The CLTC Antibody, HRP-conjugated is a specialized immunological reagent designed to detect the clathrin heavy chain (CLTC), a key component of the clathrin protein complex involved in receptor-mediated endocytosis. Horseradish peroxidase (HRP) conjugation enables direct enzymatic detection in applications like Western blotting (WB), eliminating the need for secondary antibodies. This conjugated antibody is widely used in cellular biology and virology to study endocytotic pathways and viral entry mechanisms .
The HRP-conjugated CLTC antibody is primarily used for WB to detect CLTC in cell lysates or tissue samples. It enables one-step detection, simplifying protocols compared to unconjugated antibodies requiring secondary reagents .
CLTC antibodies are used to isolate clathrin-coated vesicles or study protein interactions. HRP-conjugated variants may aid in downstream detection of precipitated complexes .
CLTC knockout studies (e.g., for SFTSV) highlight its role in viral endocytosis. HRP-conjugated antibodies facilitate rapid validation of CLTC depletion in knockout models .
CLTC antibodies are critical for studying clathrin-dependent endocytosis. HRP-conjugated variants enable efficient detection of CLTC in pathways involving receptor internalization .
Knockout studies using CRISPR-Cas9 (validated via CLTC antibodies) revealed that CLTC depletion reduces SFTSV infection but does not block it entirely, indicating dual endocytotic pathways (clathrin-dependent and lipid raft-mediated) .
Polyclonal antibodies (e.g., 26523-1-AP) show broader reactivity (human, mouse, rat, pig), while monoclonal variants (e.g., TD.1) are species-specific, enabling targeted studies .
Clathrin heavy chain 1 (CLTC), a 191.615 kDa protein belonging to the clathrin heavy chain family, serves as a critical structural component in cellular vesicle formation and trafficking. CLTC is essential for clathrin-mediated endocytosis, a fundamental process through which cells internalize molecules from their environment. Research involving CLTC enables investigation of cellular uptake mechanisms, receptor trafficking, and membrane dynamics across multiple cell types. The protein is also known by several alternative names including CHC, CHC17, and CLH-17L2 . As a highly conserved protein across mammalian species, CLTC studies provide insights into fundamental cellular functions with implications for understanding disease mechanisms where vesicular trafficking is disrupted.
CLTC antibodies specifically target the heavy chain component of clathrin-coated structures, making them ideal for investigating clathrin-dependent trafficking pathways. Unlike caveolin markers (which detect caveolae-mediated endocytosis) or dynamin antibodies (which identify multiple endocytic routes), CLTC antibodies selectively label clathrin-coated pits and vesicles. Western blot analysis consistently detects CLTC at approximately 180 kDa across multiple cell and tissue types, including human cell lines (HeLa, A431, HepG2, HL-60, Raji), and brain tissues from multiple species (mouse, rat, pig) . For comprehensive endocytosis studies, researchers should employ CLTC antibodies alongside markers for alternative pathways to distinguish between different internalization mechanisms. Choose CLTC when specifically examining receptor-mediated endocytosis processes that utilize clathrin-coated structures.
The selection between monoclonal and polyclonal CLTC antibodies depends on experimental requirements:
Antibody Type | Advantages | Optimal Applications | Example Catalog | Host |
---|---|---|---|---|
Monoclonal (e.g., clone 6D3) | High specificity, consistent lot-to-lot reproducibility, reduced background | Flow cytometry, IHC of specific epitopes | M03134-2 | Mouse |
Polyclonal | Multiple epitope recognition, enhanced signal amplification, greater tolerance to protein denaturation | Western blot, general protein detection | 26523-1-AP | Rabbit |
Monoclonal antibodies like clone 6D3 demonstrate exceptional specificity, detecting a discrete band at 180 kDa in Western blot analysis across human, mouse, and rat samples . This specificity makes them particularly valuable for flow cytometry applications where signal-to-noise ratio is critical. In contrast, polyclonal antibodies offer broader epitope recognition, potentially increasing detection sensitivity in applications like Western blotting, where they can be used at dilutions ranging from 1:2000 to 1:10000 . For experiments requiring detailed localization studies, monoclonal antibodies generally provide more precise subcellular detection with minimal background.
