THOC6 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery time may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery times.
Synonyms
THOC6 antibody; WDR58 antibody; PSEC0006 antibody; THO complex subunit 6 homolog antibody; Functional spliceosome-associated protein 35 antibody; fSAP35 antibody; WD repeat-containing protein 58 antibody
Target Names
THOC6
Uniprot No.

Target Background

Function
THOC6 Antibody, FITC conjugated, acts as a component of the THO subcomplex within the TREX complex. This complex is believed to play a crucial role in coordinating mRNA transcription, processing, and nuclear export. Specifically, THOC6 associates with spliced mRNA, not unspliced pre-mRNA. TREX is recruited to spliced mRNAs through a transcription-independent mechanism, binds to mRNA upstream of the exon-junction complex (EJC), and is recruited in a splicing- and cap-dependent manner to a region near the 5' end of the mRNA. This action is essential for mRNA export to the cytoplasm via the TAP/NFX1 pathway. The TREX complex is critical for the export of Kaposi's sarcoma-associated herpesvirus (KSHV) intronless mRNAs and subsequent infectious virus production. THOC6 also plays a role in regulating apoptosis and is involved in brain development.
Gene References Into Functions
  1. Research has identified three unrelated patients with bi-allelic mutations in THOC6. These mutations are associated with intellectual disability and other clinical features. PMID: 27102954
  2. In addition to confirming the pathogenic nature of THOC6 by providing an independent homozygous, apparently loss-of-function allele in a patient with a compatible phenotype, our data further expands the THOC6-related phenotype to include previously unreported imperforate anus and undescended testicles. PMID: 26739162
Database Links

HGNC: 28369

OMIM: 613680

KEGG: hsa:79228

STRING: 9606.ENSP00000326531

UniGene: Hs.412304

Involvement In Disease
Beaulieu-Boycott-Innes syndrome (BBIS)
Protein Families
WD repeat THOC6 family
Subcellular Location
Nucleus. Nucleus speckle.

Q&A

What is THOC6 and what cellular functions does it perform?

THOC6 (THO Complex Subunit 6 homolog) is a component of the THO subcomplex within the TREX (transcription/export) complex. It plays a critical role in coupling mRNA transcription, processing, and nuclear export. THOC6 functions to tether TREX dimers to form tetramers that permit interaction with auxiliary molecules such as ALYREF and CHTOP . Through its WD40 domains, THOC6 interacts with THOC5 to facilitate tetramerization, which is essential for proper TREX complex function . Notably, THOC6 is positioned at the TREX core interface, directly heterodimerizing and homodimerizing with THOC5 and THOC6, respectively . Beyond its role in mRNA processing, THOC6 has been implicated in negative control of apoptosis involved in brain development, highlighting its importance in neural tissue development .

How are THOC6 FITC-conjugated antibodies typically characterized?

THOC6 FITC-conjugated antibodies undergo rigorous characterization to ensure specificity and functionality. These antibodies are typically tested in multiple applications including Western blot, immunoprecipitation, and immunohistochemistry across various cell lines and tissue samples . For example, THOC6 antibodies have demonstrated positive Western blot detection in A431 cells, mouse ovary tissue, and Neuro-2a cells . Immunoprecipitation validation is often performed in cell lines such as A431 cells, while immunohistochemistry testing may utilize tissues like human ovary tumor samples . Antibody purity is assessed through methods like protein G chromatography, with high-quality antibodies typically achieving >95% purity . The reactivity profile is established against target species (commonly human, mouse, and rat), and epitope mapping identifies the specific amino acid sequences recognized by the antibody, such as amino acids 56-315 of human THOC6 .

What are the optimal experimental conditions for immunofluorescence using THOC6 FITC-conjugated antibodies?

For optimal immunofluorescence results with THOC6 FITC-conjugated antibodies, consider the following protocol:

  • Cell preparation: Culture cells on coverslips or appropriate imaging chambers until they reach 50-70% confluence .

  • Fixation: Remove growth medium and wash cells twice with PBS. Fix cells with methanol at -20°C for 5-10 minutes, which is often preferred for nuclear proteins like THOC6 .

  • Permeabilization: If not using methanol fixation, permeabilize cells with 0.1-0.5% Triton X-100 in PBS for 5-10 minutes at room temperature.

  • Blocking: Add 2 mL of blocking solution (PBS containing 10% fetal bovine serum) and incubate for 20 minutes at room temperature to reduce non-specific binding .

