INTS10 Antibody, FITC conjugated is a polyclonal antibody designed for the detection of human Integrator complex subunit 10 (INTS10), a critical component of the Integrator complex involved in RNA polymerase II (RNAPII)-mediated transcriptional regulation . Conjugated to fluorescein isothiocyanate (FITC), this reagent enables fluorescent-based applications such as flow cytometry (FACS) and immunofluorescence .
SKU | Size | Price |
---|---|---|
QA26363_50ug | 50 µg | $190.00 |
QA26363_100ug | 100 µg | $299.00 |
Functional Studies: INTS10-INTS14 interaction depends on the INTS14 VWA domain’s MIDAS pocket . Disruption reduces snRNA processing efficiency .
Structural Data: Cryo-EM and SEC analyses confirm INTS10 integrates into a 1.2 MDa Integrator subcomplex .
Cross-Reactivity: Limited to human samples; no reactivity reported in mouse/rat .
Controls: Use knockdown/knockout cell lines (e.g., INTS10-depleted HEK293T) to confirm signal specificity .
Caution: Sodium azide (preservative) requires careful handling .
Current research focuses on:
INTS10 Antibody targets Integrator complex subunit 10 (also known as C8orf35 or INT10), which is a component of the Integrator complex involved in RNA processing and transcriptional regulation. The antibody enables researchers to investigate nuclear signaling pathways and epigenetic regulation mechanisms in which INTS10 participates. The FITC-conjugated version provides direct fluorescent detection capability without requiring secondary antibodies, streamlining immunofluorescent detection protocols. This antibody is particularly valuable for studying the localization and expression patterns of INTS10 in various cellular contexts, especially in research related to transcriptional regulation and RNA processing pathways .
The INTS10 Antibody, FITC conjugated, is a polyclonal antibody raised in rabbits with specificity for human INTS10 protein. It has the following specifications:
Property | Specification |
---|---|
Target | Integrator Complex Subunit 10 (INTS10) |
Host | Rabbit |
Clonality | Polyclonal |
Species Reactivity | Human, Mouse (depending on supplier) |
Immunogen | Recombinant Human Integrator complex subunit 10 protein (341-710AA or 451-710AA) |
Isotype | IgG |
Conjugate | FITC (Fluorescein isothiocyanate) |
Excitation/Emission | 499/515 nm |
Laser Line | 488 nm |
Form | Liquid |
Purity | >95%, Protein G purified |
UniProt Number | Q9NVR2 |
Storage Buffer | 0.01M PBS, pH 7.4, 0.03% Proclin 300, 50% Glycerol |
Storage Conditions | -20°C to -80°C, avoid repeated freeze/thaw cycles and exposure to light |
This antibody is intended for research use only and should not be used for diagnostic or therapeutic purposes .
While specific applications for FITC-conjugated INTS10 antibodies have limited documentation in the provided search results, similar polyclonal INTS10 antibodies have been validated for several applications. Based on the antibody's characteristics and general principles of FITC-conjugated antibodies, the following applications are suitable:
Immunofluorescence (IF): The FITC conjugation makes this antibody directly applicable for immunofluorescence studies without secondary antibody requirements. Recommended dilutions typically range from 1:50 to 1:200, though optimal concentrations should be determined empirically for each experiment .
Flow Cytometry: FITC-conjugated antibodies are commonly used in flow cytometric analysis. For intracellular proteins like INTS10, proper cell permeabilization protocols are essential for antibody access.
Immunocytochemistry (ICC): For detection of INTS10 in cultured cells, particularly when studying nuclear localization patterns.
Non-conjugated INTS10 antibodies have been successfully used in Western blotting at concentrations around 0.4 μg/ml for human samples, suggesting that the same target epitope recognition would be valid for the FITC-conjugated version, though the conjugation might affect some applications requiring protein denaturation .
