GTF2H1 (General Transcription Factor IIH, Polypeptide 1, 62kDa) is a crucial component of the TFIIH complex involved in dual essential cellular processes: nucleotide excision repair (NER) of damaged DNA and RNA transcription by RNA polymerase II. In NER, TFIIH acts by opening DNA around lesions to facilitate excision of damaged oligonucleotides and their replacement with new DNA fragments. In transcription, GTF2H1 plays an essential role in initiation, where TFIIH is required for promoter opening and escape after pre-initiation complex (PIC) formation. The phosphorylation of the C-terminal domain of RNA polymerase II by the kinase module CAK controls transcription initiation .
This protein participates in a variety of important protein interactions. For example, retinoblastoma protein (Rb) competes with TATA-binding protein (TBP) and GTF2H1 for binding to E2F, thereby repressing E2F-mediated transactivation. Additionally, herpes simplex virus VP16 and human p53 directly interact with GTF2H1 .
GTF2H1 antibodies are available in multiple formats optimized for different experimental applications:
| Antibody Type | Host Species | Clonality | Common Applications | Target Epitopes | Examples |
|---|---|---|---|---|---|
| Anti-GTF2H1 | Rabbit | Polyclonal | WB, IHC, ELISA, IF | AA 201-300, AA 50-200, AA 50-350, N-Term | ABIN873029, ab204168, ab232982 |
| Anti-GTF2H1 | Mouse | Monoclonal (1F12-1B5) | WB, IHC, IP, RNAi | AA 1-548 | H00002965-M01 |
Most antibodies are unconjugated and purified using Protein A or antigen affinity chromatography. The immunogens used include KLH-conjugated synthetic peptides derived from human GTF2H1 and recombinant fragment proteins .
The choice between polyclonal and monoclonal GTF2H1 antibodies depends on experimental goals:
Polyclonal GTF2H1 antibodies:
Recognize multiple epitopes on the GTF2H1 protein
Provide higher sensitivity due to binding to multiple sites
Useful for detection of denatured proteins in Western blotting
Better for proteins with lower expression levels
Show broader species cross-reactivity (e.g., recognize GTF2H1 in human, mouse, rat, and sometimes other species)
Monoclonal GTF2H1 antibodies:
Recognize a single epitope (e.g., clone 1F12-1B5 targets AA 1-548)
Offer higher specificity and lower background
Provide more consistent results between batches
Particularly useful for co-immunoprecipitation experiments where specificity is critical
For initial characterization or when working with low abundance samples, polyclonal antibodies may be preferable. For highly specific applications or when background is problematic, monoclonal antibodies are often the better choice.
Proper validation of GTF2H1 antibodies is critical for generating reliable research data. A comprehensive validation approach includes:
Western blot validation:
Cross-reactivity testing:
Application-specific validation:
A well-validated antibody should show consistent results across multiple experiments and match known biological characteristics of GTF2H1.
For optimal Western blot results with GTF2H1 antibodies:
Sample preparation:
Use RIPA or NP-40 buffer with protease inhibitors
Include phosphatase inhibitors if phosphorylation status is important
Gel separation and transfer:
8-10% SDS-PAGE gels work well for the 62 kDa GTF2H1 protein
PVDF membranes generally provide better results than nitrocellulose
Blocking and antibody incubation:
Detection considerations:
Enhanced chemiluminescence (ECL) detection works well
For weaker signals, consider using more sensitive ECL substrates
Controls:
The observed molecular weight is typically 62 kDa, matching the calculated molecular weight of GTF2H1 .
For optimal immunohistochemistry (IHC) results with GTF2H1 antibodies:
Tissue preparation:
Formalin-fixed, paraffin-embedded (FFPE) sections work well
Fresh-frozen sections can also be used
Antigen retrieval:
Blocking and antibody incubation:
Detection:
Use appropriate detection systems (HRP/DAB, AP/Fast Red)
Include DAPI or hematoxylin counterstain
Controls:
For mouse tissues, consider using mouse-on-mouse detection systems to minimize background when using mouse monoclonal antibodies.
GTF2H1 antibodies are valuable tools for investigating nucleotide excision repair (NER) pathways, as demonstrated in published research:
Chromatin immunoprecipitation (ChIP) assays:
GTF2H1 antibodies can be used to study recruitment of TFIIH to sites of DNA damage
IP-grade antibodies are essential for these applications
Immunofluorescence to track DNA repair kinetics:
Using GTF2H1 antibodies to visualize recruitment to UV-induced damage sites
Can be combined with other repair factor antibodies to study recruitment timing
Analyzing NER in SWI/SNF-deficient cells:
Research has shown that SWI/SNF chromatin remodelers BRM and BRG1 promote GTF2H1 expression, impacting NER efficiency
GTF2H1 antibodies can be used to quantify protein levels in different genetic backgrounds
In BRM/BRG1-depleted cells, impaired TFIIH function was rescued by ectopic expression of GTF2H1
Research has demonstrated that DNA damage sensitivity of SWI/SNF-deficient cells depends on GTF2H1 levels, suggesting GTF2H1 as a potential predictive marker for platinum drug sensitivity in SWI/SNF-deficient cancer cells .