Successful Western blot analysis with CLTC antibodies requires careful optimization of sample preparation, antibody dilution, and detection conditions. Based on validated protocols:
Sample preparation: Load 30 μg of protein lysate per lane under reducing conditions. CLTC is effectively extracted from various tissues including brain, intestine, and cancer cell lines .
Gel electrophoresis: Use 5-20% gradient SDS-PAGE gels run at 70V (stacking)/90V (resolving) for 2-3 hours to achieve proper separation of this high molecular weight protein .
Transfer conditions: Transfer proteins to nitrocellulose membrane at 150mA for 50-90 minutes to ensure complete transfer of the large CLTC protein (180-192 kDa) .
Blocking: Block membranes with 5% non-fat milk in TBS for 1.5 hours at room temperature .
Primary antibody incubation: Incubate with anti-CLTC antibody at optimized concentration (0.5 μg/mL for monoclonal antibodies; 1:2000-1:10000 dilution for polyclonal antibodies) overnight at 4°C .
Secondary antibody: For HRP-conjugated secondary antibodies, use 1:10000 dilution and incubate for 1.5 hours at room temperature .
Detection: Develop using enhanced chemiluminescent (ECL) detection systems for optimal visualization of the specific 180 kDa CLTC band .
This methodology has been validated with multiple cell lines including HeLa, A431, and various tissue lysates, consistently producing clear, specific detection of CLTC .
Immunohistochemical detection of CLTC requires tissue-specific optimization of antigen retrieval methods and antibody concentrations:
Tissue Type | Recommended Antigen Retrieval | Antibody Concentration | Incubation Conditions | Secondary Detection System |
---|---|---|---|---|
Pancreatic cancer | EDTA buffer (pH 8.0) | 2 μg/ml (monoclonal) | Overnight at 4°C | Biotin-streptavidin-DAB |
Renal carcinoma | EDTA buffer (pH 8.0) | 2 μg/ml (monoclonal) | Overnight at 4°C | Biotin-streptavidin-DAB |
Tonsil tissue | EDTA buffer (pH 8.0) | 2 μg/ml (monoclonal) | Overnight at 4°C | Biotin-streptavidin-DAB |
Lung cancer | EDTA buffer (pH 8.0) | 2 μg/ml (polyclonal) | Overnight at 4°C | Biotin-streptavidin-DAB |
Intestinal tissue | EDTA buffer (pH 8.0) | 2 μg/ml (polyclonal) | Overnight at 4°C | Biotin-streptavidin-DAB |
Breast cancer | TE buffer (pH 9.0) | 1:500-1:2000 (polyclonal) | Overnight at 4°C | Biotin-streptavidin-DAB |
All protocols require tissue section blocking with 10% goat serum prior to primary antibody incubation . For challenging tissue types, extending the antigen retrieval time or using enzymatic retrieval methods may improve detection. When transitioning to HRP-conjugated primary antibodies, eliminate the secondary antibody step and adjust incubation time to 2 hours at room temperature, followed by direct substrate addition. Validation studies demonstrate that these protocols yield specific cytoplasmic and membrane-associated staining patterns consistent with CLTC's role in vesicular structures.
Rigorous experimental controls are critical for confirming CLTC antibody specificity:
Positive cellular controls: Include validated cell lines known to express CLTC (HeLa, HepG2, U20S) in each experimental series .
Tissue controls: Use brain tissue (mouse, rat, or pig) as a positive control due to consistently high CLTC expression .
Isotype controls: For flow cytometry, include matched isotype controls (e.g., mouse IgG for monoclonal antibodies) at identical concentrations to assess non-specific binding .
Antibody omission controls: Process samples without primary antibody to evaluate secondary antibody background and endogenous peroxidase activity, particularly important when using HRP-conjugated detection systems .
Knockdown validation: When possible, include CLTC knockdown or knockout samples to verify antibody specificity through signal reduction or elimination .
Cross-reactivity testing: Test antibodies against related proteins (e.g., CLTCL1) to ensure target specificity.
Multiple antibody validation: Use alternative CLTC antibodies recognizing different epitopes to confirm staining patterns.
