CAPG Antibody is a research tool designed to detect and quantify the expression of the capping actin protein, gelsolin-like (CAPG), an actin-binding protein critical for regulating cytoskeletal dynamics. CAPG stabilizes actin filaments by capping their barbed ends, influencing cell motility, immune cell function, and tumor progression . Antibodies targeting CAPG are employed in immunoblotting (Western blot), immunohistochemistry (IHC), immunocytochemistry (ICC), and enzyme-linked immunosorbent assays (ELISA) to study its role in physiological and pathological processes .
CAPG antibodies are utilized across diverse experimental workflows:
CAPG antibodies revealed that CAPG interacts with NF-κB transcription factors, promoting leukemic cell proliferation and survival. Knockdown of CAPG reduced AML progression in murine models, suggesting its role as an oncogene .
Immunohistochemistry with CAPG antibodies demonstrated upregulated CAPG expression in early gastric cancer (EGC), correlating with lymph node metastasis and poor prognosis. CAPG enhances migration and invasion via the Wnt/β-catenin pathway .
CAPG expression is linked to an immunosuppressive tumor microenvironment, promoting T-cell exhaustion. High CAPG levels predict shorter survival and correlate with reduced response to immunotherapy .
CAPG antibodies identified CAPG’s role in viral egress, particularly through interactions with the nucleocapsid protein VP40. CAPG inhibition disrupts actin remodeling, impairing virus release .
CAPG antibodies enable detection of elevated CAPG in early-stage cancers, such as EGC, offering diagnostic utility. For example, in gastric cancer, CAPG overexpression is observed in 94.7% of EGC cases compared to 60% in advanced gastric cancer (AGC) .
Nanobodies targeting CAPG’s actin-binding domains inhibit metastasis in breast cancer models by disrupting cytoskeletal dynamics. This approach may complement existing therapies targeting immune evasion or signaling pathways .
Precision Medicine: Integrating CAPG antibody-based diagnostics with genetic profiling to predict therapeutic responses.
Combinatorial Strategies: Pairing CAPG inhibitors with ferroptosis-inducing agents, as CAPG depletion enhances iron-dependent cell death in cancers .
Viral Pathogenesis: Exploring CAPG’s role in other viral infections beyond Ebola, using antibody-based assays to map host-virus interactions .
CAPG belongs to the gelsolin/villin family of actin-regulatory proteins. It plays a crucial role in controlling actin-based motility in non-muscle cells by capping the barbed ends of actin filaments without severing preformed filaments . This regulatory function makes it significant in studying cytoskeletal dynamics, cell migration, and related cellular processes. CAPG functions in a calcium and phosphoinositide-dependent manner, contributing to its biological significance in actin filament organization .
CAPG antibodies are primarily used for Western Blotting (WB), Immunohistochemistry (IHC), Immunofluorescence (IF), Immunocytochemistry (ICC), and Enzyme-Linked Immunosorbent Assays (ELISA) . These techniques allow researchers to detect, visualize, and quantify CAPG in various experimental contexts. The selection of a specific application depends on the research question, with Western blotting being particularly useful for quantitative analysis of protein expression levels and immunofluorescence providing insights into subcellular localization .
Most commercially available CAPG antibodies demonstrate reactivity with human samples, with many also cross-reacting with mouse and rat CAPG . Some antibodies have predicted reactivity with other species such as pig, bovine, horse, sheep, rabbit, and dog, though these predictions require experimental validation . When selecting an antibody, it's critical to verify species reactivity for your specific model organism to ensure reliable results and avoid false negatives due to species incompatibility.
Validation should include:
Positive and negative control testing using samples with known CAPG expression profiles
Comparison with previously validated antibodies through parallel staining
Assessment of specificity through Western blotting to confirm the detection of a single band at the expected molecular weight (approximately 38-39 kDa)
Evaluation of reproducibility across multiple experiments
Confirmation of expected staining patterns in relevant cell types (CAPG is abundant in macrophages and macrophage-like cells)
This systematic validation ensures reliable and reproducible results in subsequent experiments.
