CCT2 Antibody

Shipped with Ice Packs
In Stock

Description

Role in Cancer Biology

  • Tumor Growth: Silencing CCT2 in triple-negative breast cancer (TNBC) cells (MDA-MB-231) reduced viability, adhesion, and migration. In vivo studies showed CCT2 depletion impaired tumor growth in murine models .

  • Prognostic Marker: High CCT2 expression correlates with poor survival in breast cancer patients and drives tumor cell proliferation via JAK2/STAT3 signaling .

  • Immune Evasion: CCT2 suppresses CD40L expression in CD4+ T cells by inhibiting Ca²⁺-NFAT1 signaling, promoting immune escape in breast cancer .

Aggrephagy Regulation

  • Autophagy Receptor: CCT2 transitions from a chaperone to a ubiquitination-independent autophagy receptor under proteostatic stress, targeting solid protein aggregates for degradation .

  • Conserved Mechanism: Yeast studies confirm CCT2’s aggrephagy role is independent of the TRiC complex. Overexpressed Cct2 facilitates vacuolar localization of misfolded proteins (e.g., Ape1-P22L) via interactions with Atg8 and Atg11 .

Clinical and Therapeutic Implications

Study FocusKey FindingSource
Cancer PrognosisCCT2 expression correlates with TMB, MSI, and immune neoantigens in 10+ cancers
Therapeutic TargetCCT2 inhibition disrupts tumor growth and metastasis in breast cancer models
NeurodegenerationCCT2’s aggrephagy role highlights potential in treating proteinopathies (e.g., Alzheimer’s)

Validation and Protocols

  • Western Blot: Recommended dilutions range from 1:5,000 to 1:50,000 .

  • Immunofluorescence: Optimal staining achieved at 1:200–1:800 dilutions .

  • Controls: Validated in cell lines (HeLa, HEK-293, LNCaP) and tissues (brain, tumor xenografts) .

Future Directions

  • Mechanistic Studies: Elucidate CCT2’s role in disassembling mitotic checkpoint complexes and calcium signaling .

  • Therapeutic Development: Explore CCT2-targeted inhibitors for cancer and neurodegenerative diseases .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CCT2 antibody; At4g15130 antibody; dl3610w antibody; FCAALL.209 antibody; Choline-phosphate cytidylyltransferase 2 antibody; AtCCT2 antibody; EC 2.7.7.15 antibody; CTP:phosphocholine cytidylyltransferase 2 antibody; Phosphorylcholine transferase 2 antibody
Target Names
CCT2
Uniprot No.

Target Background

Function
CCT2 Antibody plays a crucial role in the biosynthesis of the phospholipid phosphatidylcholine. It catalyzes the formation of CDP-choline.
Database Links

KEGG: ath:AT4G15130

STRING: 3702.AT4G15130.1

UniGene: At.33210

Protein Families
Cytidylyltransferase family

Q&A

What is CCT2 and why is it important in cellular function?

CCT2 (Chaperonin Containing TCP1, Subunit 2 Beta) is a critical component of the CCT/TRiC chaperonin complex responsible for protein folding in eukaryotic cells. This complex plays an essential role in folding approximately 10% of newly synthesized cytosolic proteins, with actin and tubulin being among its obligate client proteins . CCT2 serves as one of eight subunits (CCT1-8) that form the barrel-shaped CCT complex, which uses ATP to fold proteins into their native conformations. Research has demonstrated that CCT2's function extends beyond structural support of the complex, as it directly influences other CCT subunits' expression levels and contributes to cellular processes including migration, proliferation, and invasiveness . The protein appears particularly relevant in cancer biology, where its expression is frequently upregulated and correlates with poor patient survival in several cancer types, notably breast cancer .

How should researchers select the appropriate CCT2 antibody for their specific applications?

Selection of the appropriate CCT2 antibody should be guided by the intended application, target species, and specific epitope requirements. When choosing between available antibodies, researchers should consider:

  • Application compatibility: Verify the antibody has been validated for your specific application (WB, IHC, ICC, FACS, etc.). For example, ABIN969010 is validated for Western Blotting, ELISA, Immunocytochemistry, and Flow Cytometry , while ABIN4886512 is validated specifically for Western Blotting and Immunohistochemistry on paraffin-embedded sections .