  • Antibody incubation: Remove blocking solution and add 1 mL of PBS/10% FBS containing THOC6 FITC-conjugated antibody at a 1:500 dilution. Incubate for 1 hour at room temperature in the dark .

  • Washing: Wash cells 2-3 times with PBS for 5 minutes each .

  • Mounting: Mount with appropriate anti-fade medium containing DAPI for nuclear counterstaining.

  • Visualization: Observe using a fluorescence microscope with appropriate FITC filter sets (excitation ~495 nm, emission ~520 nm) .

Key considerations include protecting the antibody and samples from light exposure throughout the procedure and empirically determining the optimal antibody dilution for specific cell types or tissues, as this may vary from the recommended 1:500 dilution .

How can THOC6 FITC-conjugated antibodies be used to study mRNA processing defects?

THOC6 FITC-conjugated antibodies are valuable tools for investigating mRNA processing defects, particularly in the context of THOC6 mutations or dysfunction. Researchers can employ these antibodies in several approaches:

  • Co-localization studies: Use THOC6 FITC-conjugated antibodies in conjunction with other fluorescently labeled markers of mRNA processing components to assess potential disruptions in spatial organization. This approach can reveal whether THOC6 variants affect its localization to nuclear speckles where mRNA processing occurs .

  • TREX complex integrity assessment: Combine immunofluorescence with co-immunoprecipitation assays to determine whether THOC6 variants disrupt interactions with other TREX components like THOC5. THOC6 FITC-conjugated antibodies can visualize the co-localization patterns, while biochemical approaches confirm physical interactions .

  • Analysis of splicing defects: In studies of THOC6-related disorders, THOC6 FITC-conjugated antibodies can help assess whether THOC6 variants alter the recruitment of splicing factors. Research has shown that THOC6 loss of function leads to mRNA mis-splicing rather than export defects in neural tissue .

  • Developmental studies: When investigating THOC6's role in brain development, these antibodies can be used to track THOC6 expression patterns during neurogenesis and correlate its distribution with markers of neural progenitor cells or differentiated neurons .

  • Rescue experiments: In cells expressing THOC6 variants, researchers can attempt functional rescue with wild-type THOC6 and use the FITC-conjugated antibodies to confirm proper localization and restoration of TREX tetramer formation .

This multi-faceted approach allows researchers to connect THOC6 molecular functions with observed cellular phenotypes and disease mechanisms.

What controls should be included when using THOC6 FITC-conjugated antibodies?

When conducting experiments with THOC6 FITC-conjugated antibodies, including appropriate controls is essential for result validation and interpretation:

  • Negative controls:

    • Isotype control: Use a FITC-conjugated IgG of the same isotype (e.g., rabbit IgG for rabbit polyclonal THOC6 antibodies) to assess non-specific binding .

    • Unstained samples: Include samples without any antibody to establish autofluorescence baseline.

    • Secondary-only control (for indirect methods): Apply only the secondary antibody to detect non-specific binding.

    • THOC6 knockout/knockdown cells: If available, use cells with validated THOC6 depletion to confirm antibody specificity .

  • Positive controls:

    • Cell lines with confirmed THOC6 expression: A431 cells and Neuro-2a cells have been validated for THOC6 expression and can serve as positive controls .

    • Tissue samples: Human ovary tumor tissue has demonstrated positive THOC6 immunoreactivity .

    • Recombinant THOC6: Purified or overexpressed THOC6 protein can provide a strong positive signal.

  • Technical controls:

    • Titration series: Test multiple antibody dilutions to optimize signal-to-noise ratio.

    • Antigen competition: Pre-incubate the antibody with purified THOC6 protein to demonstrate binding specificity.

    • Multiple detection methods: Validate findings using complementary techniques like Western blotting or immunoprecipitation .

  • Biological controls:

    • Wild-type vs. variant THOC6: When studying THOC6 variants, include wild-type samples for comparison .

    • Tissue/developmental controls: When examining THOC6 in development, include samples from multiple developmental stages to track expression changes .

How can I minimize photobleaching of FITC-conjugated THOC6 antibodies during imaging?

FITC is particularly susceptible to photobleaching, which can significantly impact experimental outcomes when using FITC-conjugated THOC6 antibodies. To minimize this issue, implement the following strategies:

  • Sample preparation:

    • Use high-quality anti-fade mounting media containing anti-photobleaching agents like p-phenylenediamine or ProLong Gold .

    • Store slides in the dark at 4°C when not being imaged.

    • For live-cell imaging, supplement media with anti-oxidants like vitamin C or OxyFluor.