For optimal Western blot results with CAPG antibodies:
Use standard SDS-PAGE with 10-12% acrylamide gels to properly separate proteins in the 38-39 kDa range
Transfer proteins to PVDF or nitrocellulose membranes
Block membranes with 5% non-fat milk or BSA in TBST
Dilute primary CAPG antibodies according to manufacturer recommendations (typically 1:500 - 1:2000)
Incubate with appropriate HRP-conjugated secondary antibodies
Develop using enhanced chemiluminescence detection systems
Expected result: a single band at approximately 38-39 kDa representing CAPG protein
Include appropriate positive controls (cells with known CAPG expression) and negative controls (CAPG knockout cells or isotype control antibodies) to validate results.
For successful immunofluorescence detection of CAPG:
Fix cells with 4% paraformaldehyde for 15-20 minutes at room temperature
Permeabilize with 0.1-0.5% Triton X-100 for 5-10 minutes
Block with 1-5% BSA or normal serum in PBS for 30-60 minutes
Incubate with CAPG primary antibody at optimized dilution (typically starting at 1:100-1:500)
Wash thoroughly with PBS (3-5 times)
Incubate with fluorophore-conjugated secondary antibody
Counterstain nuclei with DAPI
Mount and image
CAPG typically shows cytoplasmic localization with potential concentration at membrane ruffles or areas of actin reorganization . Dual staining with actin markers can provide valuable insights into CAPG's relationship with the actin cytoskeleton.
When including CAPG antibodies in cocktails:
Verify compatibility among all antibody isotypes, fluorophores, and buffer requirements
Conduct optimization to determine optimal concentrations of each antibody in the cocktail
Validate the cocktail through comparison with individual antibody staining to ensure no interference occurs
Consider stability issues, especially with antibodies conjugated to tandem dyes which may break down more readily in cocktails
Document objective acceptance criteria for lot-to-lot validation (e.g., percentage of positive population within 10-15% difference, median fluorescence intensity within 0.5-1 log difference)
Store prepared cocktails in amber glass vials at 2-8°C and determine stability through rigorous testing
This systematic approach ensures reliable performance of antibody cocktails that include CAPG antibodies.
Problem | Possible Causes | Solutions |
---|---|---|
No signal | Insufficient antibody concentration | Increase antibody concentration or incubation time |
Inadequate antigen retrieval | Optimize antigen retrieval protocol | |
Improper sample preparation | Ensure proper fixation and permeabilization | |
High background | Excessive antibody concentration | Titrate antibody to optimal concentration |
Insufficient blocking | Increase blocking time or use alternative blocking reagent | |
Non-specific binding | Include additional washing steps | |
Multiple bands in Western blot | Protein degradation | Use fresh samples and protease inhibitors |
Non-specific binding | Increase stringency of wash buffers | |
Cross-reactivity | Try alternative CAPG antibody clones |
Always include appropriate controls and perform antibody validation to identify and address these common issues .
Establish comprehensive quality control by:
Implementing lot-to-lot testing for each new antibody batch
Performing parallel staining with previously validated antibodies on the same sample
Establishing objective acceptance criteria for validation:
Including both quantitative measurements and visual assessment of staining patterns
Maintaining detailed documentation of antibody lot numbers, preparation dates, and quality control results
Establishing standard operating procedures for regular antibody performance monitoring
These practices ensure consistent, reliable results across experiments and minimize variability introduced by antibody performance.