  • Species reactivity: Match the antibody to your experimental model organism. Some CCT2 antibodies are human-specific (like ABIN969010) , while others demonstrate cross-reactivity with mouse and rat samples (like ABIN4886512) .

  • Epitope specificity: Consider which region of CCT2 you need to target. Different antibodies recognize distinct amino acid sequences, such as AA 414-535 or other regions, which may affect detection of protein interactions or post-translational modifications.

  • Clonality: Monoclonal antibodies offer high specificity for a single epitope with minimal batch-to-batch variation (e.g., clone 5B5C4) , while polyclonal antibodies provide broader epitope recognition but potentially more background signal .

  • Validation data: Review performance data in applications similar to your planned experiments, preferably with positive controls from relevant tissues or cell lines.

What are the standard storage and handling procedures to maintain CCT2 antibody efficacy?

To maintain optimal performance of CCT2 antibodies, researchers should implement the following storage and handling protocols:

  • Temperature management: Store antibodies at -20°C for long-term storage, with aliquoting to prevent freeze-thaw cycles that can degrade antibody performance. For working solutions, maintain at 4°C for short periods (typically 1-2 weeks).

  • Aliquoting strategy: Upon receipt, divide the antibody into small single-use aliquots (10-20 μl) to minimize freeze-thaw cycles. Each freeze-thaw cycle can reduce antibody activity by approximately 10-15%.

  • Buffer compatibility: When diluting antibodies, use buffers appropriate for the application (e.g., TBS or PBS with 0.05-0.1% BSA for Western blotting) that maintain protein stability and reduce non-specific binding.

  • Contamination prevention: Use sterile techniques when handling antibodies to prevent microbial contamination, which can degrade antibody performance and introduce experimental artifacts.

  • Carrier protein consideration: For highly diluted antibody solutions, consider adding carrier proteins (0.1-1% BSA or gelatin) to prevent antibody adsorption to tube walls.

  • Expiration monitoring: Track antibody age and performance, as efficacy typically declines over time even with optimal storage conditions. When signal quality diminishes, validation of a new antibody lot is recommended.

What are the optimal protocols for using CCT2 antibodies in Western blotting?

For optimal Western blotting with CCT2 antibodies, researchers should follow this methodological approach:

  • Sample preparation:

    • Extract proteins using RIPA buffer supplemented with protease inhibitors

    • Quantify protein concentration using Bradford or BCA assay

    • Prepare 20-30 μg of total protein per lane for cell lines or 40-50 μg for tissue samples

    • Denature samples in Laemmli buffer at 95°C for 5 minutes

  • Gel electrophoresis:

    • Use 10-12% SDS-PAGE gels for optimal resolution of CCT2 (~57 kDa)

    • Include molecular weight markers to verify target band size

    • Run at 100-120V until sufficient separation is achieved

  • Membrane transfer:

    • Transfer proteins to PVDF or nitrocellulose membrane at 100V for 1 hour (or 30V overnight at 4°C)

    • Verify transfer efficiency with reversible staining (Ponceau S)

  • Blocking and primary antibody:

    • Block membrane with 5% non-fat milk or BSA in TBST for 1 hour at room temperature

    • Dilute CCT2 antibody according to manufacturer's recommendation (typically 1:500-1:2000)

    • Incubate membrane with primary antibody solution overnight at 4°C with gentle agitation

  • Detection and visualization:

    • Wash membrane 3-4 times with TBST, 5-10 minutes each

    • Incubate with appropriate HRP-conjugated secondary antibody (1:2000-1:5000) for 1 hour at room temperature

    • Wash 3-4 times with TBST

    • Develop using ECL substrate and capture images using appropriate detection system

  • Controls and normalization:

    • Include positive control samples known to express CCT2

    • Normalize signals to established loading controls (β-actin, GAPDH) or total protein staining

    • For tagged CCT2 constructs (like CCT2-FLAG), remember that the tagged protein will appear at a slightly higher molecular weight than endogenous CCT2, allowing differentiation

For quantification, normalize CCT2 signal to total protein rather than single housekeeping proteins for more accurate comparisons, as demonstrated in research examining CCT2 overexpression effects .

How can immunoprecipitation be used to study CCT2 interactions with other chaperonin subunits?