  • Microscope settings:

    • Reduce excitation light intensity to the minimum needed for adequate signal detection.

    • Use neutral density filters to attenuate excitation light.

    • Minimize exposure time and use binning to reduce the total light dose.

    • Employ confocal microscopy with appropriate pinhole settings to reduce out-of-focus light exposure.

  • Acquisition strategies:

    • Focus on samples using differential interference contrast (DIC) or phase contrast before switching to fluorescence.

    • Capture images of FITC channels first in multi-channel experiments, as FITC bleaches more rapidly than many other fluorophores.

    • Use deconvolution software to enhance signal from lower-intensity images.

    • Consider automated acquisition systems that minimize the time samples are exposed to excitation light.

  • Alternative approaches:

    • For samples requiring extended imaging, consider using more photostable fluorophores like Alexa Fluor 488 as alternatives to FITC .

    • For time-lapse experiments, implement time-interval acquisition to reduce total light exposure.

By implementing these strategies, researchers can significantly extend the fluorescence lifetime of FITC-conjugated THOC6 antibodies and improve the quality and reproducibility of imaging data.

What are the common pitfalls in antibody validation for THOC6 FITC-conjugated antibodies?

Proper validation of THOC6 FITC-conjugated antibodies is crucial for generating reliable experimental results. Researchers should be aware of these common pitfalls and their solutions:

  • Cross-reactivity with related proteins:

    • Pitfall: The THO complex contains multiple subunits with similar structures that may cross-react with THOC6 antibodies.

    • Solution: Perform specificity testing using Western blots comparing wild-type samples with THOC6 knockdown/knockout samples. Verify a single band at the expected molecular weight (approximately 35-38 kDa for THOC6) .

  • Epitope masking in fixed tissues/cells:

    • Pitfall: Certain fixation methods may mask the THOC6 epitope, particularly if the antibody targets regions involved in protein-protein interactions.

    • Solution: Compare multiple fixation protocols (e.g., paraformaldehyde, methanol, acetone) and antigen retrieval methods. For THOC6, TE buffer pH 9.0 has been suggested for antigen retrieval in IHC applications .

  • Batch-to-batch variation:

    • Pitfall: Different antibody lots may show varying levels of specificity and sensitivity.

    • Solution: Maintain reference samples from previous successful experiments to validate new antibody lots. Consider purchasing larger quantities of a single lot for long-term studies.

  • Insufficient controls:

    • Pitfall: Relying solely on staining patterns without proper controls can lead to misinterpretation.

    • Solution: Include biological controls (cells/tissues with altered THOC6 expression) and technical controls (isotype, secondary-only) in each experiment .

  • FITC conjugation efficiency:

    • Pitfall: Inconsistent FITC conjugation can affect signal strength and reproducibility.

    • Solution: Verify the fluorophore-to-protein ratio (typically 3-5 FITC molecules per antibody is optimal). Test each new batch with standardized samples to ensure comparable performance.

  • Overlooking species-specific differences:

    • Pitfall: THOC6 shows some variation across species, which may affect antibody binding.

    • Solution: Confirm the antibody's reactivity with your species of interest. Current FITC-conjugated THOC6 antibodies primarily target human THOC6, with some cross-reactivity to mouse and rat .

By addressing these potential pitfalls, researchers can significantly improve the reliability and reproducibility of experiments using THOC6 FITC-conjugated antibodies.

How can THOC6 FITC-conjugated antibodies be used to investigate TREX complex dynamics in neural development?

Recent research has revealed THOC6's critical role in neural development, making THOC6 FITC-conjugated antibodies valuable tools for investigating TREX complex dynamics in this context. Here's a methodological approach:

  • Developmental time-course analysis:
    Use THOC6 FITC-conjugated antibodies to track expression patterns across neural development stages. Research has shown that THOC6 plays a role in regulating the transition from proliferative to neurogenic divisions during corticogenesis . Create quantitative spatiotemporal maps of THOC6 expression in developing neural tissue, correlating with markers of neural progenitors (PAX6) and post-mitotic neurons.

  • TREX complex assembly visualization:
    Combine THOC6 FITC-conjugated antibodies with differentially labeled antibodies against other TREX components (THOC1, THOC2, THOC5) to assess co-localization patterns during neural development. This approach can reveal whether TREX complex composition changes during neurogenesis and whether THOC6-dependent tetramerization varies across developmental stages .

