The TTC32 antibody targets the Tetratricopeptide Repeat Domain 32 (TTC32), a protein encoded by the TTC32 gene (Gene ID: 130502; UniProt ID: Q5I0X7) . This antibody is widely used in research to study TTC32's role in cellular processes, leveraging its reactivity across human and mouse tissues .
FITC-Conjugated TTC32 Antibody (ABIN6069969): Optimized for ELISA applications with human samples, requiring titration for optimal results .
IHC: Robust nuclear/cytoplasmic staining in paraffin-embedded mouse testis tissues .
IP: Efficient pull-down of TTC32 from mouse testis lysates .
Human Protein Atlas (HPA) Validation: Includes IHC on 44 normal and 20 cancer tissues, alongside protein array testing against 364 recombinant proteins .
No cross-reactivity with unrelated proteins confirmed via stringent antigen-affinity purification .
Subcellular Localization: TTC32 shows nuclear and cytoplasmic expression in testis tissues, suggesting roles in spermatogenesis .
Molecular Interactions: Tetratricopeptide repeats mediate protein-protein interactions, potentially influencing chaperone complexes .
Disease Relevance: While not directly linked to cancer, TTC32 antibodies are critical tools for studying genetic disorders involving protein misfolding .
TTC32 (Tetratricopeptide repeat domain 32) is a human protein-coding gene with a molecular weight of approximately 17.1 kDa. The tetratricopeptide repeat motif is a structural motif that mediates protein-protein interactions and is found in a wide range of proteins involved in various cellular processes. Research interest in TTC32 stems from its potential role in protein-protein interactions within cellular pathways. When designing experiments targeting TTC32, researchers should consider its cellular localization and expression patterns to select appropriate controls and experimental conditions .
TTC32 antibodies are available in both polyclonal and monoclonal formats, each with distinct advantages. Polyclonal antibodies, such as the rabbit-derived antibody from Sigma-Aldrich, recognize multiple epitopes on TTC32, potentially offering higher sensitivity but variable specificity between lots . Monoclonal antibodies, including mouse-derived clones OTI3E11, OTI5B4, and OTI3C2, target specific epitopes, providing consistent results between experiments but potentially lower sensitivity than polyclonals . When selecting between these formats, researchers should consider the specific requirements of their experimental approach, with monoclonals generally preferred for reproducible, long-term studies and polyclonals often advantageous for initial detection or enhancement of weak signals .
TTC32 antibodies support multiple research applications with varying dilution requirements. For Western blotting (WB), monoclonal antibodies typically require dilutions ranging from 1:1000 to 1:2000 . Immunohistochemistry (IHC) applications generally use dilutions of 1:50 to 1:200 for polyclonal antibodies and approximately 1:150 for monoclonals . Immunofluorescence (IF) typically requires 0.25-2 μg/mL for polyclonal antibodies or a 1:100 dilution for monoclonals . Flow cytometry applications generally employ a 1:100 dilution . Researchers should optimize these recommended dilutions for their specific samples and detection systems through titration experiments to achieve optimal signal-to-noise ratios .
Optimization of TTC32 antibody for Western blotting requires a systematic approach. Begin with sample preparation by testing different lysis buffers (RIPA, NP-40, or Triton X-100-based) to ensure efficient protein extraction. For electrophoretic separation, use 12-15% polyacrylamide gels to achieve optimal resolution of the 17.1 kDa TTC32 protein . Perform initial antibody titration experiments using a dilution series (e.g., 1:500, 1:1000, 1:2000, 1:5000) to determine the optimal concentration that maximizes specific signal while minimizing background . Test various blocking agents (5% BSA, 5% non-fat dry milk) and incubation conditions (1 hour at room temperature vs. overnight at 4°C). Include appropriate positive controls (HEK293T cells transfected with TTC32 expression vectors) and negative controls (non-transfected cells) to validate specificity . Document all optimization steps, maintaining consistent sample loads (20-30 μg total protein) across experiments to ensure reproducibility of results.