Immunoprecipitation (IP) represents a powerful approach to investigate CCT2's associations with other chaperonin subunits and potential client proteins:

  • Experimental design considerations:

    • Select antibodies with minimal interference to protein-protein interaction sites

    • For tagged CCT2 constructs (e.g., CCT2-FLAG), anti-tag antibodies provide excellent specificity

    • Design experiments to capture both stable and transient interactions

  • Co-immunoprecipitation protocol:

    • Harvest cells in non-denaturing lysis buffer (50 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM EDTA, 1% Triton X-100) with protease/phosphatase inhibitors

    • Clear lysate by centrifugation (14,000 × g, 10 min, 4°C)

    • Pre-clear lysate with protein A/G beads to reduce non-specific binding

    • Incubate cleared lysate with CCT2 antibody (5 μg) overnight at 4°C with gentle rotation

    • Add protein A/G beads and incubate for 2-4 hours at 4°C

    • Wash beads 4-5 times with lysis buffer

    • Elute bound proteins with Laemmli buffer at 95°C for 5 minutes

    • Analyze by SDS-PAGE and Western blotting for CCT2 and potential interaction partners

  • Interaction verification:

    • Probing for known CCT subunit interactions, such as CCT5, which directly contacts CCT2 in the complex

    • Perform reciprocal IP using antibodies against suspected interaction partners

    • Include appropriate negative controls (isotype-matched IgG or unrelated antibody)

Research demonstrates that FLAG-tagged CCT2 can successfully co-immunoprecipitate other CCT subunits, confirming integration into the functional chaperonin complex. For example, pulldown experiments using anti-FLAG agarose beads successfully co-precipitated both CCT2 and CCT5, confirming their physical association within the complex .

What methodologies are recommended for evaluating CCT2 expression in tissue samples?

For accurate assessment of CCT2 expression in tissue samples, researchers should employ these methodological approaches:

  • Immunohistochemistry (IHC) protocol:

    • Prepare formalin-fixed paraffin-embedded (FFPE) tissue sections (4-5 μm)

    • Perform antigen retrieval using citrate buffer (pH 6.0) at 95-98°C for 20 minutes

    • Block endogenous peroxidase with 3% H₂O₂ for 10 minutes

    • Block non-specific binding with 5% normal serum for 1 hour

    • Incubate with CCT2 antibody (optimized dilution, typically 1:100-1:500) overnight at 4°C

    • Apply HRP-conjugated secondary antibody for 30-60 minutes at room temperature

    • Develop with DAB substrate and counterstain with hematoxylin

    • Evaluate staining pattern, intensity, and localization

  • Tissue microarray approach:

    • Utilize tissue microarrays for high-throughput screening across multiple samples

    • Include normal adjacent tissue controls for comparison with tumor samples

    • Implement digital pathology tools for quantitative assessment of staining

  • Multiplexed immunofluorescence:

    • For co-localization studies, employ multiplexed immunofluorescence with other CCT subunits

    • Use spectral unmixing to differentiate signals when detecting multiple targets

    • Perform confocal microscopy for high-resolution subcellular localization

  • Quantitative considerations:

    • Develop consistent scoring systems (e.g., H-score or Allred score) for semi-quantitative assessment

    • Utilize automated image analysis software for unbiased quantification

    • Correlate CCT2 expression with clinicopathological parameters and patient outcomes

Research has employed these approaches to demonstrate that CCT2 expression is elevated in breast, prostate, and lung cancers compared to normal tissues, and that its expression correlates with patient survival outcomes . When evaluating CCT2 depletion in tumor models, immunohistochemistry analysis confirmed varying levels of knockdown efficiency across different tumor samples, highlighting the importance of validating target modulation in intervention studies .

How can researchers use CCT2 antibodies to investigate its role in cancer progression?