For optimal immunofluorescence results with INTS10 Antibody, FITC conjugated, researchers should follow this methodological approach:
Sample Preparation:
Fix cells with 4% paraformaldehyde (10-15 minutes at room temperature)
Permeabilize with 0.1-0.5% Triton X-100 (5-10 minutes) to allow antibody access to nuclear targets
Blocking:
Block with 1-5% BSA or 5-10% normal serum (from species different from the host of the antibody) in PBS for 30-60 minutes to reduce background
Antibody Incubation:
Optimization Parameters:
Titrate antibody concentration (begin with manufacturer's recommendation and adjust based on signal-to-noise ratio)
Test different fixation methods if standard paraformaldehyde doesn't yield optimal results
Vary permeabilization conditions depending on subcellular localization (nuclear proteins like INTS10 may require stronger permeabilization)
Include appropriate positive controls (cell lines known to express INTS10) and negative controls (blocking peptide, isotype control)
Counterstaining and Mounting:
Counterstain nuclei with DAPI (1 μg/ml for 5 minutes)
Mount with anti-fade mounting medium to prevent photobleaching of FITC
Store slides at 4°C protected from light
The optimal protocol should be established through systematic testing of these parameters, with particular attention to signal specificity in the nuclear compartment where INTS10 is predominantly localized.
When working with INTS10 Antibody, FITC conjugated, researchers may encounter several technical challenges. Here are methodological solutions to common issues:
Low Signal Intensity:
Problem: FITC has lower quantum yield compared to newer fluorophores and is susceptible to photobleaching.
Solutions:
Use higher antibody concentration (while monitoring background)
Extend incubation time to overnight at 4°C
Employ signal amplification systems compatible with FITC
Use anti-fade mounting media containing DABCO or PPD
Adjust microscope settings (longer exposure, higher gain) while balancing photobleaching concerns
Consider tyramide signal amplification (TSA) for very low abundance targets
High Background/Non-specific Binding:
Problem: Polyclonal antibodies may exhibit cross-reactivity.
Solutions:
Increase blocking time and concentration (5% BSA or 10% normal serum)
Add 0.1-0.3% Triton X-100 to antibody dilution buffer
Include 0.1-0.5% Tween-20 in wash buffers and increase washing duration
Pre-adsorb antibody with acetone powder from non-relevant tissues
Use centrifugation (10,000g for 5 min) of diluted antibody to remove aggregates
Photobleaching:
Problem: FITC is particularly susceptible to photobleaching during imaging.
Solutions:
Minimize exposure to excitation light during sample preparation and imaging
Use anti-fade mounting media specifically formulated for FITC
Image FITC channels first in multi-channel experiments
Consider using sealed nail polish or mounting media that hardens to prevent oxygen exposure
Autofluorescence Interference:
Problem: Cellular autofluorescence can overlap with FITC spectrum.
Solutions:
Include unstained controls to determine autofluorescence levels
Use Sudan Black B (0.1-0.3% in 70% ethanol) to quench autofluorescence
Employ spectral unmixing during image acquisition if available
Consider longer wavelength fluorophores if autofluorescence remains problematic
Remember that INTS10 is a nuclear protein, so nuclear localization of the signal should be evident in positive samples . Absence of nuclear localization may indicate specificity issues requiring further optimization.
Validating antibody specificity is critical for ensuring reliable research outcomes. For INTS10 Antibody, FITC conjugated, researchers should implement the following methodological approaches:
Positive and Negative Controls:
Positive Controls: Use cell lines with confirmed INTS10 expression (e.g., A549 cells have been verified for INTS10 detection by immunofluorescence)
Negative Controls:
Isotype control (rabbit IgG-FITC) at equivalent concentration
Secondary-only controls (for detecting non-specific binding)
Cells with INTS10 knockdown via siRNA or CRISPR-Cas9 (most rigorous control)
Peptide Competition Assay:
Pre-incubate antibody with excess immunizing peptide (5-10x molar concentration)
Run parallel staining with blocked and unblocked antibody
Signal elimination/reduction in the peptide-blocked sample confirms specificity
Orthogonal Method Validation:
Cross-Species Reactivity Testing:
If the antibody claims reactivity to multiple species, test each species separately
Verify that staining patterns match known expression and localization patterns for each species
Co-localization Studies:
Co-stain with antibodies targeting known INTS10 interaction partners or nuclear markers
Confirm nuclear localization consistent with INTS10's role in the Integrator complex
Analyze co-localization quantitatively using Pearson's correlation coefficient or Manders' overlap coefficient
Molecular Weight Verification:
Although not directly applicable to immunofluorescence, running Western blots with non-conjugated versions of the same antibody can verify target specificity (expected MW for INTS10 is approximately 82 kDa)
These validation steps should be systematically documented and included in research publications to support the reliability of findings obtained using INTS10 Antibody, FITC conjugated.