These controls collectively ensure that observed signals truly represent CLTC rather than technical artifacts or cross-reactivity with other proteins, providing confidence in experimental interpretations.
HRP conjugation directly to primary CLTC antibodies offers several advantages over conventional two-step detection:
Enhanced sensitivity: Direct HRP conjugation eliminates signal loss that occurs during secondary antibody binding, potentially increasing detection sensitivity by 1.5-2.5 fold, particularly beneficial for detecting low CLTC expression in certain cell types.
Reduced background: The elimination of secondary antibodies decreases non-specific binding events, resulting in improved signal-to-noise ratios in both Western blot and immunohistochemistry applications.
Time efficiency: HRP-conjugated primary antibodies streamline protocols by removing secondary antibody incubation and washing steps, reducing experimental time by approximately 2-3 hours.
Multiplexing capability: Direct conjugation facilitates multiplex experiments with antibodies from the same host species, enabling simultaneous detection of CLTC with other endocytic markers.
Effective co-localization studies require carefully optimized immunofluorescence protocols:
Cell preparation: Culture cells on coverslips and fix with 4% paraformaldehyde for 15 minutes at room temperature. For U20S cells, enzyme antigen retrieval using IHC enzyme antigen retrieval reagent for 15 minutes improves CLTC detection .
Permeabilization: Use 0.1-0.2% Triton X-100 for 10 minutes to allow antibody access to intracellular CLTC structures without disrupting membrane architecture.
Blocking: Block with 10% goat serum to minimize non-specific binding .
Primary antibody incubation: For co-localization studies, use rabbit anti-CLTC antibody (5 μg/mL) concurrently with mouse antibodies against endosomal markers (EEA1, Rab5, Rab7). Incubate overnight at 4°C .
Secondary antibody selection: Use fluorophore-conjugated secondary antibodies with spectral separation (e.g., DyLight®488 for CLTC and Alexa Fluor 594 for endosomal markers) at 1:100-1:200 dilution .
Nuclear counterstaining: Counterstain with DAPI (1 μg/mL) to provide cellular context .
Mounting and imaging: Mount with anti-fade medium and image using confocal microscopy with appropriate filter sets.
For quantitative co-localization analysis, capture z-stack images (0.3-0.5 μm intervals) and analyze using Pearson's or Mander's correlation coefficients. This approach has successfully demonstrated CLTC co-localization with early endosomal markers in multiple cell types, including HeLa and HepG2 cells .
Flow cytometric detection of intracellular CLTC requires specific protocol modifications:
Cell preparation: Harvest 1×10^6 cells, wash in PBS, and fix with 4% paraformaldehyde for 15 minutes at room temperature .
Permeabilization: Use commercial permeabilization buffer specifically designed for intracellular flow cytometry to allow antibody access while maintaining cell integrity .
Blocking: Block with 10% normal goat serum to reduce background staining .
Antibody concentration: For monoclonal CLTC antibodies, use 1 μg per 1×10^6 cells; for polyclonal antibodies, use 0.40 μg per 10^6 cells in 100 μl suspension .
Incubation conditions: Incubate for 30 minutes at 20°C rather than at 4°C to improve antibody penetration into fixed cells .
Secondary detection: For non-conjugated primary antibodies, use DyLight®488-conjugated goat anti-mouse/rabbit IgG at 5-10 μg per 1×10^6 cells .
Controls: Include both isotype control (matched mouse/rabbit IgG) and unstained samples to set proper gating parameters .
Flow cytometric analysis of HepG2 cells using this protocol successfully detects intracellular CLTC with clear separation from control populations . For HRP-conjugated CLTC antibodies, substitute a tyramide signal amplification system compatible with flow cytometry to generate fluorescent signals from the HRP activity, allowing integration into multiparameter flow cytometry panels while maintaining sensitivity.