  • Co-immunoprecipitation with imaging validation:
    Perform co-immunoprecipitation of THOC5 and other TREX components, followed by immunofluorescence validation with THOC6 FITC-conjugated antibodies. This combined approach can determine whether specific neural differentiation states correlate with changes in THOC6's interaction with THOC5 and auxiliary proteins like ALYREF .

  • RNA processing visualization:
    Combine THOC6 immunofluorescence with RNA FISH (Fluorescence In Situ Hybridization) targeting specific transcripts known to be mis-spliced in THOC6-deficient neural tissue. This method can provide spatial information about where in the nucleus these processing defects occur relative to THOC6 localization.

  • High-resolution imaging techniques:
    Employ super-resolution microscopy (STED, STORM, SIM) with THOC6 FITC-conjugated antibodies to resolve the nanoscale organization of TREX complexes in neural progenitor cells versus differentiated neurons, potentially revealing structural reorganizations during differentiation.

This multi-dimensional approach allows researchers to connect molecular-level TREX complex dynamics with cellular-level neural development processes, providing deeper insights into how THOC6 dysfunction leads to neurodevelopmental disorders .

How do THOC6 variants associated with neurodevelopmental disorders affect TREX tetramer formation and function?

THOC6 variants have been implicated in neurodevelopmental disorders such as TIDS (THOC6-related Intellectual Disability Syndrome). THOC6 FITC-conjugated antibodies can be instrumental in elucidating the molecular mechanisms of these disorders through several methodological approaches:

  • Structural impact analysis:
    Use THOC6 FITC-conjugated antibodies to visualize the nuclear localization patterns of wild-type versus variant THOC6 proteins. Research has shown that pathogenic THOC6 variants, particularly those affecting the WD40 domains, can disrupt THOC6's ability to bind THOC5, preventing TREX tetramer formation while leaving TREX dimers intact . Compare the co-localization patterns of THOC6 variants with THOC5 and other TREX components using high-resolution microscopy.

  • Functional domain mapping:
    Employ a panel of THOC6 variants with mutations in different domains to determine which regions are critical for tetramer formation. FITC-conjugated antibodies can help visualize whether specific variants alter nuclear speckle localization or protein stability. Research has demonstrated that variants like p.E188K completely disrupt THOC5-THOC6 interaction .

  • Quantitative protein-protein interaction analysis:
    Combine fluorescence resonance energy transfer (FRET) techniques with THOC6 FITC-conjugated antibodies to quantitatively measure the proximity of THOC6 to other TREX components in living cells expressing different THOC6 variants. This approach can provide dynamic information about how mutations affect complex assembly in real-time.

  • Rescue experiments:
    In cells expressing THOC6 variants, attempt phenotypic rescue with wild-type THOC6 and use FITC-conjugated antibodies to confirm proper localization and restoration of TREX tetramer formation. Quantify the degree of rescue by measuring ALYREF association with the TREX complex, which has been shown to be reduced in cells with THOC6 variants .

  • Developmental timing analysis:
    Investigate whether THOC6 variants affect the timing of neural differentiation by correlating THOC6 localization with markers of cell cycle progression and differentiation. This approach can help explain how THOC6 dysfunction leads to the specific neurodevelopmental phenotypes observed in patients .

These methodologies collectively provide a comprehensive framework for understanding how THOC6 variants disrupt normal neurogenesis at the molecular level, potentially identifying targets for therapeutic intervention.

What approaches can be used to investigate the role of THOC6 in glioma progression and potential therapeutic applications?

Recent research has identified THOC6 as a novel biomarker and potential therapeutic target in glioma, with high expression correlating with poor prognosis . THOC6 FITC-conjugated antibodies can be leveraged to investigate this connection through several sophisticated approaches:

  • Expression pattern analysis in glioma specimens:
    Use THOC6 FITC-conjugated antibodies for comprehensive immunofluorescence profiling of glioma tissue microarrays spanning different grades, histological subtypes, and IDH mutation status. Quantitative analysis has shown that THOC6 expression correlates with multiple clinical features and is significantly higher in gliomas compared to normal brain tissue (P < 0.001) . Co-stain with markers of glioma stem cells to determine if THOC6 is specifically elevated in the tumor-initiating population.

  • Functional studies in patient-derived models:
    Establish patient-derived xenografts or organoids from glioma specimens with varying THOC6 expression levels. Use THOC6 FITC-conjugated antibodies to track expression changes during tumor progression and in response to treatments. Correlate expression patterns with tumor growth dynamics and invasion capabilities.