For researchers investigating INTS10 in heterogeneous cell populations, multi-parameter flow cytometry offers powerful analytical capabilities. The following methodological approach optimizes INTS10 Antibody, FITC conjugated integration into multi-parameter panels:
Panel Design Considerations:
FITC excites at 488nm and emits at 515nm, positioning it in the first fluorescence channel (FL1) on most cytometers
Avoid fluorophores with significant spectral overlap (PE, Alexa Fluor 532)
Compatible fluorophores for multiplexing include APC (640/660nm), PE-Cy7 (480/785nm), and Pacific Blue (405/455nm)
Intracellular Staining Protocol:
Surface marker staining should precede fixation and permeabilization
Fix cells with 4% paraformaldehyde for 15-20 minutes at room temperature
Permeabilize with 0.1% Triton X-100 or commercial permeabilization buffers optimized for nuclear proteins
Block with 2-5% normal serum for 30 minutes
Incubate with INTS10 Antibody, FITC conjugated (starting dilution 1:100)
Wash thoroughly to remove unbound antibody
Assay Optimization Strategies:
Titrate INTS10 Antibody, FITC conjugated to determine optimal signal-to-noise ratio
Calculate staining index: (MFI positive - MFI negative) / (2 × SD of negative)
For nuclear proteins like INTS10, optimize permeabilization conditions (concentration, duration, temperature)
Include fluorescence minus one (FMO) controls for accurate gating
Experimental Applications:
Cell Cycle Analysis: Co-stain with DNA dyes (7-AAD, DRAQ5) to correlate INTS10 expression with cell cycle phases
Differentiation Studies: Combine with lineage markers to track INTS10 expression during cellular differentiation
Phospho-Flow Integration: Add phospho-specific antibodies to connect INTS10 with signaling pathways
Sorting Applications: Sort INTS10-positive cells for downstream applications (RNA-seq, ChIP-seq)
Data Analysis Approach:
Implement Boolean gating strategies to identify cell subpopulations based on INTS10 expression
Use dimensionality reduction techniques (tSNE, UMAP) for visualizing INTS10 distribution across cell populations
Quantify nuclear localization using nuclear/cytoplasmic ratio calculations
When implementing this approach, remember that INTS10's nuclear localization may require specialized permeabilization procedures compared to cytoplasmic proteins. This method enables quantitative assessment of INTS10 expression across different cell populations and experimental conditions.
Co-localization studies with INTS10 Antibody, FITC conjugated can provide valuable insights into protein-protein interactions and functional relationships within the Integrator complex. The following methodological framework ensures rigorous co-localization analysis:
Selection of Co-localization Partners:
Primary Targets: Other Integrator complex components (INTS1-14)
Functional Partners: RNA Polymerase II, U1/U2 snRNP components
Nuclear Domain Markers: SC35 (splicing speckles), Coilin (Cajal bodies), Fibrillarin (nucleolus)
Fluorophore Selection Strategy:
FITC (Ex/Em: 499/515nm) paired with spectrally distinct fluorophores:
Red range: Alexa Fluor 594, Texas Red, or Cy3
Far-red range: Alexa Fluor 647 or Cy5
Blue range: Pacific Blue or Alexa Fluor 405
Avoid fluorophores with spectral bleed-through into FITC channel
Advanced Microscopy Requirements:
Confocal Microscopy: Essential for accurate co-localization assessment
Optimize pinhole settings (1 Airy unit recommended)
Match Z-section thickness across channels
Nyquist sampling rate for optimal resolution
Super-Resolution Options:
Structured Illumination Microscopy (SIM): 2x resolution improvement
Stimulated Emission Depletion (STED): Superior resolution for precise co-localization
Single Molecule Localization Microscopy: For nanoscale interaction studies
Critical Control Experiments:
Negative Co-localization Control: Pair INTS10 with proteins known to occupy distinct nuclear compartments
Positive Co-localization Control: Pair with known INTS10 interacting partners
Single Wavelength Controls: Image each fluorophore separately to assess bleed-through
Antibody Order Controls: Reverse the order of antibody application to check for steric hindrance
Quantitative Analysis Protocol:
Pearson's Correlation Coefficient (PCC): Measures linear correlation between fluorescence intensities
Manders' Overlap Coefficient (MOC): Quantifies proportion of co-localizing pixels
Object-based methods: Count co-localizing vs. separate objects
Line scan analysis: Plot fluorescence intensity profiles along linear regions
Statistical validation: Compare experimental values against randomized pixel distributions
Advanced Data Interpretation:
Distinguish between true molecular interactions and spatial proximity due to nuclear organization
Consider the resolution limits of the imaging system (~200nm for confocal microscopy)
Validate key findings with complementary techniques (proximity ligation assay, co-immunoprecipitation)
This comprehensive approach enables researchers to accurately assess INTS10's spatial relationships with other nuclear proteins, providing insights into its functional roles in transcriptional regulation and RNA processing .