CCT2 antibodies can be employed in several sophisticated approaches to elucidate its functions in cancer progression:

  • Expression correlation studies:

    • Utilize CCT2 antibodies for IHC on tissue microarrays containing samples across cancer stages

    • Quantify expression levels and correlate with clinicopathological parameters

    • Perform survival analysis comparing patients with high versus low CCT2 expression

    • Research has shown that patients with genomic alterations in CCT2 (mainly gene amplification or increased mRNA) died almost 70 months sooner than patients without changes in CCT2

  • Cellular phenotype assessment:

    • Combine CCT2 detection with cellular markers of proliferation (Ki-67, EdU incorporation)

    • Assess relationship between CCT2 expression and cell cycle regulators (CDK2, CDK4)

    • Evaluate correlation between CCT2 levels and markers of invasiveness

    • Studies demonstrate that CCT2 overexpression increases proliferation index and alters cell morphology toward more invasive phenotypes

  • Mechanistic investigations:

    • Use proximity ligation assays (PLA) with CCT2 antibodies to identify novel protein interactions in situ

    • Perform ChIP-seq experiments using transcription factor antibodies to identify regulators of CCT2 expression

    • Combine CCT2 antibodies with phospho-specific antibodies to assess signaling pathway activation

  • Therapeutic response monitoring:

    • Monitor CCT2 expression changes following treatment with cytotoxic or targeted therapies

    • Assess correlation between CCT2 levels and therapeutic resistance mechanisms

    • Evaluate CCT2 as a potential predictive biomarker for specific treatment modalities

Research has demonstrated that CCT2 influences the invasiveness and proliferation of breast cancer cells, with overexpression promoting migration in cells that were not inherently motile . CCT2 depletion in a triple-negative breast cancer model prevented tumor growth, indicating its essential role in tumorigenesis and potential as a therapeutic target .

What are the methodological approaches for investigating CCT2's impact on cytoskeletal organization?

To investigate CCT2's impact on cytoskeletal organization, researchers can implement these methodological approaches:

  • Immunofluorescence co-localization analysis:

    • Co-stain cells for CCT2 and cytoskeletal components (actin, tubulin)

    • Perform high-resolution confocal microscopy to assess spatial relationships

    • Quantify co-localization using Pearson's or Mander's coefficients

    • Evaluate cytoskeletal organization in CCT2-modulated cells using phalloidin (F-actin) and anti-tubulin antibodies

  • Live-cell imaging techniques:

    • Generate fluorescently-tagged CCT2 constructs (ensuring functionality is preserved)

    • Combine with fluorescent cytoskeletal markers (LifeAct, SiR-Actin, SiR-Tubulin)

    • Perform time-lapse imaging to track dynamic changes in cytoskeletal structures

    • Analyze cytoskeletal dynamics parameters (polymerization rates, persistence)

  • Biochemical fractionation:

    • Separate soluble and insoluble (cytoskeletal) fractions using differential detergent extraction

    • Quantify distribution of actin and tubulin between fractions in CCT2-modulated cells

    • Assess polymerization status of cytoskeletal proteins using Western blotting

  • Functional cytoskeletal assays:

    • Evaluate microtubule regrowth after nocodazole-induced depolymerization

    • Assess actin dynamics using FRAP (Fluorescence Recovery After Photobleaching)

    • Quantify cellular mechanical properties using atomic force microscopy

    • Measure focal adhesion dynamics in CCT2-modulated cells

Research has established that CCT is critical for cytoskeletal function, with actin and tubulin being obligate client proteins . Studies targeting CCT with CT20p resulted in loss of actin and tubulin, and CCT2 overexpression promoted cell migration, highlighting its role in cytoskeletal regulation . The function of CCT in supporting cytoskeletal integrity directly impacts cell motility, morphology, and proliferation, making it a crucial area for cancer research .

How can CCT2 antibodies be utilized to study the effects of CCT2 depletion or overexpression?

CCT2 antibodies serve as essential tools for monitoring and validating experimental manipulations of CCT2 expression:

  • Validation of genetic manipulation approaches:

    • Confirm knockdown efficiency in CCT2 siRNA/shRNA experiments via Western blotting

    • Verify overexpression levels in CCT2 transfection or transduction experiments

    • Quantify both exogenous (tagged) and endogenous CCT2 expression

    • Research has demonstrated that CCT2-FLAG constructs can be distinguished from endogenous CCT2 by slight molecular weight differences on immunoblots

  • Monitoring approaches for inducible systems:

    • Assess temporal dynamics of CCT2 depletion in tetracycline-inducible shRNA systems

    • Optimize doxycycline concentration and treatment duration for maximal knockdown

