The TOK1 protein, encoded by the BCCIP gene, is a nuclear and cytoplasmic protein involved in:
DNA Repair: Facilitating homologous recombination (HR) and stabilizing replication forks during DNA replication stress .
Cell Cycle Regulation: Interacting with p21 (CDKN1A) to modulate cyclin-dependent kinase (CDK) activity, particularly CDK2 .
Cancer Pathways: Implicated in p53 transactivation and tumor suppression, with deficiencies linked to neural developmental defects and cancer progression .
The antibody specifically binds to the TOK1 protein, enabling its detection in biological samples.
Multiple commercial sources offer TOK1 antibodies, varying in type and specificity:
Supplier | Antibody Type | Reactivity | Applications |
---|---|---|---|
Biocompare | Polyclonal/Mono | Human | ELISA, Western Blot, IF |
Cepham Life Sciences | Polyclonal | Human | WB (1:500–1:2000), ELISA |
Proteintech | Monoclonal | Human | WB, IP, IF |
Polyclonal Antibodies: Broad epitope recognition, ideal for immunoprecipitation and Western blotting .
Monoclonal Antibodies: High specificity, often validated in knockout cell lines for precision .
The TOK1 antibody is utilized in:
Western Blot: Use SDS-PAGE under reducing conditions (e.g., DTT/BME) .
Immunofluorescence: Fix cells with 4% paraformaldehyde and permeabilize with 0.1% Triton X-100 .
Homologous Recombination: TOK1 stabilizes replication forks during replication stress, preventing genome instability .
p53 Pathway: Binds p21 and enhances its inhibition of CDK2, linking TOK1 to tumor suppression .
Oncogenic Roles: Overexpression in head and neck squamous cell carcinoma (HNSCC) correlates with poor prognosis due to E6-mediated p53 degradation .
Therapeutic Target: Inhibiting TOK1 may sensitize cancer cells to replication stress-inducing agents .
KEGG: sce:YJL093C
STRING: 4932.YJL093C
TOK-1 is a reported synonym of the BCCIP gene, which encodes the BRCA2 and CDKN1A interacting protein. This protein plays crucial roles in multiple cellular processes including DNA repair and cytoskeleton organization. The human version has a canonical amino acid length of 314 residues and a protein mass of 36 kilodaltons, with 4 distinct isoforms identified to date . Its significance lies in its dual localization in both nucleus and cytoplasm, suggesting multifunctional roles in cellular homeostasis. As a member of the BCP1 protein family, TOK-1 represents an important target for understanding fundamental cellular processes, particularly those related to genomic stability.
TOK-1 antibodies are primarily employed in several standard molecular and cellular biology techniques:
Application | Common Dilution Range | Sample Types | Detection Method |
---|---|---|---|
Western Blot | 1:500-1:2000 | Cell/tissue lysates | Chemiluminescence |
Immunofluorescence | 1:100-1:500 | Fixed cells/tissues | Fluorescence microscopy |
ELISA | 1:1000-1:5000 | Purified protein/serum | Colorimetric/fluorometric |
These applications enable researchers to detect and measure TOK-1 antigen in various biological samples, making them essential tools for studies investigating protein-protein interactions, subcellular localization, and expression levels in different experimental conditions .
Polyclonal and monoclonal TOK-1 antibodies offer distinct advantages depending on the research context:
Polyclonal TOK-1 antibodies: Recognize multiple epitopes on the TOK-1 antigen, providing higher sensitivity but potentially lower specificity. These are typically generated in rabbits and are useful for initial detection studies and applications where signal amplification is important .
Monoclonal TOK-1 antibodies: Recognize a single epitope with high specificity, ensuring consistent results across experiments. These are particularly valuable for distinguishing between closely related proteins or specific isoforms of TOK-1.
The choice between these antibody types should be guided by experimental requirements. For confirming novel interactions or conducting initial screens, polyclonal antibodies may be preferred due to their higher sensitivity. For precise mapping studies or when background is problematic, monoclonal antibodies offer superior specificity.