  • Mechanistic interrogation of signaling pathways:
    Combine THOC6 immunofluorescence with phospho-specific antibodies against components of interleukin signaling, Rho GTPase signaling, and DNA repair pathways, which enrichment analysis has linked to THOC6 high expression phenotypes in glioma . This approach can reveal how THOC6 integrates with these oncogenic pathways.

  • Immuno-therapeutic targeting validation:
    Investigate the potential of THOC6 as an immunotherapy target by assessing its cell surface accessibility in live glioma cells using non-permeabilized immunofluorescence protocols with FITC-conjugated antibodies. Research has shown that THOC6 expression in glioma is closely related to immune cell infiltration levels .

  • Drug-target interaction studies:
    Use molecular docking simulations in conjunction with experimental verification to study the interaction between THOC6 and potential anti-glioma drugs. Research has demonstrated strong molecular docking energy between several glioma drugs and THOC6, suggesting it might be a direct therapeutic target . FITC-conjugated antibodies can help validate these interactions by assessing changes in THOC6 localization or abundance following drug treatment.

  • Theranostic application development:
    Explore the potential of THOC6 FITC-conjugated antibodies themselves as theranostic agents for both imaging and targeted therapy. The high diagnostic value of THOC6 in glioma (AUC = 0.915 in ROC curve analysis) makes it a promising candidate for development of antibody-drug conjugates or imaging agents .

These methodological approaches provide a comprehensive framework for understanding THOC6's role in glioma biology and developing targeted therapeutic strategies.

What are the optimal storage and handling conditions for maintaining THOC6 FITC-conjugated antibody activity?

Proper storage and handling of THOC6 FITC-conjugated antibodies are critical for maintaining their activity and fluorescence properties. Follow these detailed guidelines:

  • Storage temperature:

    • Store antibody aliquots at -20°C for long-term preservation .

    • Avoid repeated freeze/thaw cycles by preparing small working aliquots (10-20 μL) .

    • Do not store at 4°C for extended periods (>1 week) as this can lead to gradual loss of activity.

  • Buffer conditions:

    • Optimal storage buffer typically contains PBS (pH 7.4), 0.03% Proclin-300 (or 0.01% sodium azide), and 50% glycerol as a cryoprotectant .

    • If diluting for immediate use, use fresh PBS containing 1-2% BSA or 10% FBS to maintain antibody stability and prevent non-specific binding .

  • Light protection:

    • FITC is particularly sensitive to photobleaching; store all FITC-conjugated antibodies in amber or foil-wrapped tubes .

    • Minimize exposure to light during all handling steps including thawing, aliquoting, and experimental procedures .

    • Work under subdued lighting conditions when preparing FITC-conjugated antibody solutions.

  • Contamination prevention:

    • Use sterile technique when handling antibody stocks to prevent microbial contamination.

    • Add sterile-filtered sodium azide (0.01% final concentration) for additional protection against microbial growth.

    • Never use antibodies showing visible signs of contamination (turbidity, unusual color).

  • Thawing protocol:

    • Thaw frozen aliquots rapidly at room temperature followed by brief centrifugation to collect contents.

    • Once thawed, keep on ice and protected from light until ready to use.

    • Return unused portions to -20°C promptly; do not refreeze thawed antibody more than once.

  • Shipping considerations:

    • THOC6 FITC-conjugated antibodies can typically be shipped at ambient temperature with appropriate light protection .

    • Upon receipt, promptly transfer to -20°C storage.

    • No dry ice shipping is typically required .

  • Stability monitoring:

    • Monitor antibody performance over time by including positive control samples in each experiment.

    • If decreased signal is observed, this may indicate antibody degradation requiring a fresh aliquot.

Following these detailed handling procedures will maximize the shelf-life and performance consistency of THOC6 FITC-conjugated antibodies in research applications.

How can I establish the optimal working dilution for THOC6 FITC-conjugated antibodies in different experimental systems?

Determining the optimal working dilution for THOC6 FITC-conjugated antibodies requires a systematic approach to balance specific signal with minimal background across different experimental platforms:

  • Initial titration series:

    • For immunofluorescence applications, start with a broad dilution range (e.g., 1:100, 1:250, 1:500, 1:1000, 1:2000) in blocking buffer (PBS with 10% FBS) .

    • For each dilution, process identical samples following your standard protocol, maintaining consistent exposure settings during imaging.

    • Evaluate signal-to-noise ratio quantitatively by measuring fluorescence intensity in known THOC6-positive regions versus background areas.

  • Application-specific considerations:

    • For immunofluorescence: Recommended starting dilution is 1:500 in PBS containing 10% FBS .

