The CCT4 Antibody (clone 67455-1-Ig) recognizes the delta subunit of the TRiC complex, a molecular chaperone essential for folding cytoskeletal proteins like actin and tubulin . It is produced in mice and validated for Western blot (WB), immunofluorescence (IF), and ELISA applications across human, mouse, rabbit, and pig samples .
Western Blot: Detected in LNCaP, HeLa, HEK-293, HepG2, and Jurkat cells, as well as brain tissues from pigs, rabbits, and mice .
Drosophila Studies: CCT4 knockdown reduced cell size and number in wing discs by 21% and 23%, respectively, and increased apoptosis via Caspase-3 activation .
Mechanism: CCT4 interacts with insulin/TOR signaling components (e.g., TOR, S6K) and regulates Cyclin E expression .
Hepatocellular Carcinoma (HCC):
CCT4 is a component of the chaperonin-containing T-complex (TRiC), a molecular chaperone complex essential for protein folding upon ATP hydrolysis. The TRiC complex plays a crucial role in mediating the folding of WRAP53/TCAB1, thereby regulating telomere maintenance. Furthermore, as part of the TRiC complex, CCT4 may be involved in the assembly of the BBSome, a complex implicated in ciliogenesis, regulating the transport of vesicles to cilia. The TRiC complex also contributes to the folding of actin and tubulin.
CCT4 (Chaperonin Containing TCP1 Subunit 4), also known as T-complex protein 1 subunit delta, is a component of the chaperonin-containing T-complex (TRiC), a molecular chaperone complex that assists in protein folding upon ATP hydrolysis. CCT4 has significant importance in cellular biology as it contributes to the folding of various proteins, particularly actin and tubulin. As part of the TRiC complex, it plays critical roles in regulating telomere maintenance and the assembly of BBSome, a complex involved in ciliogenesis that regulates vesicle transport to cilia . Understanding CCT4 function is essential for research into protein folding mechanisms, cellular stress responses, and various pathological conditions including cancer. Recent studies have highlighted CCT4's potential role as a biomarker in hepatocellular carcinoma and other diseases .
CCT4 antibodies are employed across multiple research applications with varying optimization requirements:
When selecting a CCT4 antibody for your research, consider the specific application requirements and choose antibodies validated for your intended application and species of interest .
Validating antibody specificity is crucial for generating reliable research data. For CCT4 antibodies, implement the following validation strategies:
Positive and negative controls: Use cell lines with known CCT4 expression levels. Several antibodies have been validated in cell lines such as HeLa, MCF-7, HEK-293, and HepG2 .
Knockdown/knockout validation: Utilize CCT4 siRNA or CRISPR-Cas9 approaches to create CCT4-depleted cells as negative controls. Published studies have employed this approach to confirm antibody specificity .
Multiple antibody approach: Use different antibodies targeting distinct epitopes of CCT4 to confirm consistent detection patterns.
Western blot analysis: Confirm the detected band corresponds to the expected molecular weight (55-58 kDa) for CCT4 .
Peptide competition assay: Pre-incubate the antibody with a blocking peptide (such as the immunogen peptide) to demonstrate signal specificity .
Cross-reactivity testing: Test the antibody against related proteins, particularly other CCT family members, to ensure specificity within this closely related protein family.
Remember that antibody validation should ideally include both positive results (detection in samples known to express CCT4) and negative controls to conclusively demonstrate specificity .
CCT4 expression patterns show distinct variations across tissues and in disease states:
Normal tissues:
CCT4 is expressed in various tissues, with particularly strong expression in tissues with high protein folding demands. The highest expression levels have been observed in brain tissues, particularly in neuronal cells .
Cancer tissues:
Significant upregulation of CCT4 expression has been documented in multiple cancer types, with hepatocellular carcinoma (HCC) being particularly well-studied:
When investigating CCT4 in pathological conditions, it's recommended to use a panel of antibodies and multiple detection methods to accurately characterize expression changes. The observed correlation between CCT4 upregulation and clinical parameters suggests it may serve as a potential prognostic biomarker and therapeutic target, particularly in HCC .
