AGO3 Antibody, HRP conjugated

Shipped with Ice Packs
In Stock

Description

Introduction to AGO3 Antibody, HRP Conjugated

The AGO3 Antibody, HRP conjugated refers to an antibody targeting Argonaute 3 (AGO3), a core component of RNA-induced silencing complexes (RISCs), covalently linked to Horseradish Peroxidase (HRP). While direct commercial availability of pre-conjugated AGO3-HRP antibodies is limited, researchers often employ HRP-conjugated secondary antibodies in combination with anti-AGO3 primaries. This article synthesizes methodologies, applications, and research findings to provide a comprehensive overview of AGO3 antibody-HRP systems.

Structure and Function of AGO3

AGO3 is a member of the Argonaute protein family, characterized by PAZ and Piwi domains. It plays a role in RNA interference (RNAi) pathways, though its catalytic activity is less prominent compared to AGO2. AGO3 is implicated in gene regulation and stress response mechanisms, with expression observed in human, mouse, and rat tissues .

Key Features of AGO3Details
Domain CompositionPAZ domain (binds RNA), Piwi domain (binds target DNA/RNA)
FunctionRNAi pathway component, gene regulation, stress response
ExpressionUbiquitous in human, mouse, rat tissues

AGO3 Antibodies in Research: Applications and Validation

Primary anti-AGO3 antibodies are validated for diverse applications, including:

  • Immunohistochemistry (IHC): Detection in paraffin-embedded tissues (e.g., human cervical carcinoma, rat brain) .

  • Western Blot (WB): Identification of AGO3 in lysates (e.g., HEK293, CHO cells) .

  • Immunofluorescence (IF): Localization in U2OS cells using DyLight®488-conjugated secondaries .

  • Flow Cytometry: Intracellular staining of Jurkat cells .

HRP Conjugation Methods for AGO3 Antibodies

For custom HRP conjugation, researchers employ cross-linking strategies:

Key Conjugation Techniques

MethodReagentsMechanismAdvantagesChallenges
Periodate OxidationSodium metaperiodate, sodium cyanoborohydrideOxidizes HRP carbohydrates to aldehydes; forms Schiff base with antibody aminesHigh yield, stable conjugatesRequires strict pH control
Maleimide-Thiol CouplingSulfo-SMCC, SATAActivates HRP with maleimide; thiolates antibody for covalent bondingSite-specific conjugationCritical antibody concentration
Lyophilization-EnhancedLyophilized HRP post-activationReduces reaction volume, increases collision frequencyHigher HRP:antibody ratioRequires specialized equipment

Example Workflow

  1. Oxidize HRP: Treat with 0.15 M sodium metaperiodate to generate aldehydes .

  2. Dialyze: Remove excess oxidizing agents using PBS .

  3. Lyophilize: Freeze-dry to concentrate reactive HRP .

  4. Conjugate: Mix with thiolated AGO3 antibody (1:4 molar ratio) and stabilize with sodium cyanoborohydride .

Immunohistochemistry

Primary anti-AGO3 antibodies (e.g., Bosterbio A04191-1) are paired with HRP-conjugated secondaries (e.g., goat anti-rabbit HRP) for chromogenic detection using DAB .

Example Protocol:

  1. Antigen Retrieval: EDTA buffer (pH 8.0) for paraffin sections .

  2. Blocking: 10% goat serum .

  3. Primary Incubation: 2 μg/mL rabbit anti-AGO3 overnight at 4°C .

  4. Secondary Incubation: HRP-conjugated goat anti-rabbit IgG for 30 minutes at 37°C .

  5. Detection: DAB/H₂O₂ substrate .

Western Blotting

AGO3 detection in lysates (e.g., HEK293, CHO cells) uses primary antibodies with HRP secondaries, enabling chemiluminescent or chromogenic readouts .

RNA Immunoprecipitation (RIP)

While not explicitly documented for AGO3, analogous Ago2-HRP systems enable co-IP of miRNAs for sequencing .

Performance Metrics and Challenges

ParameterHRP-Conjugated SecondaryCustom-Conjugated Primary
SensitivityModerate (1:1000–1:5000 dilution) High (1:25–1:5000)
Signal-to-NoiseModerate (background in complex samples)High (controlled conjugation)
Cross-ReactivityLow (species-specific secondaries) Variable (depends on antibody purity)

Key Challenges:

  • Conjugation Efficiency: HRP:antibody ratios (2–4 HRP/antibody) impact activity .