When investigating INTS10 expression changes across different experimental conditions, researchers should implement a systematic experimental design that controls for variables and ensures reproducible results. The following methodological framework is recommended:
Experimental Design Strategy:
Time-Course Analysis: Measure INTS10 expression at multiple time points following treatment (0, 2, 6, 12, 24, 48 hours)
Dose-Response Relationships: Test multiple concentrations of treatment agents to establish threshold effects
Biological Replicates: Minimum of three independent experiments with different cell passages
Technical Replicates: At least duplicate measurements within each biological replicate
Appropriate Controls: Vehicle controls, positive controls (treatments known to affect similar nuclear proteins)
Cell Models and Conditions for INTS10 Research:
Recommended Cell Lines: A549 (validated for INTS10 detection) , HEK293, HeLa
Primary Cells: Analyze tissue-specific expression patterns
Relevant Treatment Conditions:
Transcriptional inhibitors (α-amanitin, DRB)
Cell cycle synchronization (serum starvation, thymidine block)
Stress conditions (hypoxia, oxidative stress, UV irradiation)
Epigenetic modifiers (HDAC inhibitors, DNA methyltransferase inhibitors)
Quantification Methodology:
Flow Cytometry:
Measure mean fluorescence intensity (MFI) of INTS10-FITC signal
Normalize to cell count and unstained controls
Gate on relevant cell populations (viable, specific cell cycle phase)
Immunofluorescence Microscopy:
Capture images using consistent exposure settings
Measure nuclear signal intensity using automated image analysis software
Normalize to nuclear area and DAPI intensity
Analyze minimum of 50-100 cells per condition
Data Analysis Framework:
Statistical Methods:
ANOVA with appropriate post-hoc tests for multiple comparisons
Non-parametric alternatives (Kruskal-Wallis) if normality assumptions are violated
Mixed effects models for time-course experiments
Visualization Approaches:
Box plots showing distribution of expression levels
Scatter plots with overlaid means and standard deviations
Heat maps for multiple condition comparisons
Effect Size Calculation: Cohen's d or similar metrics to quantify magnitude of changes
Validation Strategy:
Confirm key findings with orthogonal methods (Western blot, RT-qPCR)
Use siRNA knockdown or CRISPR knockout to establish specificity of antibody signal
Compare findings with publicly available expression datasets
This experimental design provides a robust framework for investigating INTS10 expression changes, ensuring statistical power while controlling for technical and biological variables that could confound interpretation.
INTS10, as part of the Integrator complex, plays important roles in RNA processing and transcriptional regulation. The following methodological approaches leverage INTS10 Antibody, FITC conjugated as part of comprehensive research strategies:
Chromatin Association Studies:
Chromatin Immunoprecipitation followed by Immunofluorescence (ChIP-IF):
Perform standard ChIP protocol with non-conjugated INTS10 antibody
Visualize chromatin-bound INTS10 on spreads using INTS10-FITC antibody
Co-stain with RNA Polymerase II and transcription factors
Proximity Ligation Assay (PLA):
Combine INTS10-FITC with antibodies against transcription machinery components
Visualize protein-protein interactions as fluorescent spots
Quantify interaction frequency under different transcriptional states
Dynamic Analysis of INTS10 during Transcription:
Live Cell Imaging:
Combine fluorescently-tagged RNA Polymerase II with INTS10 immunofluorescence
Use pulse-chase labeling of nascent RNA (EU incorporation)
Track co-localization over time following transcriptional stimulus
Transcription Factory Analysis:
Visualize INTS10 in relation to transcription factories
Quantify spatial relationships during active vs. inhibited transcription
Combine with RNA FISH to correlate with specific gene expression
RNA Processing Investigation:
snRNA Maturation Analysis:
Couple INTS10-FITC immunofluorescence with FISH for precursor and mature snRNAs
Quantify co-localization coefficients between INTS10 and processing intermediates
Track processing kinetics after transcriptional pulse
Sequential Immunofluorescence:
Apply INTS10-FITC antibody along with markers for different RNA processing steps
Use quenching between rounds to build comprehensive maps of processing complexes
Correlate INTS10 localization with RNA maturation states
Functional Assessment Techniques:
Knockdown/Knockout Plus Rescue:
Deplete endogenous INTS10 using siRNA or CRISPR
Rescue with wild-type or mutant INTS10
Use INTS10-FITC antibody to confirm expression patterns of rescue constructs
Measure functional readouts (snRNA processing, transcription rates)
Domain-Specific Function Analysis:
Express truncated INTS10 variants missing specific domains
Visualize localization patterns using INTS10-FITC antibody
Correlate localization changes with functional defects
Systems-Level Integration:
Multi-omics Correlation:
Perform RNA-seq after INTS10 perturbation
Correlate transcriptomic changes with INTS10 localization by immunofluorescence
Integrate with ChIP-seq data to build comprehensive regulatory maps
Protein Interaction Network Mapping:
Use INTS10-FITC for visualization after BioID or APEX proximity labeling
Identify spatial interaction networks through co-localization with candidate proteins
Validate key interactions through co-immunoprecipitation
These methodologies provide comprehensive approaches for investigating INTS10's roles in nuclear processes, leveraging the specificity and direct visualization capabilities of INTS10 Antibody, FITC conjugated .
Before incorporating INTS10 Antibody, FITC conjugated into pivotal experiments, researchers should conduct a series of quality control tests to ensure reliability and reproducibility. This methodological quality control workflow includes:
Antibody Performance Validation:
Titration Analysis:
Test serial dilutions (1:25, 1:50, 1:100, 1:200, 1:400)
Plot signal-to-noise ratio versus concentration
Determine optimal working dilution where signal is maximal with minimal background
Specificity Testing:
Perform peptide competition assay using immunizing peptide
Compare staining pattern with published INTS10 localization data
Run parallel staining with alternative INTS10 antibodies targeting different epitopes
Cross-Reactivity Assessment:
Fluorophore Quality Control:
FITC Conjugation Efficiency:
Measure absorbance at 280nm (protein) and 495nm (FITC)
Calculate fluorophore-to-protein ratio (optimal range typically 2-6 FITC molecules per antibody)
Compare with manufacturer's specifications if provided
Photobleaching Rate Determination:
Expose labeled samples to excitation light at defined intervals
Plot signal intensity versus cumulative exposure time
Calculate half-life of fluorescence signal
Spectral Profile Verification:
Functional Validation Assays:
Positive Control Testing:
Biological Response Validation:
Test antibody performance after cellular treatments known to affect nuclear proteins
Verify expected changes in localization or intensity
Compare results with alternative detection methods (Western blot)
Reproducibility Assessment:
Perform replicate staining across different days
Calculate coefficient of variation between experiments
Establish acceptance criteria for experimental consistency
Storage and Handling Quality Control:
Freeze-Thaw Stability:
Test antibody performance after 0, 1, 3, and 5 freeze-thaw cycles
Measure signal intensity and background at each point
Determine maximum acceptable number of freeze-thaw cycles
Temperature Sensitivity:
Long-term Stability:
Test antibody from the same lot at defined intervals (0, 3, 6, 12 months)
Document any changes in performance metrics
Determine practical shelf-life under laboratory conditions
These quality control procedures should be documented systematically and incorporated into laboratory standard operating procedures to ensure consistent experimental outcomes with INTS10 Antibody, FITC conjugated.
