GTF3C5 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Phosphate Buffered Saline (PBS) containing 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days after receiving them. Delivery times may vary depending on the purchasing method or location. For specific delivery details, please contact your local distributor.
Synonyms
FLJ20857 antibody; General transcription factor 3C polypeptide 5 antibody; General transcription factor IIIC polypeptide 5 63kDa antibody; GTF3C5 antibody; TF3C epsilon antibody; TF3C-epsilon antibody; TF3C5_HUMAN antibody; TFIIIC 63 kDa subunit antibody; TFiiiC2 63 antibody; TFIIIC63 antibody; TFIIICepsilon antibody; Transcription factor IIIC 63 kDa subunit antibody; Transcription factor IIIC epsilon subunit antibody; Transcription factor IIIC subunit epsilon antibody
Target Names
GTF3C5
Uniprot No.

Target Background

Function
GTF3C5 Antibody is involved in RNA polymerase III-mediated transcription. It is an integral, tightly associated component of the DNA-binding TFIIIC2 subcomplex that directly binds to tRNA and virus-associated RNA promoters.
Database Links

HGNC: 4668

OMIM: 604890

KEGG: hsa:9328

STRING: 9606.ENSP00000361180

UniGene: Hs.495417

Protein Families
TFIIIC subunit 5 family
Subcellular Location
Nucleus.

Q&A

Basic Research Questions About GTF3C5 Antibody

For maximum shelf life and performance, GTF3C5 antibodies should be stored according to these guidelines:

  • Store at -20°C for most commercial preparations

  • Aliquot to avoid repeated freeze-thaw cycles that can degrade antibody performance

  • Some preparations contain glycerol (typically 50%) and sodium azide (0.02%) as preservatives

  • Most antibodies remain stable for one year after shipment when properly stored

  • For smaller quantities (e.g., 20μl), some formulations include 0.1% BSA as a stabilizer

Proper storage is critical as antibody degradation can lead to increased background signal, reduced sensitivity, and false results in experimental applications .

How do I troubleshoot non-specific bands in Western blots when using GTF3C5 antibodies?

Non-specific bands are a common challenge when working with GTF3C5 antibodies. The following methodological approach can help resolve these issues:

  • Optimize antibody concentration: Titrate the antibody dilution from 1:500 to 1:2400 to determine optimal signal-to-noise ratio . Based on validation data, 0.1 μg/ml has produced clean results with human and mouse samples .

  • Sample preparation considerations:

    • Use NETN lysis buffer for whole cell lysates as validated in established protocols

    • Load appropriate protein amounts (50 μg has been validated in HeLa, HEK293T, and NIH 3T3 cells)

  • Blocking optimization:

    • Try different blocking agents (5% non-fat milk vs. BSA)

    • Extend blocking time to reduce non-specific binding

  • Control experiments:

    • Run a known positive control (HeLa or NIH 3T3 lysates have been validated)

    • Include a peptide competition assay to confirm specificity

  • Detection system:

    • For chemiluminescent detection, short exposure times (10 seconds) have been effective

    • Consider using secondary antibodies with minimal cross-reactivity to the species of your samples

The expected molecular weight for GTF3C5 is 63 kDa, which should be used as the primary reference point when evaluating band specificity .

What is the relationship between GTF3C5 mutations and human disease, and how can antibodies be used to study disease mechanisms?

Recent research has identified biallelic variants in GTF3C5 associated with several clinical manifestations including hypomelanosis of Ito, seizures, and growth abnormalities . Researchers investigating these disease mechanisms can employ GTF3C5 antibodies with these methodological approaches:

  • Comparative expression analysis:

    • Western blotting to quantify protein levels in patient-derived cells versus controls

    • Immunohistochemistry in relevant tissues (brain, skin) to examine spatial distribution changes

  • Functional impact assessment:

    • Immunoprecipitation to examine protein-protein interactions that may be disrupted by mutations

    • ChIP assays to determine if DNA binding properties are altered

  • Disease model validation:

    • Use IHC with GTF3C5 antibodies in animal models of related disorders

    • Verify antibody cross-reactivity between human and model organisms (mouse and rat reactivity has been confirmed)

  • Therapeutic development:

    • Monitor GTF3C5 levels or localization in response to candidate therapeutics

    • Establish assays using these antibodies for high-throughput screening

When conducting disease-related research, it is advisable to incorporate multiple antibodies targeting different epitopes to ensure robust detection of potentially altered protein forms .

How can I optimize immunohistochemistry protocols for GTF3C5 detection in different tissue types?

