodies and other subcellular structures
Co-localization with RNA targets at molecular resolution
In vivo applications:
Ex vivo staining of tissue sections from experimental animals
Correlation with in vivo functional studies of miRNA activity
Analysis of AGO3 expression in animal models of disease
This multidisciplinary approach leverages the direct visualization capabilities of FITC-AGO3 antibodies while enriching the findings with complementary molecular data, providing a comprehensive understanding of AGO3's roles in RNA silencing complexes and disease mechanisms .
A robust experimental design with FITC-conjugated AGO3 antibodies requires these essential controls:
Antibody specificity controls:
Peptide competition/blocking: Pre-incubate antibody with immunizing peptide to block specific binding
Knockout/knockdown validation: Use AGO3 knockout or siRNA-depleted samples as negative controls
Isotype control: Include FITC-conjugated isotype-matched non-specific antibody (e.g., rabbit IgG-FITC) to assess non-specific binding
Fluorescence controls:
Autofluorescence control: Unstained samples to determine background autofluorescence
Single-color controls: When performing multi-color experiments, include single-stained samples for each fluorophore
Photobleaching control: Monitor signal stability over multiple exposures
Technical controls:
Secondary antibody-only control: If using additional layers of detection
Fixation control: Compare different fixation methods to identify potential artifacts
Blocking optimization: Samples with different blocking conditions to minimize background
Biological controls:
Positive control tissue/cells: Samples known to express AGO3 (based on literature)
Related protein controls: Include staining for other Argonaute family members (AGO1, AGO2, AGO4) to assess specificity
Treatment conditions: Include relevant positive and negative treatment controls
Validation with orthogonal methods:
Western blot correlation: Verify that IF signal intensity correlates with protein levels detected by Western blot
qPCR validation: Confirm that protein expression patterns correlate with mRNA expression
A well-designed experiment should include comprehensive documentation of antibody characteristics: host species (rabbit), clonality (polyclonal), immunogen details (synthetic peptide corresponding to amino acids 251-350/860 of human AGO3), and storage conditions (-20°C, protected from light) .
Optimizing FITC-conjugated AGO3 antibody performance across tissue types requires systematic adjustment of multiple parameters:
Additional optimization strategies:
Tissue-specific blocking:
Brain: Add 0.1-0.3% Triton X-100 in blocking solution
Liver/kidney: Include 0.1% Sudan Black to reduce autofluorescence
High-lipid tissues: Include 0.05% saponin in blocking buffer
Signal amplification options:
For tissues with low AGO3 expression, consider using a non-conjugated primary antibody with a FITC-conjugated secondary antibody for signal amplification
Tyramide signal amplification can be used for very low abundance targets
Iterative optimization:
When transitioning between different tissue types, maintain detailed records of optimization parameters to build a reference database for future experiments.
Maintaining optimal FITC fluorescence stability requires careful management of several critical factors:
Light exposure:
FITC is highly susceptible to photobleaching
Prevention strategies:
Store antibody solutions in amber vials or wrapped in aluminum foil
Work under reduced lighting conditions
Minimize exposure during microscopy by using neutral density filters
Use minimal exposure times during image acquisition
pH sensitivity:
FITC fluorescence is optimal at pH 7.5-8.0 and diminishes significantly below pH 7.0
Management approaches:
Use buffers with consistent pH and sufficient buffering capacity
Monitor pH of solutions, especially during long-term storage
Consider pH-insensitive alternatives (like Alexa Fluor 488) for acidic environments
Storage conditions:
Proper storage is critical for maintaining antibody integrity and fluorescence
Best practices:
Mounting media selection:
Mounting media significantly impact fluorescence longevity
Recommendations:
Use anti-fade mounting media containing anti-photobleaching agents
Consider media with hardening properties for long-term storage
Seal edges of coverslips with nail polish for additional protection
Chemical environment:
Certain chemicals can quench FITC fluorescence
Considerations:
Avoid high concentrations of halogens or transition metals in buffers
Minimize exposure to oxidizing agents
Be cautious with fixatives containing high aldehyde concentrations
Image acquisition strategies:
Smart imaging approaches can preserve signal
Techniques:
Focus using differential interference contrast (DIC) before switching to fluorescence
Use computational approaches (like deconvolution) to enhance low-intensity signals
Consider spectral unmixing to separate FITC signal from autofluorescence
Documentation and quantification:
Include fluorescence intensity standards in experiments
Image all experimental groups in a single session with identical settings
Use automated exposure control where possible to standardize imaging