    • Correlate GFP reporter expression with CCT2 depletion in co-expression systems

    • Studies have used 72-hour doxycycline treatment to achieve optimal CCT2 knockdown in an inducible Tet-on shRNA system

  • Phenotypic consequence assessment:

    • Quantify effects on other CCT subunits following CCT2 manipulation

    • Assess changes in client protein folding and function

    • Measure proliferation, migration, and invasion in modified cells

    • Research shows that increasing total CCT2 by 1.3-1.8-fold caused concurrent increases in CCT3, CCT4, and CCT5 levels, while silencing CCT2 by ~50% caused other CCT subunits to decrease

  • In vivo validation:

    • Confirm target modulation in xenograft or syngeneic tumor models using IHC

    • Correlate CCT2 expression with tumor growth kinetics

    • Assess heterogeneity of knockdown efficiency across tumor samples

    • Studies revealed that tumors growing despite CCT2 shRNA induction maintained detectable CCT2 levels comparable to controls, indicating selection for cells with inefficient knockdown

Research has established that CCT2 depletion results in loss of viability, reduced adherence, and impaired migration in triple-negative breast cancer cells . CCT2 overexpression promotes invasiveness and increases proliferative index, demonstrating its integral role in the chaperonin complex's activity and contribution to tumorigenesis .

What are common challenges when using CCT2 antibodies and how can they be addressed?

Researchers frequently encounter several challenges when working with CCT2 antibodies that can be systematically addressed:

  • Non-specific binding and background signal:

    • Challenge: High background or multiple bands in Western blotting

    • Solution: Optimize blocking (try 5% BSA instead of milk), increase washing stringency, and titrate antibody concentration

    • Validation approach: Include negative control samples and peptide competition assays to confirm specificity

  • Variable immunostaining patterns:

    • Challenge: Inconsistent staining across tissue sections

    • Solution: Standardize fixation protocols, optimize antigen retrieval methods, and test multiple antibody clones

    • Optimization strategy: Perform systematic titration of antibody concentration and antigen retrieval conditions

  • Cross-reactivity concerns:

    • Challenge: Potential cross-reactivity with other CCT subunits due to structural similarities

    • Solution: Select antibodies specifically validated for minimal cross-reactivity (e.g., ABIN4886512 which shows "no cross reactivity with other proteins")

    • Verification approach: Test antibody in CCT2-knockdown cells as negative controls

  • Detecting conformational changes:

    • Challenge: Antibody may not recognize CCT2 in certain conformational states within the complex

    • Solution: Use multiple antibodies targeting different epitopes or consider non-denaturing conditions

    • Alternative strategy: Use tagged CCT2 constructs when studying complex formation

  • Quantification accuracy:

    • Challenge: Reliable quantification of expression differences

    • Solution: Normalize to total protein rather than single housekeeping proteins

    • Best practice: Use multiple technical replicates and standardized exposure times for imaging

Research studies have addressed these challenges through careful antibody selection and validation. For example, researchers studying CCT2 overexpression normalized signals to total protein rather than individual loading controls, improving quantification accuracy . Additionally, when using tagged constructs, researchers confirmed the slightly higher molecular weight band of CCT2-FLAG compared to endogenous CCT2 to verify expression of the exogenous protein .

How can researchers validate CCT2 antibody specificity for their experimental system?

Rigorous validation of CCT2 antibody specificity is essential for producing reliable and reproducible research findings:

  • Genetic validation approaches:

    • Utilize CCT2 knockdown/knockout systems as negative controls

    • Test antibody in CCT2-overexpressing cells as positive controls

    • Employ CRISPR-Cas9 edited cell lines with epitope modifications

    • Compare staining patterns across multiple antibodies targeting different CCT2 epitopes

  • Biochemical validation methods:

    • Perform peptide competition assays using the immunizing peptide

    • Conduct immunoprecipitation followed by mass spectrometry to confirm target identity

    • Use recombinant CCT2 protein as a positive control in Western blotting

    • Test cross-reactivity against other purified CCT subunits

  • Technical validation strategies:

    • Confirm expected molecular weight (~57 kDa for human CCT2)

    • Verify expression patterns match known biological distribution

    • Test antibody performance across multiple applications (WB, IHC, ICC)