Optimizing IHC protocols for GTF3C5 detection requires systematic adaptation based on tissue type and fixation method:

  • Antigen retrieval optimization:

    • For FFPE tissue sections (as validated with human breast carcinoma samples), heat-mediated antigen retrieval has shown effectiveness

    • Test multiple pH conditions (citrate buffer pH 6.0 vs. EDTA buffer pH 9.0) to determine optimal epitope exposure

  • Antibody parameters:

    • Starting dilution recommendation: Follow manufacturer protocols, then optimize

    • Incubation conditions: Test both overnight 4°C and 1-2 hour room temperature protocols

    • Detection systems: DAB has been validated for GTF3C5 IHC

  • Tissue-specific considerations:

    • Brain tissue: May require extended fixation and permeabilization steps

    • Lung tissue: Has been validated as a positive control source for GTF3C5 expression

    • Cancer tissues: May show altered expression patterns requiring adjusted detection parameters

  • Controls:

    • Include tissue with known GTF3C5 expression (human breast carcinoma has been validated)

    • Incorporate a negative control by omitting primary antibody

    • Consider using tissues from GTF3C5 knockout models when available

  • Signal amplification:

    • For tissues with low expression, consider tyramide signal amplification or polymer-based detection systems

Following optimization, protocols should be standardized to ensure reproducibility across experiments and sample types.

What are the key differences between polyclonal and monoclonal GTF3C5 antibodies for specific research applications?

The choice between polyclonal and monoclonal GTF3C5 antibodies should be guided by experimental requirements:

FeaturePolyclonal GTF3C5 AntibodiesMonoclonal GTF3C5 Antibodies
Epitope RecognitionMultiple epitopes (e.g., rabbit polyclonals from Abbexa, Proteintech) Single epitope (e.g., mouse monoclonal 3F10)
SensitivityGenerally higher due to multiple epitope recognitionMay have lower sensitivity but higher specificity
Batch-to-Batch VariationHigher variation requiring validation between lotsMore consistent performance between batches
Best ApplicationsIHC, WB for general detectionCritical epitope-specific applications, quantitative assays
Cross-ReactivityOften broader (human, mouse, rat, cow) Usually more species-restricted (primarily human)
Validated UsesWB, IHC, IP, ELISA Primarily WB, ELISA, IP

For reproducibility in long-term projects, monoclonal antibodies offer more consistency, while polyclonal antibodies may provide better detection in applications where the protein conformation might be altered (e.g., denatured samples in WB) .

What strategies should I employ when using GTF3C5 antibodies for co-immunoprecipitation of protein complexes?

GTF3C5 functions as part of the TFIIIC complex, making co-immunoprecipitation (co-IP) a valuable technique for studying its protein interactions. Optimize your co-IP protocol with these methodological considerations:

  • Lysis buffer selection:

    • For maintaining protein-protein interactions, use non-denaturing buffers

    • NETN buffer has been validated for GTF3C5 applications

    • Consider supplementing with phosphatase and protease inhibitors to preserve complex integrity

  • Antibody amount optimization:

    • Start with 3 μg antibody per mg of lysate as validated in previous studies

    • For mouse lung tissue, 0.5-4.0 μg antibody per 1.0-3.0 mg lysate has been effective

  • Pre-clearing strategy:

    • Implement pre-clearing with protein A/G beads to reduce non-specific binding

    • Consider using control IgG from the same species as the GTF3C5 antibody

  • Washing stringency balance:

    • Less stringent washing preserves weaker interactions but increases background

    • More stringent washing reduces background but may disrupt physiologically relevant interactions

    • Test a gradient of salt concentrations to determine optimal conditions

  • Elution and detection:

    • For Western blot analysis, load approximately 20% of immunoprecipitated material

    • Consider native elution for downstream functional assays

When analyzing results, remember that GTF3C5 interacts with other TFIIIC subunits, which can serve as positive controls for successful co-IP experiments.

How can I quantify GTF3C5 expression levels accurately across different experimental conditions?

Accurate quantification of GTF3C5 expression requires careful experimental design and appropriate controls:

  • Western blot quantification approach:

    • Use a concentration gradient of purified recombinant GTF3C5 to create a standard curve

    • Include housekeeping protein controls (β-actin, GAPDH) for normalization

    • Ensure linear detection range by testing multiple exposure times or using digital imaging systems

  • Sample preparation standardization:

    • Consistent protein extraction methods across all samples

    • Accurate protein quantification prior to loading (BCA or Bradford assay)

    • Load a dilution series of a reference sample to verify linearity of detection

  • Data analysis methods:

    • Use digital image analysis software with background subtraction

    • Normalize GTF3C5 signal to loading controls

    • Present data as fold change relative to appropriate control conditions

  • Alternative quantification methods:

    • Consider ELISA-based approaches for more precise quantification

    • qPCR for mRNA levels (as complementary data to protein levels)

  • Validation across methods:

    • Confirm Western blot findings with immunofluorescence quantification

    • For critical findings, verify with mass spectrometry-based protein quantification

By implementing these methodological approaches, researchers can achieve reproducible and accurate quantification of GTF3C5 expression changes across experimental conditions.

How do I address background issues in immunofluorescence staining with GTF3C5 antibodies?