Successful immunohistochemistry with TTC32 antibodies depends on several critical steps. Tissue fixation significantly impacts epitope preservation—compare 10% neutral buffered formalin with other fixatives if initial results are suboptimal. Antigen retrieval is essential; test both heat-induced epitope retrieval (citrate buffer pH 6.0 or EDTA buffer pH 9.0) and enzymatic retrieval methods to determine optimal conditions for TTC32 epitope accessibility . When using polyclonal antibodies, implement a 1:50 to 1:200 dilution range, while monoclonal antibodies typically perform best at approximately 1:150 . Optimize incubation parameters (temperature, duration) and detection systems (ABC, polymer-based) based on signal intensity and background levels. Include appropriate positive tissue controls with known TTC32 expression and negative controls (primary antibody omission, isotype controls) to confirm staining specificity . Counterstain optimization is also important—adjust hematoxylin timing to achieve nuclear definition without obscuring specific TTC32 signals.
When encountering weak or absent signals in TTC32 immunofluorescence, implement a systematic troubleshooting approach. First, verify antibody activity using a positive control sample (such as HEK293T cells overexpressing TTC32) . If the control fails, assess antibody quality through a dot blot or Western blot using recombinant TTC32 protein. For fixation-related issues, compare paraformaldehyde (PFA) fixation with methanol or acetone fixation, as different fixatives preserve different epitopes . Enhance epitope accessibility by testing various permeabilization methods (0.1-0.5% Triton X-100, 0.1-0.2% Saponin) and durations. If background is high but specific signal is weak, optimize blocking conditions (test 2-5% BSA, normal serum, or commercial blocking reagents) and increase antibody concentration (use up to 2 μg/mL for polyclonal or 1:50 dilution for monoclonal antibodies) . Extend primary antibody incubation to overnight at 4°C and experiment with signal amplification systems (tyramide signal amplification, more sensitive fluorophores). Document all modifications to establish an optimized protocol for future experiments.
Validating TTC32 antibody specificity requires a multi-faceted approach. Begin with knockdown/knockout controls by performing siRNA/shRNA-mediated knockdown or CRISPR-Cas9 knockout of TTC32 in your experimental system, expecting significant signal reduction in these samples . Implement overexpression controls using cells transfected with TTC32 expression vectors compared to empty vector controls . For epitope competition assays, pre-incubate the antibody with excess immunizing peptide (such as the sequence YEAMDDYTSAIEVQPNFEVPYYNRGLILYRLGYFDDALEDFKKVLDLNPGFQDATLSLKQTILDKEE for the Sigma antibody) before application to samples, which should substantially reduce specific signals . Cross-validate findings using multiple TTC32 antibodies targeting different epitopes (e.g., comparing results from OTI3E11, OTI5B4, and OTI3C2 clones) . Verify molecular weight consistency by confirming that the detected band in Western blots appears at the expected 17.1 kDa size . Document all validation experiments comprehensively, as these will strengthen the reliability of your research findings.
Investigating TTC32 protein-protein interactions requires complementary methodological approaches. For co-immunoprecipitation experiments, use TTC32 antibodies coupled to protein A/G beads to pull down TTC32 and associated proteins from cell lysates, followed by mass spectrometry or Western blotting for suspected interacting partners . Proximity ligation assays (PLA) can visualize interactions in situ by using TTC32 antibody (1:100 dilution) paired with antibodies against suspected interaction partners . For bimolecular fluorescence complementation (BiFC), create fusion constructs of TTC32 and putative interaction partners with split fluorescent protein fragments. Yeast two-hybrid screening using TTC32 as bait can identify novel binding partners. GST pull-down assays with recombinant GST-tagged TTC32 incubated with cell lysates allow for in vitro verification of interactions. When interpreting interaction data, consider the tetratricopeptide repeat motif's structural properties and established interaction patterns to contextualize findings. Control experiments should include non-specific IgG for co-IP and cells expressing only one fusion partner for BiFC.