GTF2H1 antibodies can provide insights into virus-host interactions, as demonstrated in dengue virus research:
Transcriptional response analysis:
Studies show that antibody-dependent dengue virus entry alters host responses that support the viral life cycle
GTF2H1 antibodies can help monitor changes in transcription factor complexes during infection
Tracking TFIIH complex stability:
Using GTF2H1 antibodies to monitor TFIIH complex integrity during viral infection
Combined with antibodies against other TFIIH components to assess complex assembly
Investigating viral manipulation of transcription:
When studying virus-host interactions, coupling GTF2H1 antibody techniques with transcriptomic analyses can provide more comprehensive understanding of how viruses manipulate host transcription machinery.
GTF2H1 and related transcription factors have been implicated in neurological development:
Developmental studies:
Research on related transcription factors like Gtf2i and Gtf2ird1 shows they play important roles in developing brain
GTF2H1 antibodies can help characterize expression patterns in neural tissues
Neurodevelopmental disorder models:
GTF2H1 is part of the TFIIH complex essential for transcription and DNA repair
Mutations affecting TFIIH components have been linked to neurodevelopmental disorders
Antibodies can help characterize expression in disease models
DNA binding properties:
Investigations using GTF2H1 antibodies in neural tissues require careful optimization of fixation and permeabilization protocols to maintain tissue architecture while enabling antibody penetration.
When working with GTF2H1 antibodies in Western blotting, several common issues may arise:
No signal or weak signal:
Increase antibody concentration (try 1:200-1:500 for weak signals)
Increase protein loading (20-40 μg total protein)
Extend primary antibody incubation (overnight at 4°C)
Use more sensitive detection substrates
Verify GTF2H1 expression in your specific sample type
Multiple bands or non-specific binding:
Use more stringent washing (increase TBST washes to 4-5 times, 5-10 minutes each)
Optimize blocking conditions (try 5% BSA instead of milk)
Reduce primary antibody concentration
Verify antibody specificity with appropriate controls
Unexpected molecular weight:
High background:
Use fresher blocking reagents
Increase washing stringency
Reduce antibody concentration
Use higher quality secondary antibodies
For optimal results, store antibodies according to manufacturer recommendations (typically -20°C) and avoid repeated freeze-thaw cycles by preparing small aliquots.
When working with challenging tissue samples for GTF2H1 immunohistochemistry:
Fixation-resistant tissues:
Extended antigen retrieval (20-30 minutes)
Try alternative retrieval methods (pressure cooker, enzyme-based retrieval)
Consider dual retrieval approaches (heat followed by enzymatic)
High background in specific tissues:
For liver, kidney, or brain tissues that often show high background:
Use longer blocking steps (2 hours)
Include additional blocking agents (avidin/biotin block if using biotinylated secondaries)
Consider tyramide signal amplification for specific signal enhancement
Weak staining:
Titrate antibody concentration (try 1:50-1:200 range)
Extend primary antibody incubation to 48 hours at 4°C
Use polymer-based detection systems for signal enhancement
Consider paraffin section thickness (4-5 μm optimal)
Tissue-specific considerations:
For challenging samples, performing a titration series with both antigen retrieval conditions and antibody concentrations can help identify optimal conditions.
When faced with contradictory results from different GTF2H1 antibodies:
Epitope differences:
Antibodies targeting different regions of GTF2H1 may give different results
N-terminal antibodies vs. central domain antibodies may detect different forms
Check if the epitopes are accessible in your experimental conditions
Isoform specificity:
Confirm which isoforms your antibodies detect
Different antibodies may recognize different splice variants
Post-translational modifications:
Complex formation:
GTF2H1 exists in the TFIIH complex
Some epitopes may be masked when in complex
Native vs. denaturing conditions can give different results
Cross-reactivity:
When publishing contradictory results, include detailed methods sections specifying exact antibody catalog numbers, dilutions, and incubation conditions to enable proper replication.
Based on research showing GTF2H1 regulation by SWI/SNF chromatin remodelers BRM and BRG1, optimal experimental design should include:
Gene expression analysis:
RT-qPCR to measure GTF2H1 mRNA levels following BRM/BRG1 knockdown
ChIP-qPCR using BRM/BRG1 antibodies to verify direct binding to GTF2H1 promoter
Analysis of other TFIIH subunits to assess specificity of regulation
Protein level and stability assessment:
Western blotting with GTF2H1 antibodies following BRM/BRG1 depletion
Pulse-chase experiments to determine if protein stability is affected
Co-immunoprecipitation to assess TFIIH complex integrity
Functional rescue experiments:
Complementation with ectopic GTF2H1 to rescue phenotypes
Testing nucleotide excision repair capacity
Analyzing transcriptional activity using reporter assays
Cell type considerations:
Research has shown that DNA damage sensitivity in SWI/SNF-deficient cells correlates with GTF2H1 levels, suggesting GTF2H1 could potentially be used as a marker for platinum drug sensitivity in SWI/SNF-deficient cancers .