Troubleshooting weak CLTC signals requires systematic optimization:
Issue | Potential Cause | Solution |
---|---|---|
Weak/absent signal | Insufficient protein loading | Increase sample loading to 30-50 μg per lane |
Inefficient transfer of high MW protein | Extend transfer time to 90 minutes; reduce current to 150mA | |
Suboptimal primary antibody concentration | For monoclonal: increase to 1-2 μg/mL; For polyclonal: use 1:2000 dilution | |
Multiple bands | Protein degradation | Add protease inhibitors to lysis buffer; keep samples on ice |
Cross-reactivity | Try alternative CLTC antibody clone; increase blocking to 5% milk for 2 hours | |
High background | Insufficient blocking | Increase blocking time to 2 hours; use 5% BSA instead of milk |
Excessive antibody concentration | Dilute primary antibody further; reduce incubation to 2 hours at RT |
For HRP-conjugated primary antibodies specifically, dilute 2-5 fold more than non-conjugated versions to prevent excessive background, and reduce substrate incubation time. CLTC consistently appears at approximately 180 kDa in multiple cell types and tissues , so any significant deviation from this molecular weight should be carefully validated. Consider using gradient gels (5-20%) for optimal separation and resolution of this high molecular weight protein .
Variations in CLTC staining patterns reflect its biological functions and should be interpreted accordingly:
Punctate cytoplasmic staining: Represents clathrin-coated vesicles and is the predominant pattern in most cell types. Particularly prominent in cell lines with high endocytic activity (HepG2, HeLa) .
Perinuclear concentration: Often observed in secretory cells and indicates association with the trans-Golgi network for protein sorting. Evident in pancreatic tissue samples .
Membrane-associated staining: Represents clathrin-coated pits at the plasma membrane during active endocytosis. More prominent in highly endocytic cell types and tissues with significant receptor-mediated uptake .
Diffuse cytoplasmic signal: May indicate technical issues or, in certain pathological conditions, disruption of normal clathrin structures. Not typically observed in properly fixed healthy cells .
Nuclear staining: Generally considered non-specific for CLTC antibodies and should prompt further validation.
Immunohistochemical analysis of different tissues demonstrates that CLTC staining intensity varies naturally, with particularly strong signals in renal carcinoma, pancreatic cancer, and tonsil tissue . These variations reflect tissue-specific requirements for clathrin-mediated endocytosis and intracellular trafficking, rather than technical artifacts. Comparative analysis across multiple tissue types should consider these biological differences when interpreting relative expression levels.
Managing cross-reactivity in complex tissues requires several targeted approaches:
Antibody selection: Prioritize monoclonal antibodies (e.g., clone 6D3) for applications in complex tissues, as they demonstrate higher specificity than polyclonal alternatives .
Titration optimization: Perform careful antibody titration experiments to identify the minimum concentration that produces specific signal. For IHC, start with 2 μg/ml for monoclonal antibodies and adjust based on signal-to-noise ratio .
Extended blocking: Increase blocking time to 2 hours with 10% serum from the same species as the secondary antibody to reduce non-specific binding .
Absorption controls: Pre-incubate antibody with recombinant CLTC protein prior to staining to confirm specificity through signal elimination.
Alternative fixation: If formalin-fixed tissues show high background, consider alternative fixatives (e.g., acetone or methanol) that may preserve the CLTC epitope while reducing cross-reactivity.
Sequential staining: For multiplexed detection, implement sequential rather than simultaneous staining protocols with complete antibody stripping between rounds.
Peptide competition: Use the immunizing peptide in parallel staining experiments to identify specific vs. non-specific signals.
For tissues known to present cross-reactivity challenges (e.g., brain tissue with high endogenous peroxidase activity), modify the protocol to include additional peroxidase blocking steps (3% H2O2 for 15 minutes) before antibody incubation . When using HRP-conjugated primary antibodies, these blocking steps become particularly critical to prevent false-positive signals.
Super-resolution imaging of CLTC requires specific protocol adaptations:
Sample preparation: Culture cells on high-precision coverslips (#1.5H, 170±5 μm thickness) to minimize spherical aberrations. For tissue sections, use 5 μm or thinner sections to reduce light scattering.
Fixation optimization: Use 4% paraformaldehyde with 0.2% glutaraldehyde to better preserve nanoscale structures. The addition of glutaraldehyde improves structural integrity critical for resolving individual clathrin-coated pits.
Antibody selection: Use monoclonal antibodies (like clone 6D3) that target specific CLTC epitopes for highest precision in structure localization .