When researchers encounter discrepancies between results obtained with INTS10 Antibody, FITC conjugated and alternative detection methods, a systematic troubleshooting approach is essential. The following methodological framework helps reconcile contradictory findings:
Technical Variance Analysis:
Method-Specific Limitations Assessment:
Immunofluorescence with FITC-conjugated antibodies: Limited to fixed samples, potential artifacts from fixation and permeabilization
Western blot: Detects denatured proteins, may miss conformational epitopes
RNA-based methods: Measure transcript levels, which may not correlate with protein abundance
Mass spectrometry: High specificity but variable sensitivity for different peptides
Epitope Accessibility Evaluation:
Map epitope regions recognized by different detection methods
Assess how sample preparation affects epitope exposure
Consider post-translational modifications that might mask epitopes
Detection Threshold Comparison:
Determine limit of detection for each method
Normalize signals to appropriate standards
Consider relative sensitivity of each technique for INTS10 detection
Experimental Design Reconciliation:
Sample Preparation Harmonization:
Standardize cell lysis conditions across methods
Use identical fixation protocols when possible
Process samples in parallel to minimize batch effects
Biological Context Alignment:
Ensure identical cell types, passages, and growth conditions
Match treatment timing and concentrations precisely
Control for cell cycle phase and confluence level
Quantification Strategy Standardization:
Develop comparable normalization approaches
Use shared reference standards across methods
Apply similar statistical treatments to raw data
Antibody-Specific Investigation:
Epitope Mapping:
Cross-Reactivity Profiling:
Perform immunoprecipitation followed by mass spectrometry
Identify all proteins captured by the antibody
Compare actual versus intended targets
Functionality Assessment:
Determine if FITC conjugation affects antibody binding properties
Test native versus conjugated versions of the same antibody clone
Evaluate impact of storage conditions on performance
Resolution Through Orthogonal Approaches:
Genetic Validation Strategy:
Generate INTS10 knockdown/knockout models
Test all detection methods on these models
Identify which methods accurately reflect INTS10 depletion
Complementary Technique Integration:
Combine immunofluorescence with proximity ligation assay
Validate Western blot findings with immunoprecipitation
Correlate protein levels with mRNA expression
Functional Readout Correlation:
Assess downstream effects of INTS10 perturbation
Determine which detection method best predicts functional outcomes
Use functional data to resolve contradictory expression results
Decision Framework for Results Interpretation:
Preponderance of Evidence Approach:
Weight findings based on methodological rigor
Consider consistency across biological replicates
Prioritize results from complementary techniques showing concordance
Literature Contextualization:
Compare contradictory findings with published studies
Evaluate concordance with established INTS10 biology
Consider novel biological insights that might explain discrepancies
Transparent Reporting:
Document all contradictory findings
Present complete methodological details for each technique
Discuss potential sources of variance in research communications
As molecular biology techniques continue to evolve, INTS10 Antibody, FITC conjugated presents opportunities for integration into emerging research applications. The following methodological approaches represent promising future directions:
Advanced Imaging Technologies:
Lattice Light-Sheet Microscopy:
Track INTS10 dynamics in live cells with minimal phototoxicity
Visualize 3D nuclear organization with isotropic resolution
Capture rapid relocalization events during transcriptional responses
Expansion Microscopy:
Physically expand samples to achieve super-resolution with standard confocal microscopy
Resolve spatial relationships between INTS10 and interaction partners
Map precise subnuclear distributions relative to chromatin domains
Correlative Light and Electron Microscopy (CLEM):
Identify INTS10-positive regions with fluorescence microscopy
Examine ultrastructural context with electron microscopy
Bridge molecular identification with structural biology
Single-Cell Analysis Platforms:
Imaging Mass Cytometry:
Combine INTS10-FITC with metal-labeled antibodies
Profile dozens of proteins simultaneously in tissue sections
Create high-dimensional spatial maps of INTS10 in relation to cellular phenotypes
Spatial Transcriptomics Integration:
Correlate INTS10 protein localization with transcriptional territories
Link protein distribution to local gene expression patterns
Map functional nuclear domains based on combined protein-RNA profiles
Microfluidic-Based Single-Cell Western Blotting:
Analyze INTS10 expression heterogeneity at single-cell resolution
Correlate with other proteins in the Integrator complex
Identify rare cell states with altered INTS10 regulation
Functional Genomics Applications:
CRISPR Screening Visualization:
Map phenotypic consequences of genetic perturbations on INTS10 localization
Screen for factors regulating INTS10 expression or nuclear distribution
Identify synthetic interactions within RNA processing pathways
Targeted Protein Degradation Studies:
Monitor INTS10 depletion kinetics after treatment with PROTACs or dTAGs
Visualize real-time consequences of acute versus chronic INTS10 loss
Correlate degradation timing with functional impairments
Optogenetic Control of Nuclear Organization:
Combine with light-inducible clustering tools to relocalize INTS10
Assess functional consequences of disrupting normal subnuclear distribution
Create spatiotemporal maps of INTS10 function
Clinical Research Applications:
Liquid Biopsy Analysis:
Detect INTS10 in circulating tumor cells or exosomes
Correlate expression with disease progression or treatment response
Develop minimally invasive monitoring approaches
Precision Medicine Approaches:
Profile INTS10 status in patient-derived samples
Correlate with transcriptional dysregulation signatures
Identify potential for targeting RNA processing dependencies
Therapeutic Response Monitoring:
Track changes in INTS10 expression or localization during treatment
Correlate with efficacy of transcription-targeted therapeutics
Develop pharmacodynamic biomarkers based on INTS10 status
These emerging applications represent promising directions for leveraging INTS10 Antibody, FITC conjugated in cutting-edge research, particularly in contexts where direct visualization of INTS10 can provide insights into nuclear organization and function that complement other molecular approaches .