The dual localization of TOK-1 in both nuclear and cytoplasmic compartments necessitates specific sample preparation approaches:
Nuclear extraction protocol:
Harvest cells and wash with ice-cold PBS
Resuspend in hypotonic buffer (10 mM HEPES pH 7.9, 10 mM KCl, 1.5 mM MgCl₂)
Add NP-40 to 0.5% final concentration
Centrifuge to separate cytoplasmic (supernatant) and nuclear (pellet) fractions
Extract nuclear proteins with high-salt buffer (20 mM HEPES pH 7.9, 420 mM NaCl, 1.5 mM MgCl₂, 0.2 mM EDTA)
Cytoskeletal fraction protocol:
Extract soluble proteins with Triton X-100 buffer
Collect insoluble fraction enriched in cytoskeletal components
Solubilize with sample buffer containing 6M urea
Including appropriate subcellular markers (e.g., Lamin A/C for nuclear fraction, GAPDH for cytoplasmic fraction) in Western blot analysis is essential to confirm fractionation efficiency and avoid misinterpretation of localization data .
Optimizing immunofluorescence for TOK-1 detection requires attention to several critical parameters:
Including positive controls (cells known to express TOK-1) and negative controls (primary antibody omission and isotype controls) is crucial for confirming specificity .
Proper validation of TOK-1 antibody specificity requires multiple complementary approaches:
Control Type | Implementation | Purpose |
---|---|---|
Knockout/knockdown | CRISPR-Cas9 or siRNA against TOK-1 | Confirms signal is specific to target |
Peptide competition | Pre-incubate antibody with immunizing peptide | Verifies epitope specificity |
Overexpression | Transfect TOK-1 expression construct | Demonstrates signal increases with target abundance |
Cross-reactivity | Test across multiple species and related proteins | Confirms specificity versus related proteins |
Multiple antibodies | Use antibodies targeting different TOK-1 epitopes | Corroborates detection pattern |
The conformation-dependent nature of some antibody epitopes (as seen with TOC1) suggests that researchers should consider how protein interactions or modifications might affect epitope accessibility . This becomes particularly important when investigating TOK-1's role in protein complexes involved in DNA repair mechanisms.
Distinguishing between the four identified TOK-1 isoforms requires sophisticated antibody selection and experimental design:
Epitope-specific antibodies: Select antibodies targeting regions unique to specific isoforms. Custom antibodies may be required for isoform-specific epitopes not covered by commercial offerings.
2D gel electrophoresis: Combine isoelectric focusing with SDS-PAGE followed by Western blotting to separate isoforms based on both molecular weight and charge differences.
Immunoprecipitation coupled with mass spectrometry: Pull down TOK-1 using a pan-isoform antibody, then identify specific isoforms by mass spectrometry analysis of unique peptides.
RNA interference: Design siRNAs targeting isoform-specific exons, then use Western blotting to confirm which bands correspond to which isoforms.
Recombinant protein standards: Express each isoform recombinantly and use as size markers and positive controls.
This multi-faceted approach ensures reliable discrimination between TOK-1 isoforms, which is essential for understanding their potentially distinct functional roles .
TOK-1's involvement in protein complexes, particularly those related to DNA repair mechanisms, can lead to epitope masking issues similar to those observed with other conformation-dependent antibodies like TOC1 . Several approaches can address this challenge:
Sample denaturation optimization: Test various denaturing conditions to expose masked epitopes without compromising antibody recognition. For Western blotting, adjust SDS concentration, heating time, and reducing agent concentration.
Epitope retrieval methods: For fixed tissues or cells, evaluate different antigen retrieval methods:
Heat-induced epitope retrieval (citrate buffer pH 6.0 or Tris-EDTA pH 9.0)
Enzymatic retrieval (proteinase K, trypsin)
Detergent-based methods (Triton X-100, SDS)
Proximity ligation assay (PLA): This technique can detect TOK-1 within protein complexes even when conventional antibody approaches fail due to epitope masking.
Crosslinking followed by immunoprecipitation: Use reversible crosslinkers to stabilize protein complexes before immunoprecipitation, then reverse crosslinking for analysis.
Native gel electrophoresis: Preserve protein complexes during separation, then detect TOK-1 using overlay techniques with labeled antibodies.
These approaches recognize that, similar to TOC1's behavior with tau oligomers, TOK-1 epitopes may be concealed in certain protein configurations but revealed in others .
Integrating antibody-based detection with functional genomics provides a comprehensive understanding of TOK-1's roles:
ChIP-seq analysis: Use TOK-1 antibodies for chromatin immunoprecipitation followed by sequencing to identify DNA binding sites if TOK-1 functions in transcriptional regulation.