Studying CCT4's interactions within the TRiC complex requires specialized techniques that preserve native protein-protein interactions:
Co-immunoprecipitation (Co-IP):
Use CCT4 antibodies to pull down the protein along with its binding partners
Western blot for other TRiC components (CCT1-8) to confirm interactions
Gentle lysis conditions (non-ionic detergents like NP-40) help preserve complex integrity
Proximity ligation assay (PLA):
Enables visualization of protein-protein interactions in situ
Requires primary antibodies from different host species for CCT4 and its interacting partners
Can provide spatial information about interactions within subcellular compartments
Cross-linking mass spectrometry:
Chemical cross-linking stabilizes transient interactions
MS analysis identifies cross-linked peptides, providing structural information
Particularly valuable for understanding the quaternary structure of the TRiC complex
Bioluminescence resonance energy transfer (BRET)/Fluorescence resonance energy transfer (FRET):
Tag CCT4 and potential binding partners with appropriate donor/acceptor pairs
Monitor real-time interactions in live cells
Useful for studying dynamics of complex assembly/disassembly
When investigating CCT4's role in telomere maintenance, consider chromatin immunoprecipitation (ChIP) experiments to assess potential associations with telomeric regions. For BBSome assembly studies, combine co-IP with sucrose gradient fractionation to isolate complexes at different assembly stages .
Investigating CCT4's role in stress response and proteostasis requires a multifaceted experimental approach:
Stress induction protocols:
Heat shock: Expose cells to elevated temperatures (42-45°C) for 30-60 minutes
Oxidative stress: Treat with hydrogen peroxide (100-500 μM) or paraquat
Proteasome inhibition: Apply MG132 (5-10 μM) for 4-24 hours
ER stress: Thapsigargin (1-5 μM) or tunicamycin treatment
Monitor CCT4 expression, localization, and activity changes under each condition
CCT4 modulation techniques:
CRISPR-Cas9 knockout: For complete elimination of CCT4
siRNA knockdown: For transient reduction (typically 70-90%)
Overexpression systems: To assess gain-of-function effects
Compare effects on proteostasis markers across these conditions
Proteostasis assessment:
Protein aggregation assays: Filter trap assay, detergent insolubility fractionation
Protein folding reporters: Luciferase refolding assay, GFP-fusion protein folding
Proteasome activity assays: Fluorogenic substrate cleavage
Autophagy flux measurements: LC3-II/LC3-I ratio, p62 degradation
Client protein analysis:
Identify CCT4-dependent substrates using immunoprecipitation followed by mass spectrometry
Monitor folding kinetics of known clients (e.g., actin, tubulin) using pulse-chase experiments
Assess effects of CCT4 depletion on client protein solubility and function
When analyzing results, consider that the TRiC complex functions as a cohesive unit, so changes in CCT4 may affect the entire complex's functionality. Include additional chaperone markers (HSP70, HSP90) to determine whether observed effects are specific to TRiC or reflect broader changes in the proteostasis network .
The relationship between CCT4 expression and cancer progression has been extensively studied, particularly in hepatocellular carcinoma:
For researchers studying CCT4 in cancer, it's advisable to employ multiparameter analysis combining expression levels with genetic alterations and correlate these with clinical parameters. The evidence suggests CCT4 could serve as both a prognostic biomarker and potential therapeutic target, particularly in HCC, though further validation studies are needed .
Sample preparation is critical for successful CCT4 detection across different applications:
Cell lysis: Use RIPA buffer or NP-40 based lysis buffers with protease inhibitors
Protein estimation: BCA or Bradford assay to ensure equal loading
Sample denaturation: Heat at 95°C for 5 minutes in Laemmli buffer with DTT or β-mercaptoethanol
Recommended loading: 20-50 μg of total protein per lane
Separation: 10-12% SDS-PAGE gels provide optimal resolution for CCT4 (55-58 kDa)
Fixation: 10% neutral buffered formalin for 24-48 hours
Processing: Standard paraffin embedding
Sectioning: 4-5 μm sections
Antigen retrieval: Most effective with TE buffer pH 9.0 (alternative: citrate buffer pH 6.0)
Blocking: 5-10% normal serum (matching secondary antibody host) with 1% BSA
Fixation: 4% paraformaldehyde for 15-20 minutes at room temperature
Permeabilization: 0.1-0.5% Triton X-100 for 5-10 minutes
Blocking: 5% BSA or 10% normal serum for 30-60 minutes
Antibody incubation: Overnight at 4°C or 1-2 hours at room temperature
Fixation: 2-4% paraformaldehyde
Permeabilization: Critical for intracellular CCT4 detection using 0.1% saponin or commercial permeabilization buffers
Blocking: 5% normal serum or 2% BSA
Single cell preparation: Gentle cell dissociation to maintain epitope integrity
Always validate these protocols for your specific cell type or tissue and adjust parameters as needed for optimal results.