For researchers seeking to adapt INTS10 Antibody, FITC conjugated for specialized applications, the following protocol modifications offer methodological solutions to extend utility beyond standard techniques:
Super-Resolution Microscopy Optimization:
STORM/PALM Imaging Protocol:
Increase antibody concentration (1:25 - 1:50) to maximize labeling density
Add oxygen scavenging system (glucose oxidase/catalase) to buffer
Include thiol compounds (MEA, BME) at 10-100 mM to enhance photoswitching
Acquire 10,000-30,000 frames with low 488nm laser power
Process with localization algorithms (ThunderSTORM, FIJI)
STED Microscopy Adaptation:
Use higher antibody concentration (1:25) to improve signal strength
Mount in ProLong Glass or TDE-based medium to match refractive index
Optimize depletion laser power to balance resolution and photobleaching
Consider using alternative antifade agents compatible with STED
Tissue-Specific Processing Methods:
Formalin-Fixed Paraffin-Embedded (FFPE) Tissues:
Extend antigen retrieval (citrate buffer pH 6.0, 20 minutes)
Add tyramide signal amplification step for low-abundance detection
Increase antibody concentration (1:25 - 1:50) and incubation time (overnight)
Include Sudan Black B treatment (0.1% in 70% ethanol) to reduce autofluorescence
Frozen Tissue Sections:
Optimize fixation (2% PFA, 10 minutes) to preserve nuclear architecture
Increase permeabilization time with 0.5% Triton X-100 (15-20 minutes)
Add 0.1% saponin to antibody dilution buffer to enhance nuclear penetration
Block with mouse/human serum (5%) for highly autofluorescent tissues
Flow Cytometry Enhancements:
High-Throughput Screening Protocol:
Optimize in 96-well format with automated sample processing
Increase cell concentration to 1-2 × 10^6 cells/mL for nuclear proteins
Add 0.1% NP-40 to permeabilization buffer for enhanced nuclear access
Include RNase treatment step to reduce background
Optimize compensation matrix for FITC with spectral overlap corrections
Cell Sorting Applications:
Use higher antibody concentration (1:50) to improve separation resolution
Adjust FSC/SSC gates to account for fixation-induced changes
Implement hierarchical gating strategy (viability → nuclear size → INTS10 intensity)
Add 5 μM DRAQ5 for simultaneous DNA content analysis
Live Cell Adaptation Strategies:
Antibody Delivery Methods:
Fragment antibody to Fab using papain digestion to enhance nuclear penetration
Conjugate with cell-penetrating peptides (TAT, Penetratin)
Optimize electroporation parameters for direct cytoplasmic delivery
Package in cationic lipid nanoparticles for transient transfection
Microinjection Protocol:
Dilute antibody to 0.5-1.0 mg/mL in injection buffer
Add 0.5% glycerol to stabilize during microinjection
Co-inject with fluorescent dextran for injection control
Maintain cells at 37°C throughout injection process
Multiplexed Detection Enhancements:
Sequential Staining Protocol:
Implement cyclic immunofluorescence with antibody stripping
Use 0.1M glycine (pH 2.5) or 6M urea for efficient stripping
Image FITC channels early in the sequence to minimize photobleaching
Register images using nuclear counterstain as fiducial marker
Tyramide Signal Amplification Integration:
Convert direct FITC detection to indirect tyramide-based amplification
Use anti-FITC HRP-conjugated secondary antibody
Develop with tyramide-488 for 5-10 minutes
Inactivate HRP with 1% sodium azide between cycles
These specialized protocol modifications extend the utility of INTS10 Antibody, FITC conjugated beyond standard applications, enabling researchers to address specialized research questions while maintaining specificity for the INTS10 target .