Proximity-dependent biotin identification (BioID): Fuse TOK-1 with a biotin ligase to identify proximal proteins in living cells, then confirm interactions using co-immunoprecipitation with TOK-1 antibodies.
CRISPR screens with antibody readouts: Combine genome-wide CRISPR screening with antibody-based detection of TOK-1 to identify genes affecting TOK-1 stability, localization, or post-translational modifications.
Single-cell analysis: Couple single-cell RNA-seq with antibody-based protein detection (CITE-seq) to correlate TOK-1 protein levels with transcriptomic changes at the single-cell level.
Multiplex imaging: Use TOK-1 antibodies in cyclic immunofluorescence or mass cytometry to understand spatial relationships with other proteins across different cellular states.
This integrated approach leverages the specificity of antibody-based detection while harnessing the power of genomic technologies, similar to strategies used in developing next-generation antibody discovery platforms .
Inconsistent antibody performance can significantly impact experimental reproducibility. For TOK-1 antibodies, consider these systematic troubleshooting approaches:
Antibody validation across conditions: Test each new antibody lot using positive controls under standardized conditions. Document optimal working dilutions for each application.
Sample preparation optimization:
For proteins involved in DNA repair like TOK-1, cell cycle synchronization may be necessary to obtain consistent results
Consider how different lysis buffers affect protein complex stability
Test fresh vs. frozen samples to determine impact on epitope integrity
Epitope accessibility assessment: Similar to observations with conformation-dependent antibodies like TOC1 , TOK-1 epitopes may be variably accessible depending on protein conformation:
Test different fixation protocols for immunofluorescence
For Western blotting, compare reducing vs. non-reducing conditions
Consider native vs. denaturing conditions based on experimental goals
Secondary antibody optimization: Test multiple secondary antibodies to identify optimal combinations that minimize background while maximizing specific signal.
Signal enhancement strategies: For low abundance targets, consider amplification systems like tyramide signal amplification or polymer-based detection systems.
Maintaining detailed laboratory records of conditions that yield consistent results is essential for reliable TOK-1 detection across experiments.
When different detection methods yield conflicting results regarding TOK-1 expression or localization, systematic reconciliation approaches are necessary:
Method-specific validation: Evaluate each method independently with appropriate controls:
For Western blotting: Include recombinant TOK-1 standards and knockdown controls
For immunofluorescence: Compare multiple fixation methods and include co-localization with known interacting partners
For ELISA: Establish standard curves with recombinant protein and validate antibody pairs for specificity
Orthogonal validation: Confirm antibody-based findings using antibody-independent methods:
Validate protein expression with RNA-level measurements (qPCR, RNA-seq)
Use tagged TOK-1 constructs to confirm localization patterns
Apply mass spectrometry to verify protein identity and modifications
Systematic comparison of experimental variables: When methods disagree, systematically evaluate:
Cell/tissue preparation differences (lysis methods, fixation protocols)
Epitope accessibility under different conditions
Potential interference from post-translational modifications
Cross-reactivity with related proteins
Multi-antibody consensus approach: Use multiple antibodies targeting different TOK-1 epitopes and consider results reliable only when supported by multiple antibodies.
This systematic approach helps distinguish true biological variation from technical artifacts, particularly important when studying multifunctional proteins like TOK-1 that may adopt different conformations in various cellular contexts .
Developing quantitative assays for TOK-1 requires careful optimization and validation:
Quantitative Western blotting protocol:
Use infrared fluorescence-based detection systems (e.g., LI-COR) for broader linear dynamic range
Include calibration curves with recombinant TOK-1 standards at known concentrations
Normalize to validated housekeeping proteins appropriate for experimental conditions
Apply statistical methods to determine limits of detection and quantification
ELISA/AlphaLISA development:
Test multiple antibody pairs to identify optimal capture and detection antibodies
Establish standard curves with recombinant TOK-1
Validate assay parameters: sensitivity, specificity, precision, accuracy
Optimize sample dilution protocols to ensure measurements fall within linear range
High-content imaging quantification:
Develop automated image analysis workflows for quantifying TOK-1 signal intensity
Include calibration standards in each experiment
Account for cell-to-cell variability through single-cell analysis approaches
Validate findings using orthogonal methods
Flow cytometry-based quantification:
Use antibody binding capacity (ABC) beads to establish standard curves
Apply compensation controls to account for spectral overlap
Include isotype controls to establish background thresholds
Validate with cells expressing known quantities of TOK-1
Each of these approaches should be validated following similar principles to those used in antibody discovery platforms, ensuring reliability and reproducibility .