  • Buffer Incompatibility: Detergents (e.g., SDS) disrupt conjugation .

  • Storage Stability: Lyophilized conjugates require -20°C storage .

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 orders. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timelines.
Synonyms
Protein argonaute-3 (Argonaute3) (hAgo3) (Argonaute RISC catalytic component 3) (Eukaryotic translation initiation factor 2C 3) (eIF-2C 3) (eIF2C 3), AGO3, EIF2C3
Target Names
AGO3
Uniprot No.

Target Background

Function
AGO3 plays a crucial role in RNA-mediated gene silencing (RNAi). It binds to short RNAs, such as microRNAs (miRNAs), and suppresses the translation of mRNAs that are complementary to them. It is believed to contribute to the stabilization of small RNA derivatives (siRNA) derived from processed RNA polymerase III-transcribed Alu repeats containing a DR2 retinoic acid response element (RARE) in stem cells. Subsequently, AGO3 participates in the siRNA-dependent degradation of a subset of RNA polymerase II-transcribed coding mRNAs by recruiting a mRNA decapping complex involving EDC4. While possessing RNA slicer activity, it exhibits this activity only on select RNAs possessing 5'- and 3'-flanking sequences to the region of guide-target complementarity.
Gene References Into Functions
  1. Research demonstrates that recombinant AGO3 loaded with miR-20a effectively cleaves complementary target RNAs. PMID: 29040713
  2. A study has unveiled a novel example of a vertebrate protein factor, Argonaute-3 (AGO3), specifically influencing the guide-to-passenger-strand ratio of the miRNA let-7a. This research proposes a multifaceted mechanism for the observed impact of AGO3 on the let-7a-3p passenger strand expression and activity. PMID: 24100239
  3. The N-terminal domain of AGO3 inhibits cleavage activity. Amino acids 1-64 exhibit a largely unstructured conformation and form a substantial loop encircling a region comprising two helices and another unstructured loop. PMID: 23665583
  4. The PIWI domain of AGO3 is capable of slicing, but our AGO3 model suggests multiple interactions between residues in the PIWI and N domains. These interactions may enhance protein rigidity or induce other properties that contribute to the observed slicing deficiency of AGO3. PMID: 23748378
  5. DICER- and AGO3-dependent generation of retinoic acid-induced DR2 Alu RNAs regulates human stem cell proliferation. PMID: 23064648
  6. AGO3 exhibits the capacity to load microRNAs efficiently in the absence of AGO1 and AGO2, despite a significant reduction in global microRNA expression. PMID: 22474261
  7. Reliable predictions of miRNA affinity to both AGO2 and AGO3 proteins have been achieved. PMID: 21634124
  8. The specificity of RNA interference is contingent upon the relative concentrations of AGO1, AGO3, and AGO4 compared to AGO2. PMID: 18771919
  9. EIF@C3 protein is expressed in both Schwann and neuron-type differentiating cells. PMID: 19393748

Show More

Hide All

Database Links

HGNC: 18421

OMIM: 607355

KEGG: hsa:192669

STRING: 9606.ENSP00000362287

UniGene: Hs.657659

Protein Families
Argonaute family, Ago subfamily
Subcellular Location
Cytoplasm, P-body.

Q&A

What is AGO3 and why is it important in molecular biology research?

AGO3 (Argonaute 3, also known as EIF2C3) is a member of the Argonaute family of proteins that play critical roles in RNA interference (RNAi). It functions by binding to short RNAs such as microRNAs (miRNAs) and repressing the translation of mRNAs complementary to them. Unlike AGO2, AGO3 lacks endonuclease activity but still contributes to gene silencing through translational repression .

AGO3 research has significant implications for understanding:

  • Post-transcriptional gene regulation mechanisms

  • miRNA function and specificity

  • RNA-based therapeutic approaches

  • Cellular pathways affected by non-coding RNAs

AGO3 contains both PAZ and PIWI domains characteristic of the Argonaute family and has a molecular weight of approximately 97 kDa . It localizes primarily in cytoplasm, P-bodies, and the nucleoplasm, where it participates in various RNA regulatory processes .

What applications are suitable for HRP-conjugated AGO3 antibodies?