When investigating GTF2H1's role in DNA repair, include these essential controls:
Positive controls:
Known NER-deficient cells (XPA-/-, XPC-/-)
UV-sensitive cell lines
Cells treated with known TFIIH inhibitors
Negative controls:
Complemented cells (GTF2H1-deficient cells with restored expression)
Isogenic wild-type cells
Cells with mutations in non-NER pathways
Technical controls:
Antibody specificity controls (blocking peptides, isotype controls)
siRNA off-target controls (multiple siRNAs, rescue experiments)
DNA damage verification (CPD antibodies for UV damage)
Functional readouts:
Direct repair assays (Unscheduled DNA synthesis, Host cell reactivation)
Survival assays with DNA damaging agents
TFIIH complex integrity assessment
Research has demonstrated that XPD recruitment to local UV damage was impaired in BRM and BRG1 depleted cells, but could be rescued by ectopic expression of GTF2H1, confirming specificity of the observed repair defects .
Distinguishing GTF2H1's dual functions requires carefully designed experiments:
Chromatin association studies:
ChIP-seq to identify genome-wide binding sites during transcription vs. repair
Compare GTF2H1 binding before and after DNA damage induction
Co-occupancy analysis with transcription vs. repair factors
Mutational analysis:
Distinguish domain-specific functions using domain-specific antibodies
Correlate antibody binding with specific functional outcomes
Temporal dynamics:
Time-course experiments following DNA damage
Synchronized cell populations to separate cell-cycle effects
Co-localization studies:
Combine with markers specific to transcription (RNA Pol II phospho-CTD)
Compare with repair-specific markers (γH2AX, XPC)
Triple staining to identify transition points between functions
Functional separation:
Use α-amanitin to inhibit transcription while studying repair function
Use specifically timed UV irradiation to study repair without affecting global transcription
Research has shown that GTF2H1 depletion impacts both transcription levels and DNA repair capacity, with ectopic expression of GTF2H1 rescuing XPD recruitment to local UV damage sites .
When analyzing GTF2H1 expression changes in disease contexts:
Cancer studies:
Neurodegenerative diseases:
Changes may reflect altered transcriptional regulation
Consider impact on both RNA Pol II transcription and DNA repair capacity
Correlate with markers of DNA damage accumulation
Viral infections:
Developmental disorders:
Quantification approaches:
Use both mRNA (RT-qPCR) and protein (Western blot) quantification
Consider complex formation and stability
Normalize to appropriate housekeeping genes/proteins
When publishing GTF2H1 expression data from disease models, include detailed information about antibody validation, quantification methods, and statistical analysis to ensure reproducibility.
Several innovative technologies are advancing GTF2H1 antibody applications:
Proximity labeling approaches:
BioID or APEX2 fusions with GTF2H1 to identify context-specific interaction partners
Allows identification of transient interactions during transcription vs. repair
Live-cell imaging techniques:
Antibody fragments (nanobodies) for live-cell visualization
CRISPR-based tagging for endogenous protein tracking
Super-resolution microscopy to visualize TFIIH complex assembly
Single-cell applications:
Mass cytometry (CyTOF) with GTF2H1 antibodies for single-cell protein analysis
Integration with single-cell transcriptomics
Spatial transcriptomics combined with GTF2H1 immunostaining
Automated high-content screening:
High-throughput imaging of GTF2H1 recruitment to damage sites
Screening for compounds that modulate TFIIH function
Machine learning algorithms to classify phenotypes
CRISPR-based genetic screens:
Combining CRISPR screens with GTF2H1 antibody-based readouts
Identifying novel regulators of TFIIH complex assembly and function
These emerging technologies offer opportunities to study GTF2H1 with unprecedented spatial and temporal resolution in both normal and disease states.
Current limitations in GTF2H1 antibody research include:
Specificity challenges:
Cross-reactivity with related proteins in some species
Solution: Develop and validate highly specific monoclonal antibodies
CRISPR knockout validation to confirm specificity
Post-translational modification detection:
Limited availability of modification-specific antibodies
Solution: Develop antibodies against known GTF2H1 modifications
Combine with mass spectrometry for comprehensive PTM mapping
Complex assembly detection:
Difficulty distinguishing free vs. TFIIH-complexed GTF2H1
Solution: Develop conformation-specific antibodies
Proximity ligation assays to visualize complex formation
Quantification standardization:
Variability in antibody performance between lots
Solution: Develop recombinant protein standards
Establish quantitative benchmarks across laboratories
Tissue penetration in thick sections:
Limited antibody penetration in brain and other dense tissues
Solution: Develop smaller antibody fragments
Tissue clearing techniques compatible with immunostaining
Future directions should include development of highly specific recombinant antibodies with standardized validation protocols to enhance reproducibility across laboratories.