Fluorophore selection: For STORM/PALM techniques, conjugate antibodies to photoswitchable fluorophores (Alexa Fluor 647 or Atto 488); for STED microscopy, use STED-optimized dyes (STAR 580, STAR 635P).
Buffer composition: For STORM imaging, use oxygen-scavenging buffer systems containing glucose oxidase, catalase, and thiol compounds to enhance fluorophore photoswitching.
Immunolabeling density: Use F(ab) fragments rather than full IgG to reduce the distance between fluorophore and target, improving localization precision.
Image acquisition: Collect 10,000-30,000 frames at 10-100 Hz for STORM/PALM reconstruction, adjusting laser power to maintain optimal fluorophore switching rates.
This approach enables visualization of individual clathrin-coated structures at 20-30 nm resolution, revealing the characteristic triskelion assembly of CLTC that cannot be resolved with conventional microscopy. For correlative studies, consider combining with electron microscopy to validate structural observations.
Quantitative analysis of CLTC-mediated endocytosis in live cells requires:
Construct selection: Utilize CLTC-GFP/RFP fusion constructs that maintain functional endocytic capacity. Alternatively, use Fab fragments of CLTC antibodies conjugated to cell-permeable fluorophores for endogenous protein labeling.
Transfection optimization: For exogenous expression, maintain low expression levels (30-50% of endogenous CLTC) to prevent disruption of normal endocytic dynamics. Use inducible expression systems to control protein levels precisely.
Image acquisition parameters:
Temporal resolution: 1-2 frames/second to capture the typical 60-90 second lifetime of clathrin-coated pits
Spatial sampling: Use 100-150 nm pixel size with high NA objectives (≥1.4)
Imaging duration: 5-10 minutes per position to track complete endocytic cycles
Illumination: Employ TIRF or Highly Inclined Laminated Optical sheet microscopy to reduce background
Quantification metrics:
Density: Number of CLTC structures per μm²
Dynamics: Lifetime distribution of CLTC structures (from appearance to disappearance)
Size: Maximum intensity as proxy for pit size
Mobility: Mean square displacement analysis of tracked structures
Cargo incorporation: Dual-color analysis with labeled cargo proteins
Analysis software: Utilize particle tracking algorithms (e.g., TrackMate in ImageJ or custom MATLAB scripts) for automated detection and analysis of hundreds of endocytic events per cell.
This approach enables robust quantification of CLTC-mediated endocytosis under various experimental conditions, including drug treatments, genetic manipulations, or disease models. For validation, compare findings with fixed-cell immunofluorescence using antibodies documented to work in HeLa or HepG2 cells .
Integration of CLTC antibodies into multiplexed proteomic approaches requires strategic planning:
Mass cytometry (CyTOF) applications:
Conjugate CLTC antibodies to rare earth metals (e.g., 159Tb, 169Tm) with minimal spectral overlap
Validate metal-conjugated antibodies using spike-in controls of known CLTC-expressing cells
Implement dual-labeling with alternative CLTC clones to confirm specificity
Combine with markers for endocytic compartments (Rab5, Rab7, Rab11) labeled with distinct metals
Multiplexed immunofluorescence:
Employ cyclic immunofluorescence with CLTC antibodies in initial rounds
Use complete antibody stripping between cycles (pH 2.5 glycine buffer or commercial antibody removal solutions)
Validate signal elimination before subsequent staining rounds
Implement tissue-specific autofluorescence quenching protocols
Proximity ligation approaches:
Combine CLTC antibodies with antibodies against interaction partners
Use species-specific PLA probes compatible with the selected CLTC antibody host
Validate with known CLTC interactors (AP2 complex components)
Quantify interaction frequencies in different subcellular compartments
Spatial proteomics integration:
For CODEX or MIBI-TOF approaches, validate CLTC antibody compatibility with metal conjugation chemistries
Test signal preservation after multiple staining/stripping cycles
For each new tissue type, establish optimal antibody concentration using titration experiments
These approaches enable comprehensive mapping of CLTC-associated protein networks across different cellular contexts. When transitioning these techniques to tissue samples, begin with validation in tissues known to show strong CLTC staining (tonsil, renal tissue, pancreatic tissue) before proceeding to more challenging samples.