Researchers commonly encounter several issues when working with CCT4 antibodies. Here are solutions to the most frequent problems:
Increase protein loading (50-100 μg)
Optimize primary antibody concentration (try serial dilutions)
Extend primary antibody incubation (overnight at 4°C)
Use enhanced chemiluminescence (ECL) substrates with higher sensitivity
Check transfer efficiency with reversible protein stains (Ponceau S)
Increase blocking time (2 hours) and concentration (5-10% serum)
Add 0.1-0.3% Triton X-100 to antibody diluent
Include 0.05-0.1% Tween-20 in wash buffers
Use more stringent washing (increase number and duration of washes)
Try alternative blocking agents (2% BSA, 5% milk, commercial blockers)
Use fresh sample preparation with protease inhibitors
Include phosphatase inhibitors if studying post-translational modifications
Increase gel percentage for better resolution
Verify with CCT4 knockout/knockdown controls
Use affinity-purified antibodies
Validate with peptide competition assays
Employ antibodies targeting non-conserved regions of CCT4
Pre-adsorb antibody with related proteins
Include knockout/knockdown controls to verify specificity
Standardize sample preparation protocols
Include internal loading controls (β-actin, GAPDH)
Prepare larger batches of antibody dilutions
Document lot numbers and maintain consistent reagent sources
Employ positive controls (cell lines with known CCT4 expression)
When troubleshooting persistent issues, consider testing multiple antibodies against different epitopes of CCT4 to determine if the problem is antibody-specific or related to your experimental system.
Accurate quantitative analysis of CCT4 expression requires rigorous methodology and appropriate controls:
Ensure linear range of detection:
Load a concentration gradient of samples
Verify signal is within linear range of detection system
Normalization strategies:
Use housekeeping proteins (GAPDH, β-actin) for loading control
Consider total protein normalization (Ponceau S, SYPRO Ruby) for more accurate results
Image acquisition:
Use cooled CCD camera systems rather than film
Avoid signal saturation
Analysis software:
Scoring systems:
H-score (intensity × percentage of positive cells)
Allred score (combines intensity and proportion)
Digital image analysis with color deconvolution
Observer variability reduction:
Blinded analysis by multiple observers
Use of automated scanning and analysis platforms
Controls for normalization:
Standardization:
Use calibration beads to standardize fluorescence intensity
Establish defined gates based on negative controls
Analysis parameters:
Mean/median fluorescence intensity (MFI)
Percentage of positive cells above threshold
Controls:
Reference gene selection:
Use multiple reference genes validated for your experimental system
Verify reference gene stability across conditions
Analysis methods:
ΔΔCt method with appropriate normalization
Standard curve method for absolute quantification
When comparing CCT4 expression across different conditions or disease states, always process and analyze all samples simultaneously to minimize batch effects. For clinical correlations, employ multivariate statistical analyses to account for confounding variables .
CCT4 antibodies are valuable tools for investigating its potential as a cancer biomarker, particularly in hepatocellular carcinoma (HCC). Here's a methodological approach for such studies:
Tissue microarray (TMA) analysis:
Multiplex immunofluorescence:
Co-stain for CCT4 and other cancer markers
Assess cellular localization and co-expression patterns
Combine with markers of proliferation (Ki-67) or other oncogenic pathways
Liquid biopsy approaches:
Develop assays to detect CCT4 in patient serum or circulating tumor cells
Compare with established biomarkers (e.g., AFP for HCC)
Evaluate sensitivity and specificity for early detection
Prognostic correlation studies:
Stratify patients based on CCT4 expression levels
Perform Kaplan-Meier survival analysis
Calculate hazard ratios using Cox regression
Compare with established prognostic markers
Therapeutic response prediction:
Assess CCT4 expression before and after treatment
Correlate expression levels with response to specific therapies
Develop predictive models incorporating CCT4 status
Investigating CCT4's functional roles in protein folding requires specialized techniques spanning biochemical, cellular, and structural approaches:
In vitro folding assays:
Luciferase refolding: Measure refolding of denatured luciferase in the presence/absence of functional CCT4
Target protein folding kinetics: Monitor folding of known CCT4 clients (actin, tubulin) using circular dichroism or fluorescence spectroscopy
ATP hydrolysis coupling: Correlate ATPase activity with folding efficiency
Client protein identification:
Co-immunoprecipitation with CCT4-specific antibodies followed by mass spectrometry
Proximity labeling approaches (BioID, APEX) with CCT4 as the bait protein
Differential analysis of protein aggregation following CCT4 depletion
CCT4 loss-of-function studies:
siRNA knockdown: Transient reduction of CCT4 (70-90%)
CRISPR-Cas9 knockout: Complete elimination (consider inducible systems as complete loss may be lethal)
Dominant-negative mutants: Express ATPase-deficient CCT4 to disrupt function
Real-time folding visualization:
FRET-based folding sensors to monitor conformational changes in client proteins
Pulse-chase experiments to track nascent protein folding
Live-cell imaging with fluorescently tagged client proteins
Structure-function studies:
Site-directed mutagenesis of key CCT4 residues
Cryo-EM analysis of TRiC complex with client proteins in different states
Crosslinking mass spectrometry to map interaction interfaces
When designing these experiments, consider that CCT4 functions as part of the larger TRiC complex, so isolated studies of CCT4 may not fully recapitulate its physiological function. Include complementation experiments (rescuing knockdown/knockout with wild-type vs. mutant CCT4) to establish specific functional roles .