Investigating post-translational modifications (PTMs) of TTC32 requires specialized experimental design. For phosphorylation analysis, enrich phospho-proteins using titanium dioxide or immobilized metal affinity chromatography before TTC32 immunoprecipitation with available antibodies . Mass spectrometry analysis can identify specific modified residues and modification types. When studying ubiquitination, perform immunoprecipitation under denaturing conditions to disrupt non-covalent interactions, followed by Western blotting with both TTC32 and ubiquitin antibodies. For site-directed mutagenesis studies, create point mutations at predicted modification sites (based on computational analysis) and compare the function and localization of wild-type and mutant TTC32. To assess the dynamics of modifications, treat cells with modulators of specific PTM pathways (phosphatase inhibitors, proteasome inhibitors) before TTC32 analysis. Control experiments should include samples treated with specific PTM-removing enzymes. Document the buffer compositions and experimental conditions comprehensively, as PTM detection often requires specialized approaches beyond standard protocols.
Maintaining TTC32 antibody activity requires proper storage and handling. For long-term storage, keep antibodies at -20°C, as recommended by manufacturers across multiple product lines . Reconstitution protocols vary by formulation—for lyophilized antibodies, add the recommended volume of sterile distilled water or PBS to achieve a final concentration of approximately 1 mg/mL . For working aliquots, store at 2-8°C for up to two weeks to minimize freeze-thaw cycles . When handling glycerol-containing formulations, avoid centrifugation which can create concentration gradients . Implement antibody validation at regular intervals (every 6-12 months) using positive controls to monitor potential activity loss over time. For carrier-free antibodies intended for conjugation, perform additional desalting steps as recommended by manufacturers . Maintain detailed records of receipt date, lot number, aliquoting dates, and thaw cycles for each antibody to track potential sources of variability in experimental results.
Buffer compositions significantly impact TTC32 antibody performance across applications. For Western blotting, TBST (Tris-buffered saline with 0.1% Tween-20) is standard, but reducing Tween-20 concentration to 0.05% may decrease background when using monoclonal antibodies . In immunohistochemistry, adding 0.1-0.3% Triton X-100 to PBS-based buffers enhances tissue penetration, particularly important for the rabbit polyclonal antibody . For immunofluorescence, PBS with 1-2% BSA provides effective blocking while preserving epitope accessibility for all TTC32 antibody types . In flow cytometry applications, PBS with 0.5-1% BSA and 0.1% sodium azide helps maintain antibody stability during longer incubations . When using glycerol-containing antibodies (like the Sigma product), ensure thorough mixing before dilution to prevent concentration inconsistencies . For carrier-free applications or conjugation chemistry, phosphate buffers without preservatives are preferred, as demonstrated in the reconstitution recommendations for lyophilized formats . Document optimized buffer compositions for each application to ensure experimental reproducibility.
Implementing rigorous quality control for TTC32 antibodies requires comprehensive metrics. Establish lot-to-lot validation protocols by testing each new antibody lot against a reference lot using Western blotting or immunofluorescence with standardized positive controls . Develop specificity assessment procedures including blocking peptide competition assays using the immunogen sequence (YEAMDDYTSAIEVQPNFEVPYYNRGLILYRLGYFDDALEDFKKVLDLNPGFQDATLSLKQTILDKEE) . Create sensitivity benchmarks by determining detection limits using serial dilutions of recombinant TTC32 protein or lysates from cells expressing varying TTC32 levels. Implement reproducibility monitoring through technical replicates and periodic retesting of archived samples. For long-term studies, establish antibody performance tracking using control lysates or tissues with documented staining patterns and signal intensities. Create application-specific validation panels (different fixation methods for IHC, various blocking agents for IF) . Document all quality control results in a dedicated database, including images of expected results, to facilitate troubleshooting and ensure consistent experimental outcomes throughout your research program.