The landscape of CLTC detection is evolving with several technological innovations:
Nanobody and single-domain antibody approaches: These smaller binding proteins (15-20 kDa versus 150 kDa for traditional antibodies) offer superior tissue penetration and reduced distance between target and label, improving spatial resolution in microscopy applications by approximately 10-15 nm.
Site-specific conjugation techniques: Controlled conjugation chemistry ensures precise labeling at defined antibody sites rather than random lysine residues, improving batch-to-batch consistency and maintaining antigen-binding capacity of CLTC antibodies.
Recombinant antibody production: Moving from hybridoma-based to recombinant production platforms reduces lot-to-lot variation for monoclonal antibodies like clone 6D3 , ensuring consistent experimental results.
Bispecific antibody formats: Dual-specificity antibodies targeting both CLTC and cargo proteins enable direct visualization of specific cargo-selective endocytic events without requiring multiple labeling steps.
Intrabodies and genetically encoded sensors: Antibody-based intracellular sensors that can be expressed in living cells provide dynamic readouts of CLTC activity without requiring cell permeabilization.
These technologies collectively enhance the specificity, sensitivity, and versatility of CLTC detection methods. As these approaches mature, researchers should validate new reagents against established standards such as the monoclonal and polyclonal antibodies documented in the search results .
Successful integration of CLTC antibodies in correlative light and electron microscopy (CLEM) requires:
Fixation compatibility: Use 4% paraformaldehyde with 0.1% glutaraldehyde to balance antigen preservation for immunolabeling with ultrastructural preservation required for EM.
Antibody selection: Choose primary antibodies validated across multiple applications, as those with demonstrated performance in both Western blot and immunofluorescence applications are more likely to work in CLEM protocols .
Gold conjugation for immuno-EM: For pre-embedding approaches, use ultra-small gold particles (1-2 nm) conjugated to secondary antibodies with silver enhancement for optimal visualization.
Section thickness optimization: For post-embedding immuno-EM, use 70-90 nm sections on EM grids to balance structural integrity with antibody accessibility.
Fiducial markers: Incorporate multimodal fiducial markers (fluorescent and electron-dense) during sample processing to ensure precise registration between light and electron microscopy images.
Sequential detection strategy:
Begin with fluorescence imaging of CLTC
Document coordinates precisely
Process the same sample for EM
Relocate the same regions using fiducial markers
Correlate structures between modalities
This approach enables the correlation of functional information from fluorescence with the ultrastructural details provided by EM, revealing both the molecular composition and morphology of clathrin-coated structures. Validation studies demonstrate that CLTC antibodies from commercial sources successfully label clathrin-coated vesicles that can be identified by their characteristic coat in EM images .
CLTC antibodies are playing crucial roles in elucidating disease mechanisms:
Cancer research: Immunohistochemical studies using CLTC antibodies reveal altered endocytic dynamics in multiple cancer types, including pancreatic, renal, and lung carcinomas . The specific staining patterns in these tissues enable quantitative assessment of changes in clathrin-dependent trafficking that correlate with disease progression.
Neurodegenerative disorders: CLTC antibodies help visualize abnormal clathrin distribution in neuronal tissues, contributing to understanding how endocytic defects may participate in pathologies such as Alzheimer's and Parkinson's diseases.
Infectious disease mechanisms: CLTC antibodies enable tracking of pathogen entry through clathrin-mediated endocytosis, revealing how viruses and bacteria exploit this pathway.
Genetic disorders: The MRD56 genetic disorder linked to CLTC mutations can be studied using specific antibodies that distinguish between normal and mutant protein localization .
Drug resistance mechanisms: CLTC antibodies help reveal how altered endocytosis contributes to therapeutic resistance in cancer by affecting drug internalization and receptor recycling.
For these studies, researchers should select antibodies validated across multiple tissue types relevant to the disease under investigation. The immunohistochemical protocols described for human cancer tissues provide starting points for developing disease-specific applications, with antibody concentrations and antigen retrieval methods optimized for the specific tissue context.
Selection of optimal CLTC antibodies should follow these evidence-based criteria:
Application validation: Choose antibodies specifically validated for your intended application. For example, clone 6D3 (monoclonal) and catalog #26523-1-AP (polyclonal) have demonstrated performance across multiple applications including Western blot, IHC, IF, and flow cytometry .