    • For Western blotting: Typical starting range is 1:500-1:2000 .

    • For immunoprecipitation: Use 0.5-4.0 μg antibody per 1.0-3.0 mg of total protein lysate .

    • For immunohistochemistry: Begin with 1:20-1:200 dilution range .

  • Fine-tuning:

    • Once an approximate optimal range is identified, perform a narrower titration (e.g., 1:400, 1:500, 1:600) to pinpoint the ideal concentration.

    • The optimal dilution provides the strongest specific signal with minimal background fluorescence.

    • Document imaging parameters (exposure time, gain settings) along with antibody dilution for reproducibility.

  • Cell/tissue type optimization:

    • Different sample types may require adjusted dilutions due to variations in target abundance and accessibility.

    • For human samples, verify reactivity specifically with human THOC6 .

    • For cross-species applications, validate reactivity with mouse or rat samples if using those model systems .

  • Fixation method considerations:

    • If using paraformaldehyde fixation, slightly higher antibody concentrations may be needed compared to methanol fixation.

    • For paraffin-embedded tissues, antigen retrieval methods significantly impact optimal antibody dilution. Test both TE buffer pH 9.0 and citrate buffer pH 6.0 for THOC6 antibodies .

  • Quantitative assessment:

    • Use image analysis software to plot signal-to-noise ratios across dilutions.

    • The inflection point where additional antibody concentration no longer significantly improves specific signal represents the optimal working dilution.

  • Validation across lots:

    • Repeat titration with new antibody lots, as conjugation efficiency may vary.

    • Maintain reference samples from successful experiments to calibrate new antibody preparations.

This methodical approach ensures consistent, reproducible results while minimizing antibody consumption and experimental artifacts.

What experimental design is optimal for comparing normal and pathological THOC6 expression patterns in neural tissues?

To rigorously compare normal and pathological THOC6 expression patterns in neural tissues, researchers should implement a comprehensive experimental design that addresses biological variability while maximizing technical consistency:

  • Sample selection and preparation:

    • Normal tissues: Include samples from multiple developmental stages (embryonic, fetal, postnatal, adult) to capture age-dependent THOC6 expression patterns .

    • Pathological tissues: Select specimens representing different conditions where THOC6 dysfunction has been implicated (neurodevelopmental disorders, gliomas) .

    • Processing consistency: Use identical fixation protocols across all samples (4% paraformaldehyde followed by either frozen sectioning or paraffin embedding with standardized antigen retrieval).

    • Section orientation: For developmental studies, prepare both coronal and sagittal sections to comprehensively map THOC6 distribution across neuroanatomical regions.

  • Staining protocol optimization:

    • Multiplex immunofluorescence design:

      • THOC6 FITC-conjugated antibody (green channel)

      • Neural progenitor markers (PAX6, Nestin) in red channel

      • Post-mitotic neuronal markers (TUJ1, NeuN) in far-red channel

      • Nuclear counterstain (DAPI) in blue channel

    • Technical controls:

      • Include secondary-only controls for each fluorophore

      • Process serial sections with isotype control antibodies

      • Include positive control tissues with known THOC6 expression (e.g., A431 cells, human ovary tissue)

  • Quantitative image acquisition:

    • Standardized microscopy settings:

      • Use identical exposure parameters across all samples

      • Acquire z-stacks (0.5-1μm step size) to capture the full tissue depth

      • Include reference fluorescence standards in each imaging session

    • Comprehensive sampling strategy:

      • For each specimen, image multiple fields (minimum 5-10) across relevant neuroanatomical regions

      • Use random sampling approaches to avoid selection bias

      • For developmental studies, establish consistent anatomical landmarks to ensure comparable regions are analyzed across ages

  • Analytical approach:

    • Quantitative metrics:

      • Measure THOC6 fluorescence intensity at cellular and subcellular levels

      • Quantify co-localization coefficients between THOC6 and nuclear speckle markers

      • Analyze nuclear vs. cytoplasmic distribution ratios

    • Cell-type specific analysis:

      • Segregate measurements based on co-expression with cell-type specific markers

      • Compare THOC6 levels in proliferating vs. post-mitotic populations

    • Statistical analysis:

      • Use hierarchical linear mixed models to account for nested data structure

      • Implement false discovery rate correction for multiple comparisons

      • Validate key findings with orthogonal techniques (Western blotting, qPCR)

  • Validation in experimental models:

    • In vitro validation:

      • Compare THOC6 localization patterns in neural progenitor cultures derived from control vs. patient iPSCs

      • Track THOC6 expression changes during directed neuronal differentiation

    • In vivo validation:

      • Utilize conditional Thoc6 knockout mouse models targeting specific neural populations

      • Confirm antibody specificity using tissue from these models

This comprehensive experimental design enables robust comparison of THOC6 expression patterns across normal development and pathological conditions while controlling for technical variables that could confound interpretation.