Incorporating TOK-1 antibodies into advanced imaging technologies opens new possibilities for understanding its functional dynamics:
Super-resolution microscopy applications:
STORM/PALM imaging: Conjugate TOK-1 antibodies with photo-switchable fluorophores to achieve nanometer resolution
SIM imaging: Use structured illumination to improve resolution 2-fold beyond diffraction limit
Expansion microscopy: Physically expand specimens to visualize TOK-1 distribution within protein complexes
Live-cell imaging strategies:
Develop cell-permeable TOK-1 nanobodies conjugated to fluorescent proteins
Use split-GFP complementation systems where one fragment is fused to anti-TOK-1 nanobodies
Apply FRET-based biosensors to monitor TOK-1 interactions with binding partners
Correlative light and electron microscopy (CLEM):
Use TOK-1 antibodies conjugated to both fluorescent tags and electron-dense particles
Correlate fluorescence microscopy with ultrastructural details from electron microscopy
Apply cryogenic techniques to preserve native protein states
Lattice light-sheet microscopy:
Achieve gentle, high-speed 3D imaging of TOK-1 dynamics
Combine with adaptive optics for deep tissue imaging
Integrate with optogenetic tools to manipulate TOK-1 function while imaging
These approaches would significantly advance our understanding of TOK-1's spatial and temporal dynamics during processes like DNA repair, similar to how conformation-specific antibodies have enhanced our understanding of tau pathology .
While the query specifies avoiding commercial questions, the research aspects of therapeutic antibody development are scientifically relevant:
Target validation considerations:
Determine whether TOK-1 modulation affects tumor-specific vulnerabilities
Evaluate whether TOK-1 inhibition creates synthetic lethality in cancer cells with specific mutations
Assess potential on-target toxicity in normal tissues with high DNA repair requirements
Antibody engineering approaches:
Develop antibodies that distinguish between normal and pathological TOK-1 conformations
Engineer cell-penetrating antibodies to reach nuclear TOK-1
Consider domain-specific inhibitory antibodies that block specific functions while preserving others
Combination therapy research:
Study how TOK-1-targeted approaches might sensitize cells to DNA-damaging agents
Investigate potential synergies with other DNA repair inhibitors
Explore combinations with immune checkpoint inhibitors if TOK-1 modulation affects antigen presentation
Delivery system development:
This research focus aligns with broader trends in antibody-drug conjugate development, which show expansion in both publications and clinical trials over the past decade .
Integrating TOK-1 antibody-based studies with multi-omics approaches provides comprehensive insights into its biological functions:
Integrated proteomics workflows:
Antibody-based purification of TOK-1 complexes followed by mass spectrometry
Phospho-proteomics to map how TOK-1 affects signaling networks
Proximity labeling (BioID/APEX) followed by mass spectrometry to identify spatial interactors
Quantitative proteomics comparing wild-type and TOK-1-deficient cells
Transcriptomics integration:
RNA-seq following TOK-1 modulation to identify regulated genes
ChIP-seq using TOK-1 antibodies to map potential DNA binding sites
Single-cell RNA-seq combined with antibody-based protein quantification (CITE-seq)
Nascent RNA sequencing to distinguish direct from indirect effects
Epigenomic connections:
ATAC-seq to determine chromatin accessibility changes associated with TOK-1 function
CUT&RUN using TOK-1 antibodies for high-resolution chromatin binding profiles
DNA methylation profiling to correlate with TOK-1 activity in DNA repair processes
Metabolomic correlations:
Isotope tracing combined with TOK-1 immunoprecipitation to identify associated metabolic enzymes
Metabolomic profiling of TOK-1-deficient cells to identify altered pathways
Analysis of metabolic dependencies in cells with different TOK-1 expression levels
Such integrated approaches would provide systems-level insights into TOK-1 function, similar to comprehensive strategies used in developing novel antibody discovery platforms .