    • Evaluate lot-to-lot consistency with standardized positive controls

  • Orthogonal validation:

    • Compare antibody-based detection with mRNA expression (qRT-PCR)

    • Correlate with tagged-protein expression in overexpression systems

    • Confirm subcellular localization matches known distribution patterns

Research has utilized multiple validation approaches when studying CCT2. For example, in overexpression studies, researchers confirmed that the CCT2-FLAG construct was incorporated into the CCT complex by performing pulldown experiments and detecting interacting partners like CCT5 . Additionally, when studying CCT2 depletion in vivo, researchers performed IHC analysis of tumor tissues to confirm the efficacy of knockdown, revealing that tumors that grew despite CCT2 shRNA induction had escaped knockdown .

What strategies can optimize CCT2 detection in challenging sample types?

Detecting CCT2 in challenging sample types requires optimization strategies tailored to specific technical limitations:

  • Fixed tissue samples:

    • Challenge: Antigen masking due to fixation and processing

    • Optimization: Test multiple antigen retrieval methods (heat-induced vs. enzymatic)

    • Protocol enhancement: Extended antigen retrieval (30-40 minutes) in citrate buffer (pH 6.0)

    • Signal amplification: Consider tyramide signal amplification for low-abundance detection

  • Archived/FFPE samples:

    • Challenge: Protein degradation and modification in long-stored samples

    • Approach: Select antibodies targeting preserved epitopes (often C-terminal regions)

    • Technical solution: Test multiple antibodies recognizing different regions (e.g., AA 414-535)

    • Control strategy: Include recently processed samples as positive controls

  • Primary patient samples:

    • Challenge: Limited material and heterogeneous expression

    • Methodology: Implement multiplex IHC to maximize information from minimal tissue

    • Analysis strategy: Use digital pathology tools for objective quantification

    • Validation: Correlate with patient-matched fresh frozen samples when possible

  • Low-expressing systems:

    • Challenge: Weak signal from samples with low CCT2 expression

    • Technical solution: Use high-sensitivity detection systems (Super Signal West Femto)

    • Protocol enhancement: Extend primary antibody incubation (overnight at 4°C)

    • Enrichment approach: Consider subcellular fractionation to concentrate target protein

Research has successfully detected CCT2 in challenging samples by implementing these optimization strategies. For example, when analyzing CCT2 expression in tumor tissues from mice with inducible CCT2 knockdown, researchers were able to detect varying levels of CCT2 despite the challenging nature of the samples, revealing that tumors growing despite knockdown induction had maintained CCT2 expression .

How can CCT2 antibodies be utilized in studying the relationship between CCT2 and cancer therapeutic resistance?

CCT2 antibodies can be strategically employed to investigate its potential role in therapeutic resistance mechanisms:

  • Expression correlation with resistance phenotypes:

    • Compare CCT2 levels between treatment-sensitive and resistant cell lines

    • Monitor expression changes during acquired resistance development

    • Correlate patient response to therapy with pre-treatment CCT2 expression

    • Analyze CCT2 levels in paired pre- and post-treatment samples

  • Mechanistic investigation approaches:

    • Assess CCT2-dependent protein folding of known resistance mediators

    • Evaluate CCT2 modulation's impact on therapeutic sensitivity

    • Investigate CCT2's influence on stress response pathways following treatment

    • Determine if CCT2 inhibition can resensitize resistant cells to standard therapies

  • Combination therapy assessment:

    • Use CCT2 antibodies to confirm target engagement of CCT-directed therapeutics

    • Monitor CCT2 expression/activity during combination treatment approaches

    • Develop IHC-based companion diagnostics for CCT2-targeted therapies

    • Evaluate changes in CCT2 complex formation following treatment interventions

  • Biomarker development strategy:

    • Develop standardized IHC protocols for CCT2 assessment in clinical samples

    • Establish threshold values correlating with treatment response

    • Implement multiplexed detection with other resistance markers

    • Validate prognostic/predictive value in retrospective patient cohorts

Research has established that targeting CCT with the peptide CT20p, which directly interacts with CCT2, resulted in loss of actin and tubulin, suggesting a potential therapeutic approach . Additionally, the finding that CCT2 depletion prevents tumor growth in vivo indicates that targeting this chaperonin subunit could overcome resistance mechanisms by interfering with fundamental tumor cell viability requirements .