The emerging role of CCT4 in cancer progression makes it an attractive therapeutic target. Antibodies serve multiple functions in this research area:
Target validation approaches:
Immunohistochemical profiling across cancer types to identify high-expression tumors
Correlation of expression with clinical outcomes to establish prognostic significance
CCT4 knockdown/knockout studies to confirm dependency in cancer models
Drug development support:
Screening assays: Use CCT4 antibodies in ELISA or AlphaScreen formats to identify small molecule binders
Target engagement: Cellular thermal shift assays (CETSA) with CCT4 antibodies to confirm compound binding
Mechanism of action studies: Evaluate effects on CCT4 protein interactions using co-IP with CCT4 antibodies
Antibody-based therapeutics:
Intrabodies: Engineered antibody fragments expressed intracellularly to inhibit CCT4 function
Antibody-drug conjugates: For cancers with cell-surface expression or during internalization
CAR-T targeting: If tumor-specific expression patterns can be established
Pathway modulation assessment:
Monitor PI3K/AKT pathway components following CCT4 modulation
Evaluate effects on client protein folding and function
Assess combination therapy approaches targeting CCT4 and related pathways
Predictive biomarker development:
Develop standardized IHC protocols to stratify patients for clinical trials
Create companion diagnostic assays using validated CCT4 antibodies
Monitor treatment response through serial sampling (liquid biopsy approaches)
Research indicates that CCT genetic alterations affect the PI3K/AKT pathway, suggesting potential synergy between CCT4-targeted therapies and existing PI3K/AKT inhibitors. The significant association of CCT4 upregulation with poor prognosis in HCC provides strong rationale for therapeutic development in this indication .
When developing therapeutics targeting CCT4, consider its essential role in normal cell physiology and establish a therapeutic window that allows targeting of cancer cells while minimizing toxicity to normal tissues.
Investigating CCT4's role in specific processes like ciliogenesis requires specialized experimental approaches:
Ciliary structure and formation analysis:
Primary cilium induction: Serum starvation (24-48 hours) in appropriate cell lines (RPE-1, IMCD3)
CCT4 manipulation: siRNA knockdown, CRISPR knockout, or overexpression
Immunofluorescence microscopy:
Use antibodies against ciliary markers (acetylated tubulin, Arl13b) with CCT4 co-staining
Quantify cilia number, length, and morphology
Image acquisition with high-resolution confocal or super-resolution microscopy
BBSome assembly and function:
Co-immunoprecipitation: Use CCT4 antibodies to pull down complex and probe for BBSome components
Sucrose gradient fractionation: Analyze BBSome assembly intermediates with/without CCT4
FRAP (Fluorescence Recovery After Photobleaching): Study dynamic transport of BBSome components in the presence/absence of CCT4
Rescue experiments: Express wild-type vs. mutant CCT4 in depleted cells
Live imaging approaches:
Fluorescently-tagged CCT4: Monitor localization during ciliogenesis
Time-lapse microscopy: Track ciliary growth kinetics following CCT4 perturbation
Photoactivatable or photoswitchable tags: Monitor protein movement and turnover
Biochemical interaction studies:
In vitro binding assays with purified components
Proximity labeling (BioID, APEX) with CCT4 as bait in ciliated cells
Crosslinking mass spectrometry to identify interaction interfaces
Functional readouts:
Hedgehog signaling assays: Gli1 transcriptional activity using luciferase reporters
Ciliary protein trafficking: Monitor Smoothened localization to cilia following Hedgehog pathway activation
Cell migration assays: Wound healing or transwell migration to assess functional consequences
When designing these experiments, consider the timeframe of ciliogenesis (typically 24-48 hours post-serum starvation) and ensure CCT4 manipulation is maintained throughout this period. For transient knockdown approaches, confirm protein depletion at the endpoint of your experiment, as protein turnover rates may affect interpretation of results .