HRP-conjugated AGO3 antibodies are versatile tools applicable to multiple research techniques:

ApplicationTypical DilutionSample TypesDetection Method
Western Blotting1:1000-1:5000Cell/tissue lysatesChemiluminescence
ELISA1:100-1:500Purified protein, serumColorimetric/TMB substrate
Immunohistochemistry1:50-1:200FFPE tissuesDAB substrate
Immunocytochemistry1:100-1:500Fixed cellsDAB/AEC substrate
Flow Cytometry1:50-1:100Fixed/permeabilized cellsEnzyme-substrate reaction

How should I validate an HRP-conjugated AGO3 antibody before experimental use?

Thorough validation is essential for reliable results:

  • Positive controls: Use cell lines known to express AGO3 (e.g., HeLa, N2A, or SHG-44 cells)

  • Negative controls:

    • AGO3 knockout cells (ideally AGO1/2/3 triple knockout for complete effect)

    • Secondary-only controls (for background assessment)

    • Isotype controls (for non-specific binding assessment)

  • Specificity testing:

    • Western blot should show a single band at ~97 kDa

    • Immunoprecipitation followed by mass spectrometry

    • Peptide competition assay with the immunogen

  • Cross-reactivity assessment: Test against other Argonaute family members (AGO1, AGO2, AGO4) to ensure specificity, particularly important as they share structural similarities .

  • Antibody titration: Determine optimal concentration by testing serial dilutions (typically 1:500-1:5000) to maximize signal-to-noise ratio.

What buffer compositions are optimal for HRP-conjugated AGO3 antibody applications?

Buffer optimization is critical for maintaining antibody functionality and reducing background:

For Western Blotting:

  • Blocking buffer: 5% non-fat dry milk or 3-5% BSA in TBS-T (TBS + 0.05-0.1% Tween-20)

  • Antibody dilution buffer: 1-3% BSA in TBS-T (avoid sodium azide as it inhibits HRP activity)

  • Wash buffer: TBS-T with 0.05-0.1% Tween-20

  • pH range: Maintain between 7.2-7.6 for optimal HRP activity

For Immunoprecipitation:

  • Lysis buffer: 25 mM Tris pH 8.0, 300 mM NaCl, 5% glycerol, 0.04% Triton X-100

  • Wash buffer: 10-50 mM amine-free buffer (e.g., HEPES, MES, MOPS, or phosphate) at pH 6.5-8.5

Important Considerations:

  • Avoid buffers containing nucleophilic components like primary amines and thiols, as they may interfere with HRP conjugation chemistry

  • Sodium azide is an irreversible inhibitor of HRP and must be avoided in working solutions

  • EDTA and common non-buffering salts and sugars have minimal effect on conjugation efficiency

  • For long-term storage (>1 week), add stabilizing proteins like 1% BSA to prevent activity loss

How can I optimize signal detection when using HRP-conjugated AGO3 antibodies?

Signal optimization strategies:

  • Substrate selection based on detection needs:

Substrate TypeSensitivityDurationApplication
ECL (standard)MediumMinutes to hoursRoutine Western blots
ECL Plus/FemtoHighHoursLow abundance proteins
Radiance QVery highExtendedTrace amount detection
DABMediumPermanentIHC/ICC visualization
TMBHighStable colorELISA readout
  • Exposure optimization:

    • For digital imaging: Test multiple exposure times (1 sec to 5 min)

    • For film: Use multiple exposures (15 sec, 1 min, 5 min)

    • Avoid overexposure that can mask subtle differences in expression

  • Temperature considerations:

    • Conduct incubations at room temperature (20-25°C) for consistent enzyme kinetics

    • Allow substrates to equilibrate to room temperature before use

    • Maintain consistent temperature during development

  • Signal enhancement strategies:

    • Use signal enhancer reagents compatible with HRP

    • Consider protein concentration steps for samples with low AGO3 expression

    • Optimize membrane blocking to reduce background without affecting specific signal

What are the best approaches for multiplex detection involving AGO3 and other proteins?