What novel approaches can integrate CCT2 antibodies with advanced imaging technologies?

Integration of CCT2 antibodies with cutting-edge imaging technologies opens new avenues for investigating its cellular functions:

  • Super-resolution microscopy applications:

    • Apply STORM or PALM imaging to visualize CCT2 distribution at nanoscale resolution

    • Map CCT2's spatial relationship with client proteins and other chaperonin subunits

    • Quantify changes in CCT2 nanodomains following cellular perturbations

    • Technical approach: Use directly labeled primary antibodies to minimize localization error

  • Live-cell imaging strategies:

    • Employ CCT2 intrabodies for real-time tracking of endogenous CCT2

    • Utilize CRISPR-Knock-in approaches to tag endogenous CCT2 with fluorescent proteins

    • Combine with biosensors for ATP consumption or protein folding activity

    • Correlate CCT2 dynamics with cytoskeletal remodeling during migration

  • Tissue-level analytical methods:

    • Implement multiplexed ion beam imaging (MIBI) for simultaneous detection of multiple targets

    • Apply clearing techniques with whole-mount immunolabeling for 3D visualization

    • Utilize spatial transcriptomics with protein detection for multi-omic analysis

    • Correlate CCT2 distribution with tissue architecture and microenvironmental features

  • Multi-modal imaging approaches:

    • Combine functional imaging (FRET) with structural analysis (SRM)

    • Integrate live-cell imaging with subsequent immunofluorescence

    • Implement correlative light and electron microscopy (CLEM) for ultrastructural context

    • Develop computational image analysis pipelines for quantitative feature extraction

While not explicitly mentioned in the search results, these approaches represent logical extensions of current CCT2 research methodologies. The demonstrated importance of CCT2 in cytoskeletal organization and cancer progression makes it an ideal candidate for advanced imaging investigations that could reveal new mechanistic insights into its function .

How can researchers investigate the potential of CCT2 as a therapeutic target using antibody-based approaches?

Antibody-based approaches offer versatile strategies for evaluating CCT2 as a therapeutic target:

  • Target validation strategies:

    • Utilize antibodies to confirm CCT2 overexpression across cancer types and subtypes

    • Correlate expression with clinical outcomes to prioritize cancer indications

    • Evaluate CCT2 expression in treatment-resistant populations

    • Research has demonstrated CCT2 upregulation in breast, prostate, and lung cancers compared to normal tissues

  • Functional inhibition approaches:

    • Develop function-blocking antibodies targeting CCT2's interaction interfaces

    • Test intracellular antibody delivery systems (lipid nanoparticles, cell-penetrating peptides)

    • Evaluate antibody-induced CCT2 degradation approaches

    • Assess phenotypic consequences in comparison to genetic CCT2 depletion

  • Antibody-drug conjugate development:

    • Investigate internalization efficiency of CCT2 antibodies

    • Optimize linker-payload combinations for maximum efficacy

    • Assess target specificity across normal and malignant tissues

    • Evaluate potential resistance mechanisms to CCT2-targeted therapies

  • Imaging and theranostic applications:

    • Develop CCT2 antibody-based imaging agents for patient stratification

    • Implement dual-function theranostic approaches combining imaging and therapy

    • Utilize CCT2 antibodies for intraoperative guidance in surgical resection

    • Evaluate pharmacodynamic biomarkers of CCT2-targeted therapies

Research has established CCT2 as a compelling therapeutic target, demonstrating that CCT2 depletion in a syngeneic murine model of triple-negative breast cancer prevented tumor growth . Additionally, the observation that CCT2 influences other CCT subunits suggests that targeting this single subunit could disrupt the entire chaperonin complex, potentially offering a therapeutic strategy with broad implications for cancer treatment . The identification of CT20p as a peptide that directly interacts with CCT2 provides proof-of-concept for targeted therapeutic development .

What are the emerging applications of CCT2 antibodies in precision medicine?