Species reactivity: Verify confirmed reactivity with your experimental species. The documented antibodies show reactivity with human, mouse, rat, and pig samples, with consistent detection of the expected 180 kDa band across species .
Epitope location: Consider epitope location relative to functional domains or potential post-translational modifications in CLTC. This information may affect antibody performance in certain applications.
Clone type selection: For applications requiring high specificity (flow cytometry, specific epitope detection), prioritize monoclonal antibodies like clone 6D3 . For applications benefiting from signal amplification (Western blot of low-abundance samples), consider polyclonal antibodies .
Publication record: Review antibodies with demonstrated performance in peer-reviewed literature, particularly for challenging applications or tissues.
Lot-to-lot consistency: For longitudinal studies, consider antibody production methods that ensure consistency, with recombinant antibodies offering advantages over traditional hybridoma-derived antibodies.
Validation controls: Confirm that antibodies have been validated against proper controls, including CLTC knockdown/knockout samples when available .
By applying these criteria systematically, researchers can select CLTC antibodies most likely to perform reliably in their specific experimental context, minimizing technical variability and ensuring reproducible results.
Preserving the activity of HRP-conjugated antibodies requires specific handling practices:
Storage temperature: Store at -20°C in a non-frost-free freezer to prevent freeze-thaw cycles that damage both antibody binding capacity and HRP enzymatic activity .
Buffer composition: Optimal storage buffer should contain 50% glycerol and PBS with 0.02% sodium azide at pH 7.3 . Note that sodium azide can inhibit HRP activity and must be removed before use through dialysis or dilution.
Aliquoting strategy: Upon receipt, divide into small single-use aliquots (5-10 μL) to avoid repeated freeze-thaw cycles. Each cycle can reduce activity by approximately 10-20%.
Thawing procedure: Thaw rapidly at room temperature and immediately return unused portion to -20°C. Avoid keeping at room temperature longer than necessary.
Working dilution preparation: Always prepare working dilutions fresh on the day of use. Diluted antibody solutions lose activity at approximately 5-10% per day at 4°C.
Stabilizers for working solutions: Add 1-2% BSA to diluted working solutions to maintain antibody stability during immunoassays.
Light exposure: Minimize exposure to light, which can generate reactive oxygen species that damage both the antibody and HRP enzyme.
Long-term stability: HRP-conjugated antibodies typically maintain >80% activity for 6-12 months when stored properly. Monitor performance over time using consistent positive controls.
Contamination prevention: Use sterile technique when handling antibody solutions to prevent microbial growth that can degrade protein components.
By following these practices, researchers can maintain optimal activity of HRP-conjugated CLTC antibodies, ensuring consistent experimental results throughout the reagent's useful life.
CLTC antibodies are finding novel applications across multiple research frontiers:
Organoid research: CLTC antibodies enable investigation of endocytic trafficking in 3D organoid systems, providing insights into tissue-specific endocytic processes in more physiologically relevant models than traditional 2D culture.
Super-resolution microscopy: Advanced imaging techniques combined with highly specific CLTC antibodies are revealing previously unobservable details of clathrin cage assembly and disassembly kinetics at the nanoscale level.
Liquid biopsy applications: Detection of extracellular vesicle-associated CLTC in biological fluids is emerging as a potential biomarker approach for diseases involving altered vesicular trafficking.
Cryo-electron tomography integration: Correlating fluorescent CLTC antibody labeling with cryo-ET provides unprecedented insights into the native 3D architecture of clathrin-coated structures in their cellular context.
Machine learning-enhanced image analysis: Combining CLTC immunolabeling with AI-based image analysis enables automated quantification of endocytic dynamics across thousands of cells, identifying subtle phenotypes impossible to detect manually.
Spatial transcriptomics correlation: Integrating CLTC protein detection with spatial transcriptomics reveals relationships between local gene expression patterns and endocytic activity in complex tissues.
Drug delivery optimization: CLTC antibodies help characterize clathrin-dependent uptake mechanisms of nanoparticle-based therapeutics, informing rational design of delivery systems targeting this pathway.