For simultaneous detection of AGO3 and other targets:

  • Sequential detection method:

    • First detection: Use HRP-conjugated AGO3 antibody and develop

    • Stripping: Use commercially available HRP stripping buffer (e.g., Azure AC2154)

    • Second detection: Apply antibody against second target

    • Note: Complete stripping should be confirmed before re-probing

  • Parallel detection strategies:

    • Size-based separation: If target proteins differ significantly in molecular weight

    • Different reporter systems: Combine HRP with alkaline phosphatase conjugates

    • Fluorescent multiplex Western blotting: If specialized imaging equipment is available

  • Important considerations for multiplex detection:

    • Antibody compatibility: Ensure species compatibility to avoid cross-reactivity

    • Stripping efficiency: Incomplete stripping can lead to false positives

    • Signal bleed-through: Ensure complete quenching of first signal before second detection

    • Loading controls: Use differently sized housekeeping proteins for internal validation

How can HRP-conjugated AGO3 antibodies be used to study microRNA-mediated gene regulation?

Advanced methodological approaches:

  • RNA Immunoprecipitation (RIP) followed by qRT-PCR:

    • Crosslink protein-RNA complexes in vivo

    • Immunoprecipitate AGO3 using HRP-conjugated antibody with protein A/G beads

    • Extract and reverse transcribe associated RNAs

    • Quantify specific miRNAs by qPCR

    • Note: HRP activity should be inhibited before RNA extraction to prevent RNA degradation

  • Enhanced Crosslinking Immunoprecipitation (eCLIP-seq):

    • This technique allows mapping of AGO3 binding sites within 3'-UTRs with high resolution

    • Comparison between wild-type and AGO1/2/3 knockout cells reveals differential binding patterns

    • Results have indicated that AGO binding within 3'-UTRs is poorly correlated with gene repression

    • Combine with RNA-seq to correlate AGO3 binding with gene expression changes

  • Interactome analysis using IP-mass spectrometry:

    • Immunoprecipitate AGO3 protein complexes

    • Analyze by mass spectrometry to identify protein binding partners

    • Compare interactome changes under different cellular conditions

    • Validate key interactions using reciprocal co-immunoprecipitation

  • Reporter assays for functional validation:

    • Use LightSwitch 3'UTR Reporter systems to study functional impact of miRNA-3'UTR interactions

    • Co-transfect with miRNA mimics or inhibitors to modulate AGO3 function

    • Measure effects on luciferase reporter activity to quantify repression

How do I interpret discrepancies between AGO3 binding and gene repression outcomes?

Research has revealed intriguing contradictions that require careful analysis:

  • Binding vs. functional effects:

    • Recent studies show AGO binding within 3'-UTRs is poorly correlated with gene repression

    • Many clusters are associated with increased (not decreased) steady state levels of mRNA in wild-type versus knockout cells

    • The strongest AGO-binding cluster within MYC 3'-UTR showed opposite-than-expected expression effects

  • Analytical framework for interpreting contradictory results:

    • Examine the role of cooperative effects between multiple AGO proteins

    • Consider the impact of knocking out AGO1, AGO2, and AGO3 together to achieve full effects

    • Evaluate alternative AGO3 functions beyond simple miRNA-mediated repression

    • Assess potential regulatory roles in mRNA stability rather than translation

  • Technical considerations for discrepancy analysis:

    • Temporal dynamics: AGO3 effects may vary with time after miRNA engagement

    • Cell-type specificity: Different outcomes may reflect cell-specific cofactor availability

    • miRNA abundance: Saturation effects at high miRNA concentrations

    • 3'-UTR context: Surrounding sequence elements may influence AGO3 function

  • Recommended validation approaches:

    • Parallel analysis with multiple techniques (RIP, eCLIP, reporter assays)

    • Time-course experiments to capture dynamic effects

    • Dose-response studies with miRNA mimics/inhibitors

    • Careful controls including AGO1/2/3 single and combinatorial knockouts

What are the current technical limitations of HRP-conjugated AGO3 antibodies in research applications?

Understanding limitations is crucial for accurate data interpretation:

  • Signal amplification constraints:

    • Direct HRP-conjugated primary antibodies provide less signal amplification than two-step detection systems

    • The indirect primary-secondary approach allows multiple secondary antibodies to bind each primary antibody, enhancing sensitivity

    • This limitation is particularly relevant for detecting low-abundance AGO3 in certain cell types

  • Technical considerations for specific applications:

    • Immunofluorescence: HRP conjugates require enzymatic development and are less suitable than fluorophore conjugates

    • FACS analysis: Limited dynamic range compared to fluorescent conjugates

    • Long-term storage: Potential loss of HRP activity over time even with proper storage

    • Multiplexing capability: Challenges in simultaneous detection of multiple targets

  • Experimental design adjustments:

    • For low-abundance targets: Consider using unconjugated primary followed by HRP-conjugated secondary

    • For quantitative applications: Include standard curves and linear range determination

    • For challenging samples: Implement signal enhancement strategies or more sensitive detection systems

    • For long-term projects: Prepare smaller aliquots to minimize freeze-thaw cycles that reduce activity

How can I address non-specific background when using HRP-conjugated AGO3 antibodies?