CCT2 antibodies hold significant potential for advancing precision medicine approaches through several emerging applications:

  • Diagnostic and prognostic stratification:

    • Development of standardized IHC protocols for clinical assessment

    • Establishment of CCT2 expression as a prognostic biomarker

    • Integration into multi-biomarker panels for patient stratification

    • Research has demonstrated that genomic alterations in CCT2 correlate with significantly reduced survival (approximately 70 months shorter) in cancer patients

  • Therapeutic response prediction:

    • Implementation of companion diagnostic assays for CCT-targeted therapies

    • Correlation of baseline CCT2 expression with treatment outcomes

    • Monitoring of CCT2 levels during treatment to detect resistance development

    • Integration with other molecular features for response prediction algorithms

  • Circulating tumor cell analysis:

    • Utilization of CCT2 antibodies in CTC enrichment and characterization

    • Correlation of CCT2-positive CTCs with disease progression

    • Assessment of CCT2 expression heterogeneity in disseminated disease

    • Longitudinal monitoring during treatment for early resistance detection

  • Theranostic approach development:

    • Creation of dual-function antibodies for imaging and therapeutic delivery

    • Implementation of image-guided therapeutic strategies

    • Development of CCT2-targeted radioimmunoconjugates

    • Integration with emerging immune-based therapeutic approaches

The demonstrated essentiality of CCT2 for tumor growth in vivo establishes its potential as a therapeutic and diagnostic target . The correlation between CCT2 expression and cancer progression, combined with its influence on cell invasion and proliferation, positions CCT2 as a promising biomarker for precision medicine applications in oncology .

What knowledge gaps remain in CCT2 biology that antibody-based research could address?

Despite significant advances, several critical knowledge gaps in CCT2 biology could be addressed through innovative antibody-based research approaches:

  • Substrate selectivity mechanisms:

    • Development of proximity-based proteomics approaches using CCT2 antibodies

    • Investigation of client-specific interaction domains through epitope-specific antibodies

    • Characterization of CCT2's role in substrate recognition versus other subunits

    • Research notes the "lack of a full understanding of CCT substrate selectivity in vivo" as a significant challenge

  • Conformational dynamics during protein folding:

    • Creation of conformation-specific antibodies recognizing different CCT2 states

    • Implementation of FRET-based sensors to monitor conformational changes

    • Investigation of ATP-dependent structural rearrangements within the complex

    • Development of real-time assays for folding activity measurement

  • Tissue-specific functions and regulation:

    • Comprehensive tissue atlas of CCT2 expression and interactome across organ systems

    • Characterization of tissue-specific post-translational modifications

    • Investigation of context-dependent functions beyond cytoskeletal regulation

    • Assessment of microenvironmental influences on CCT2 activity

  • Therapeutic targeting optimization:

    • Identification of functionally critical epitopes for inhibitory antibody development

    • Investigation of potential synergistic targets for combination approaches

    • Characterization of resistance mechanisms to CCT2-targeted interventions

    • Determination of optimal patient populations for CCT2-directed therapies

How might technological advances in antibody engineering impact future CCT2 research?

Emerging innovations in antibody engineering are poised to revolutionize CCT2 research through several transformative approaches:

  • Intracellular antibody technologies:

    • Development of cell-penetrating antibodies for live-cell CCT2 targeting

    • Creation of nanobodies/single-domain antibodies for intracellular applications

    • Implementation of intrabodies for real-time tracking of endogenous CCT2

    • Engineering of destabilizing antibodies for targeted CCT2 degradation

  • Multi-specific antibody platforms:

    • Design of bispecific antibodies targeting CCT2 and complementary cancer markers

    • Development of antibodies simultaneously targeting multiple CCT subunits

    • Creation of immunotherapy approaches redirecting immune cells to CCT2-expressing tumors

    • Engineering of antibody-based molecular glues to disrupt critical CCT2 interactions

  • Antibody-based proximity labeling:

    • Implementation of TurboID or APEX2-conjugated antibodies for proximity proteomics

    • Development of split-enzyme complementation systems for interaction mapping

    • Creation of light-activatable crosslinking antibodies for transient interaction capture

    • Integration with mass spectrometry for comprehensive interactome characterization

  • Synthetic biology integration:

    • Engineering of antibody-based synthetic signaling circuits

    • Development of optogenetic or chemogenetic antibody-based control systems

    • Creation of logic-gated antibody functions for context-dependent activity

    • Implementation of closed-loop systems for dynamic CCT2 monitoring and targeting

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.