How can multi-omics approaches incorporate THOC6 FITC-conjugated antibodies to understand RNA processing in development and disease?

Integrating THOC6 FITC-conjugated antibodies into multi-omics frameworks offers powerful opportunities to understand the complex role of THOC6 in RNA processing across developmental and disease contexts:

  • Spatial transcriptomics integration:

    • Combine THOC6 immunofluorescence with in situ sequencing or Visium spatial transcriptomics to correlate THOC6 protein distribution with transcriptome-wide expression patterns.

    • This approach can reveal spatial relationships between THOC6 localization and specific mRNA processing events, particularly in developing neural tissues where THOC6 regulates the transition from proliferative to neurogenic divisions .

    • Method: Perform THOC6 immunofluorescence imaging, record coordinates, then conduct spatial transcriptomics on the same tissue section, allowing computational integration of protein localization with transcriptome data.

  • ChIP-seq and CLIP-seq correlation:

    • Use THOC6 antibodies for Chromatin Immunoprecipitation sequencing (ChIP-seq) to identify genomic regions where THOC6 associates with chromatin during transcription.

    • Complement with Cross-Linking Immunoprecipitation sequencing (CLIP-seq) to identify THOC6-bound RNA targets.

    • Correlate these datasets to construct comprehensive maps of how THOC6 coordinates transcription with RNA processing.

    • This approach can identify specific gene sets whose expression depends on THOC6-mediated TREX tetramer formation .

  • Single-cell multi-modal analysis:

    • Apply THOC6 FITC-conjugated antibodies in CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing) workflows to simultaneously profile THOC6 protein levels and transcriptomes in thousands of individual cells.

    • This approach can reveal cell type-specific THOC6 functions and identify populations particularly vulnerable to THOC6 dysfunction.

    • In glioma research, this method could stratify tumor cells based on THOC6 expression and correlate with transcriptional programs driving malignancy .

  • Proteome-wide interaction networks:

    • Use proximity-dependent biotin labeling (BioID or TurboID) with THOC6 as the bait protein to identify its complete interactome.

    • Validate key interactions with co-immunofluorescence using THOC6 FITC-conjugated antibodies.

    • Map how these interaction networks change during neural development or in response to stress conditions.

    • This approach can expand our understanding beyond known TREX components like THOC5 to discover novel THOC6 partners .

  • Functional genomics integration:

    • Combine CRISPR screens targeting RNA processing factors with THOC6 immunofluorescence to identify genetic interactions.

    • In parallel, perform transcriptome-wide analysis of splicing patterns to connect genetic perturbations with specific RNA processing outcomes.

    • This approach can systematically map the RNA processing network dependent on THOC6 function.

By integrating these multi-omics approaches with THOC6 FITC-conjugated antibody applications, researchers can construct comprehensive models of how THOC6 dysfunction leads to specific developmental disorders or contributes to cancer progression .

What emerging technologies might enhance the utility of THOC6 FITC-conjugated antibodies in basic and translational research?

Several cutting-edge technologies are poised to dramatically expand the applications of THOC6 FITC-conjugated antibodies in both fundamental mechanistic studies and translational medicine:

  • Super-resolution microscopy advancements:

    • Implementation of expansion microscopy (ExM) with THOC6 FITC-conjugated antibodies can physically expand samples to achieve ~70 nm resolution using standard confocal microscopy.

    • This approach could reveal nanoscale organization of TREX complexes and distinguish between dimeric and tetrameric TREX formations that are critical to understanding THOC6 function .

    • Lattice light-sheet microscopy combined with adaptive optics enables high-speed, low-phototoxicity 3D imaging of THOC6 dynamics in living cells or tissue, potentially revealing previously unobservable kinetics of TREX assembly.

  • Optogenetic protein control systems:

    • Development of light-inducible THOC6 degradation systems where THOC6 is tagged with photosensitive domains allowing precise spatiotemporal control of THOC6 levels.

    • FITC-conjugated antibodies would serve as validation tools to confirm localized protein depletion.

    • This technology would enable unprecedented studies of acute THOC6 loss in specific subcellular regions or cell populations within developing neural tissues.