Systematic troubleshooting approaches:

  • Common sources of background and their solutions:

ProblemLikely CauseSolution
Uniform backgroundInsufficient blockingIncrease blocking time/concentration
Speckled backgroundAntibody aggregationCentrifuge antibody before use
Edge effectsUneven dryingMaintain humidity during incubations
High background in specific cell typesEndogenous peroxidase activityInclude peroxidase quenching step
Non-specific bands on Western blotCross-reactivityOptimize antibody dilution; use knockout controls
  • Optimization strategies:

    • Titrate antibody concentration to find optimal signal-to-noise ratio

    • Test different blocking agents (milk, BSA, normal serum)

    • Include 0.05-0.3% Tween-20 in wash buffers to reduce hydrophobic interactions

    • Perform additional wash steps with larger volumes

    • For IHC/ICC, include an endogenous peroxidase quenching step (0.3% H₂O₂ in methanol)

  • Validation controls:

    • Include isotype controls to assess non-specific binding

    • Use AGO3 knockout/knockdown samples as negative controls

    • Perform peptide competition assays to confirm specificity

What are the strategies for detecting low abundance AGO3 protein in experimental samples?

Enhanced detection methodologies:

  • Sample preparation optimization:

    • Enrich AGO3 by immunoprecipitation before detection

    • Use phosphatase inhibitors to preserve potentially phosphorylated forms

    • Optimize cell lysis conditions (25 mM Tris pH 8.0, 300 mM NaCl, 5% glycerol, 0.04% Triton X-100)

  • Signal enhancement approaches:

    • Use high-sensitivity substrates like Radiance Q

    • Implement tyramide signal amplification (TSA) for tissue sections

    • Consider using biotin-streptavidin systems for additional amplification

    • Extend primary antibody incubation time (overnight at 4°C)

  • Detection system optimization:

    • Use cooled CCD camera systems for digital capture of faint signals

    • Consider longer exposure times with low-background substrates

    • Use signal enhancers compatible with HRP detection systems

    • For Western blots, use PVDF membranes (rather than nitrocellulose) for higher protein binding capacity

  • Technical considerations:

    • Fresh reagent preparation is critical for maximum sensitivity

    • Allow substrates to equilibrate to room temperature before use

    • Keep exposure to light minimal until development

    • Consider concentration steps for dilute samples

How do different fixation and permeabilization protocols affect AGO3 detection?

Optimal sample preparation is crucial for accurate AGO3 visualization:

  • Impact of fixation methods on AGO3 epitope preservation:

Fixation MethodAdvantagesDisadvantagesRecommended Applications
4% ParaformaldehydeGood structural preservationMay mask some epitopesICC/IF for subcellular localization
Methanol/AcetoneBetter preservation of nuclear epitopesPotential protein extractionNuclear AGO3 detection
Formalin (FFPE tissue)Compatible with archived samplesRequires antigen retrievalIHC in tissue sections
Gentle fixation (0.5-2% PFA)Maintains protein interactionsLess structural preservationRIP and protein complex studies
  • Permeabilization optimization:

    • Triton X-100 (0.1-0.5%): Effective for nuclear AGO3 detection

    • Saponin (0.1%): Gentler permeabilization preserving cytoplasmic structures

    • Digitonin (0.001-0.01%): Selective plasma membrane permeabilization for cytoplasmic AGO3

    • Note: AGO3 localizes to cytoplasm, P-bodies, and nucleoplasm, requiring balanced permeabilization

  • Antigen retrieval methods for tissue sections:

    • Heat-induced epitope retrieval: Citrate buffer (pH 6.0) or EDTA (pH 9.0)

    • Enzymatic retrieval: Proteinase K (2-5 μg/ml) for 5-15 minutes

    • Combined approach: Mild enzymatic treatment followed by heat retrieval

    • Optimization is tissue-dependent and may require method comparison

  • Validation strategy:

    • Compare multiple fixation/permeabilization methods with the same antibody lot

    • Confirm specificity with appropriate controls for each condition

    • Evaluate subcellular localization patterns against published data

    • Document optimal conditions for reproducibility

How can HRP-conjugated AGO3 antibodies contribute to understanding non-canonical AGO3 functions?