Background issues in immunofluorescence can significantly impact data interpretation. Follow this systematic troubleshooting approach:

  • Fixation and permeabilization optimization:

    • Test different fixatives (4% paraformaldehyde vs. methanol)

    • Adjust permeabilization conditions (0.1-0.5% Triton X-100 for varying times)

    • Consider adding a brief post-fixation quenching step with NH₄Cl to reduce autofluorescence

  • Blocking enhancement:

    • Extend blocking time (1-2 hours or overnight)

    • Test different blocking agents (normal serum from the secondary antibody species)

    • Add 0.1-0.3% Triton X-100 to blocking solution to improve penetration

  • Antibody dilution optimization:

    • Perform a titration series to determine optimal primary antibody concentration

    • For secondary antibodies, use highly cross-adsorbed versions to minimize non-specific binding

  • Controls to implement:

    • Secondary-only control to assess non-specific secondary binding

    • IgG isotype control at the same concentration as primary antibody

    • Peptide competition control to verify specificity

  • Imaging considerations:

    • Collect autofluorescence control images

    • Implement spectral unmixing if available

    • Standardize exposure settings across all samples

By systematically addressing these factors, researchers can significantly improve signal-to-noise ratio in GTF3C5 immunofluorescence applications .

What are the critical quality control steps to validate GTF3C5 antibody specificity before experimental use?

Before employing GTF3C5 antibodies in critical experiments, implement these validation steps:

  • Western blot validation:

    • Run positive control lysates from tissues/cells with known GTF3C5 expression (HeLa, HEK293T, NIH 3T3)

    • Verify band at expected molecular weight (63 kDa)

    • Test in knockout/knockdown systems if available

  • Cross-reactivity assessment:

    • If using across species, validate in each target species

    • For human-specific applications, test in multiple cell types

  • Specificity controls:

    • Peptide competition assay using the immunogen peptide

    • Comparison with alternative antibodies targeting different epitopes

    • Correlation with mRNA expression data

  • Lot-to-lot consistency:

    • For polyclonal antibodies, validate each new lot against previous lots

    • Maintain reference lysates as internal controls for long-term projects

  • Application-specific validation:

    • For IHC: Include positive and negative control tissues

    • For IP: Verify by mass spectrometry when establishing a new protocol

    • For IF: Confirm localization pattern with published literature

Implementing these quality control measures ensures reliable and reproducible results with GTF3C5 antibodies across various applications .

How can GTF3C5 antibodies be utilized in studying the relationship between transcription factor dysregulation and disease phenotypes?

Recent findings linking GTF3C5 variants to clinical manifestations open new research avenues where GTF3C5 antibodies play a crucial role:

  • Patient-derived sample analysis:

    • Compare GTF3C5 expression patterns in samples from patients with hypomelanosis of Ito, seizures, or growth abnormalities to healthy controls

    • Assess protein localization changes that might result from disease-associated mutations

  • Transcriptional dysregulation studies:

    • Use ChIP assays with GTF3C5 antibodies to map binding site alterations in disease states

    • Combine with RNA-seq to correlate binding changes with transcriptional outcomes

  • Protein complex integrity assessment:

    • Employ co-IP with GTF3C5 antibodies to determine if disease-associated variants disrupt TFIIIC complex formation

    • Compare complex composition between normal and pathological conditions

  • Neural tissue investigations:

    • Given the association with seizures, examine GTF3C5 expression in neural tissues using immunohistochemistry

    • Study subcellular localization in neurons using high-resolution imaging techniques

  • Therapeutic monitoring:

    • Develop assays using GTF3C5 antibodies to assess the impact of potential therapeutic interventions

    • Track changes in protein expression or localization during treatment

These approaches enable researchers to mechanistically connect GTF3C5 dysfunction to clinical phenotypes, potentially revealing novel therapeutic targets .

What considerations are important when using GTF3C5 antibodies in chromatin immunoprecipitation (ChIP) experiments?

ChIP experiments with GTF3C5 antibodies require specific optimizations to effectively study its genomic binding patterns:

  • Antibody selection criteria:

    • Choose antibodies validated for IP applications

    • Consider using polyclonal antibodies that recognize multiple epitopes to enhance capture efficiency

    • Verify specificity using Western blot prior to ChIP application

  • Crosslinking optimization:

    • Test different formaldehyde concentrations (0.5-1.5%) and times (5-20 minutes)

    • For studying transient interactions, consider using dual crosslinking with additional agents like disuccinimidyl glutarate (DSG)

  • Chromatin fragmentation:

    • Optimize sonication conditions to achieve 200-500 bp fragments

    • Verify fragmentation by agarose gel electrophoresis before proceeding

  • IP conditions:

    • Use 3-5 μg antibody per ChIP reaction as a starting point based on IP protocols

    • Include appropriate controls: IgG negative control, histone marks as positive controls

    • Consider sequential ChIP (re-ChIP) to study co-occupancy with other transcription factors

  • Data analysis approach:

    • Focus analysis on tRNA genes and other RNA polymerase III targets

    • Compare binding patterns with other TFIIIC components to validate functional relevance

    • Correlate binding with expression data to establish regulatory relationships

ChIP-seq with GTF3C5 antibodies can provide valuable insights into the genomic distribution of TFIIIC complexes and their regulatory roles in normal and disease states.

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.