While CCT4's role in cancer has been extensively studied, emerging research suggests broader implications in various disease processes:
Neurodegenerative disorders:
Protein misfolding is central to neurodegenerative diseases
CCT4 may influence α-synuclein and tau protein folding
Brain tissues show high CCT4 expression, suggesting neuronal importance
Research methods:
Animal models expressing disease-associated protein aggregates
CCT4 modulation in neuronal cell cultures
Co-localization studies with disease-specific protein aggregates
Cardiovascular disease:
Proteostasis disruption contributes to cardiomyopathies
CCT4's role in cytoskeletal protein folding may affect cardiac function
Experimental approaches:
Tissue-specific CCT4 knockout in cardiac models
Pressure overload studies with CCT4 modulation
Analysis of CCT4 expression in heart failure samples
Infectious diseases:
Some pathogens may hijack host chaperone systems
CCT4 could be involved in viral protein folding or assembly
Investigation methods:
Infection models with CCT4 knockdown
Interaction studies between viral proteins and CCT4
Small molecule modulators of CCT4 as potential antivirals
Metabolic disorders:
Protein quality control plays a role in obesity and diabetes
CCT4 may influence insulin signaling pathway components
Approaches:
Analysis in metabolic tissues (liver, adipose, pancreas)
Diet-induced obesity models with CCT4 modulation
Glucose tolerance testing following CCT4 manipulation
Future research should focus on tissue-specific functions of CCT4 and developing conditional knockout models to address potential embryonic lethality of complete CCT4 loss. The development of specific small molecule modulators of CCT4 function would provide valuable tools for investigating its role in these various disease contexts.
Recent technological advances are significantly enhancing CCT4 antibody quality, specificity, and application range:
Recombinant antibody technology:
Single B cell cloning from immunized animals
Phage display libraries for epitope-specific selection
Advantages:
Reduced batch-to-batch variability
Renewable source without animal immunization
Potential for engineering enhanced properties
Nanobody and single-domain antibody platforms:
Smaller antigen-binding fragments with improved tissue penetration
Applications:
Super-resolution microscopy with reduced linkage error
Intracellular expression as functional inhibitors ("intrabodies")
Improved accessibility to conformational epitopes
Multiplexed antibody validation:
CRISPR knockout cell lines as gold-standard controls
Automated high-throughput validation pipelines
Orthogonal validation across multiple applications
Standardized reporting of validation data
Advanced imaging applications:
Expansion microscopy compatible antibodies
DNA-PAINT and other super-resolution techniques
Clearing-compatible antibodies for 3D tissue imaging
Applications to visualize CCT4 in cellular nanocompartments
Antibody conjugates and modifications:
Site-specific conjugation strategies
Novel fluorophores with improved properties
Bifunctional antibodies for proximity detection
Mass cytometry (CyTOF) compatible metal conjugates
These technological advances enable more precise investigation of CCT4's subcellular localization, interaction partners, and functional roles across different physiological and pathological contexts. Researchers should stay informed about validation standards and emerging technologies to select the most appropriate antibodies for their specific applications.
Systems biology offers powerful frameworks to contextualize CCT4 within broader cellular networks:
Multi-omics data integration:
Combine antibody-based proteomics with:
Transcriptomics (RNA-seq) to correlate mRNA and protein levels
Phosphoproteomics to map post-translational modifications
Interactomics (IP-MS) to define protein-protein interactions
Integration methods:
Network analysis tools (Cytoscape, STRING)
Correlation analysis across data types
Machine learning approaches for pattern recognition
Pathway analysis and modeling:
Map CCT4 interactions to known signaling pathways
Identify enriched pathways following CCT4 perturbation
Create mathematical models of chaperone networks
Simulate effects of CCT4 modulation on proteostasis
Single-cell multi-parameter analysis:
Single-cell proteomics with CCT4 antibodies
Mass cytometry (CyTOF) for high-dimensional analysis
Multiplexed imaging (CODEX, MIBI) for spatial context
Reveals cell-to-cell variability in CCT4 expression and function
Perturbation biology approaches:
CRISPR screens combined with CCT4 antibody readouts
Small molecule library screening
Genetic interaction mapping (synthetic lethality)
Identifies context-dependent functions and vulnerabilities
Clinical data correlation:
Integrate CCT4 expression data with:
Patient outcomes and survival
Treatment responses
Genetic backgrounds
Build predictive models for personalized medicine approaches
These approaches can reveal how CCT4 functions within the broader chaperone network and proteostasis system. For example, research has already identified connections between CCT4 genetic alterations and the PI3K/AKT pathway in HCC, demonstrating how systemic approaches can uncover unexpected functional relationships .
When implementing these approaches, standardization of antibody-based detection methods is crucial for generating comparable datasets across different studies and experimental platforms.