Exploring novel AGO3 activities:

  • Beyond miRNA-mediated repression:

    • Recent findings suggest AGO3 may have functions independent of canonical miRNA pathways

    • Some AGO3-associated mRNAs show increased (rather than decreased) expression in wild-type vs. knockout cells

    • Methodological approaches to study non-canonical functions include:

      • Comprehensive interactome analysis

      • RNA-seq comparing AGO3 single knockout vs. AGO1/2/3 triple knockout

      • Selective mutation of AGO3 domains to dissect functional requirements

  • AGO3 in specialized cellular contexts:

    • P-bodies: AGO3 localizes to these cytoplasmic RNA processing centers

    • Stress granules: Potential roles in stress response pathways

    • Nuclear functions: AGO3 also localizes to nucleoplasm, suggesting nuclear roles

    • Research methods to explore these contexts include:

      • Co-localization studies with compartment-specific markers

      • Selective isolation of subcellular compartments followed by IP

      • FRAP (Fluorescence Recovery After Photobleaching) to assess dynamics

  • AGO3 interaction with non-coding RNAs beyond miRNAs:

    • Potential interactions with long non-coding RNAs

    • Roles in chromatin remodeling and transcriptional regulation

    • Approaches include:

      • CLIP-seq targeting various RNA populations

      • RNA pulldown followed by mass spectrometry

      • Chromatin immunoprecipitation to assess DNA-associated functions

What recent technological advances improve the specificity and sensitivity of AGO3 detection?

Cutting-edge methodological improvements:

  • Advanced antibody engineering approaches:

    • Recombinant antibody technology ensures batch-to-batch consistency

    • Single-chain variable fragments (scFvs) for improved tissue penetration

    • Site-specific conjugation strategies to maintain antigen recognition

    • Implementation considerations:

      • Validation against traditional antibodies

      • Optimization of conjugation chemistry

      • Application-specific testing

  • Enhanced signal development systems:

    • Tyramine signal amplification (TSA) compatible with HRP conjugates

    • Metal-enhanced DAB precipitation for electron microscopy applications

    • Chemiluminescent substrates with extended signal duration

    • Technical requirements:

      • Specialized equipment for some detection methods

      • Careful optimization to prevent overdevelopment

      • Additional controls for background assessment

  • Single-cell and spatial transcriptomics integration:

    • Combining AGO3 protein detection with RNA analysis at single-cell level

    • Spatial mapping of AGO3-RNA interactions in tissue context

    • Implementation approaches:

      • Sequential immunodetection and RNA hybridization protocols

      • Computational integration of protein and RNA datasets

      • Validation with orthogonal methods

How does AGO3 function differ from other AGO family members in experimental systems?

Comparative analysis of Argonaute proteins:

  • Functional distinctions between AGO family members:

    • AGO2 possesses endonuclease/slicer activity; AGO3 lacks this capability

    • AGO1/2/3 knockout studies show that all three are needed for maximum effect on gene expression

    • AGO3 appears to have distinct roles in specific cellular contexts

    • Research approaches for comparative studies:

      • Selective knockdown/knockout of individual AGO proteins

      • Chimeric protein construction to map domain-specific functions

      • Parallel AGO1/2/3/4 immunoprecipitation to identify unique binding partners

  • Experimental approaches to differentiate AGO functions:

    • RNA binding specificity:

      • CLIP-seq analysis of each AGO protein reveals distinct RNA target preferences

      • AGO3 shows binding patterns not strictly correlated with gene repression

    • Protein interactome distinctions:

      • IP-mass spectrometry to identify unique binding partners

      • Proximity labeling approaches (BioID, APEX) to map spatial interactions

    • Subcellular localization differences:

      • AGO3 shows distinct localization patterns in P-bodies and nucleoplasm

      • Co-localization studies with compartment-specific markers

  • Interpreting AGO redundancy vs. specificity:

    • Context-dependent functions may explain contradictory experimental results

    • Cell type-specific roles require careful experimental design

    • Developmental timing may affect relative importance of different AGO proteins

    • Research design considerations:

      • Use of multiple cell types and differentiation states

      • Combined knockouts to address functional redundancy

      • Quantitative analysis of binding affinities and target preferences

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 2024 Thebiotek. All Rights Reserved.