  • Microfluidic tissue models:

    • Integration of THOC6 immunofluorescence with organ-on-chip technologies modeling the developing human cortex.

    • This approach allows controlled manipulation of signaling pathways while monitoring THOC6 expression and localization in real-time.

    • Particularly valuable for studying how THOC6 regulates the transition from proliferative to neurogenic divisions during corticogenesis .

  • AI-enhanced image analysis:

    • Deep learning algorithms trained on THOC6 immunofluorescence patterns could automatically identify subtle alterations in subcellular localization not detectable by conventional analysis.

    • This approach could classify THOC6 expression patterns in glioma samples to predict patient outcomes, building on findings that THOC6 expression correlates with prognosis (AUC = 0.915) .

    • Federated learning approaches could integrate THOC6 immunofluorescence data across multiple research institutions while preserving data privacy.

  • Nanobody and aptamer alternatives:

    • Development of FITC-conjugated nanobodies or aptamers against THOC6 would provide smaller probes with superior tissue penetration and reduced immunogenicity.

    • These alternative binding molecules could access epitopes in crowded nuclear environments that are inaccessible to conventional antibodies.

    • Particularly valuable for studying THOC6 in the context of dense ribonucleoprotein complexes.

  • Theranostic applications:

    • Dual-function THOC6 antibodies conjugated with both imaging agents and therapeutic payloads could simultaneously visualize and treat gliomas with high THOC6 expression.

    • This approach leverages findings that THOC6 is a potential target for anti-glioma drugs and has strong diagnostic value .

    • Could enable personalized treatment monitoring where therapy is guided by THOC6 expression patterns.

These emerging technologies promise to transform how researchers utilize THOC6 FITC-conjugated antibodies, potentially accelerating the translation of basic THOC6 biology into clinical applications for neurodevelopmental disorders and glioma treatment.

What are the key unresolved questions about THOC6 function that could be addressed using FITC-conjugated antibodies?

Despite significant advances in understanding THOC6 biology, several crucial questions remain unresolved that could be specifically addressed using THOC6 FITC-conjugated antibodies:

  • Developmental regulation of TREX complex composition:

    • How does THOC6 expression and localization change across neural development stages?

    • Is THOC6-mediated TREX tetramer formation differentially regulated during the transition from neural progenitor proliferation to differentiation?

    • FITC-conjugated antibodies would enable high-resolution temporal mapping of THOC6 dynamics during corticogenesis, potentially revealing stage-specific functions .

  • Cell type-specific roles of THOC6:

    • Does THOC6 have unique functions in specific neural cell types that explain the selective vulnerability in neurodevelopmental disorders?

    • Are there differences in THOC6 expression or TREX complex composition between neural stem cells, progenitors, and differentiated neuronal subtypes?

    • FITC-conjugated antibodies combined with cell type-specific markers would allow systematic characterization of THOC6 across neural lineages.

  • Subcellular dynamics and trafficking:

    • How does THOC6 traffic between different nuclear compartments in response to cellular stress or developmental signals?

    • What is the half-life and turnover rate of THOC6 in different cellular contexts?

    • Live-cell imaging using FITC-conjugated anti-THOC6 nanobodies could reveal these dynamic properties.

  • Structure-function relationships of THOC6 variants:

    • How do specific THOC6 mutations found in patients differentially affect interaction with other TREX components and auxiliary proteins?

    • Do different mutations create distinct molecular phenotypes that correlate with clinical severity?

    • FITC-conjugated antibodies could visualize how different variants affect nuclear localization patterns and co-localization with TREX partners .

  • Mechanistic basis of THOC6 elevation in glioma:

    • What upstream regulatory mechanisms drive THOC6 overexpression in gliomas?

    • Does THOC6 directly contribute to tumorigenesis or represent a compensatory response?

    • What specific RNA processing events mediated by THOC6 promote glioma progression?

    • FITC-conjugated antibodies could track THOC6 expression during glioma evolution and in response to therapeutic interventions .

  • Interactome dynamics:

    • Beyond the core TREX components, what other proteins interact with THOC6 in a context-dependent manner?

    • How do these interactions change during development or in disease states?

    • FITC-conjugated antibodies could be used in proximity ligation assays to map interaction networks in situ.

  • RNA target specificity:

    • Does THOC6 show preference for specific RNA sequences or structures?

    • Are THOC6-dependent RNA processing events transcript-specific or global?

    • Combining THOC6 FITC immunofluorescence with RNA FISH could reveal spatial relationships between THOC6 and its target transcripts.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.