CAPG has been implicated in various cancer types, and antibody-based approaches can reveal:
Expression level changes in tumor vs. normal tissues using IHC and Western blotting
Subcellular localization alterations during cancer progression through IF/ICC
Association with invasion and metastasis by correlating CAPG expression with clinical outcomes
Interaction with other actin-regulatory proteins through co-immunoprecipitation and proximity ligation assays
Changes in CAPG phosphorylation status using phospho-specific antibodies
When designing such studies, researchers should:
Include appropriate cancer and normal control tissues
Correlate CAPG expression with established cancer markers
Consider CAPG's calcium-dependent regulation when analyzing results
Evaluate CAPG in the context of other gelsolin family members
For flow cytometric analysis with CAPG antibodies:
Determine whether surface or intracellular staining is required (CAPG is primarily intracellular)
For intracellular staining:
Optimize fixation and permeabilization conditions
Use appropriate permeabilization buffers compatible with cytoskeletal proteins
Ensure adequate washing to reduce background
When using in antibody cocktails:
Establish clear gating strategies based on positive and negative controls
Include proper compensation controls if using multiple fluorochromes
These considerations help ensure accurate detection and quantification of CAPG-positive populations.
To study CAPG's calcium-dependent functions:
Design experiments comparing CAPG localization and interactions under varying calcium concentrations
Use calcium chelators (EGTA) or ionophores (A23187) to manipulate calcium levels
Employ co-immunoprecipitation with CAPG antibodies under different calcium conditions to identify calcium-dependent binding partners
Consider using proximity ligation assays to detect in situ interactions between CAPG and actin or other binding partners
Compare results with other calcium-regulated actin-binding proteins (gelsolin family) as controls
Combine with site-directed mutagenesis of calcium-binding domains to correlate structure with function
This multi-faceted approach provides insights into how calcium regulates CAPG's actin-capping activity.
When faced with discrepant results:
Review antibody specifications thoroughly, noting the specific epitopes targeted by each clone
Consider whether post-translational modifications might affect epitope accessibility
Evaluate antibody validation data and supporting literature for each clone
Perform additional validation using genetic approaches (siRNA knockdown, CRISPR knockout)
Test multiple antibody clones side-by-side on the same samples
Consider the possibility of detecting different CAPG isoforms or splice variants
Consult published literature for consensus on CAPG expression patterns in your specific cell/tissue type
Combining multiple antibodies targeting different epitopes provides more robust verification of CAPG expression and localization.
For rigorous quantitative analysis:
Western blot densitometry:
Normalize CAPG signals to loading controls (β-actin, GAPDH)
Use standard curves with recombinant CAPG for absolute quantification
Employ statistical analysis across multiple biological replicates
Immunofluorescence quantification:
Measure integrated density or mean fluorescence intensity
Perform subcellular localization analysis through colocalization coefficients
Use automated image analysis software for unbiased quantification
Flow cytometry:
Report median fluorescence intensity ratios compared to isotype controls
Analyze percentage of positive cells using clearly defined gating strategies
Consider population heterogeneity in your analysis
Each approach has strengths and limitations, and combining multiple quantitative methods strengthens data reliability.
Emerging applications include:
Super-resolution microscopy to visualize CAPG's precise localization relative to actin filament barbed ends
Proximity labeling approaches (BioID, APEX) combined with CAPG antibodies for proteomic identification of the CAPG interactome
Antibody-based biosensors to monitor dynamic changes in CAPG conformation or activity in live cells
Integration with single-cell technologies to understand cellular heterogeneity in CAPG expression and function
Application in liquid biopsies as potential cancer biomarkers
Development of therapeutic approaches targeting CAPG in diseases with aberrant cytoskeletal regulation
These emerging applications represent the cutting edge of CAPG research and offer exciting opportunities for new discoveries.
Researchers can advance the field by:
Publishing detailed antibody validation protocols and results, including negative findings
Employing genetic controls (knockouts, knockdowns) alongside antibody-based detection
Contributing to antibody validation repositories and databases
Adopting the validation guidelines from ICCS/ESCCA consensus recommendations
Implementing multicenter validation studies to assess reproducibility across laboratories
Developing and sharing standardized positive control materials (cell lines, recombinant proteins)
Reporting detailed experimental conditions that affect antibody performance
Capping Protein Gelsolin-Like, also known as CapG, is a member of the gelsolin family of actin-modulating proteins. These proteins play a crucial role in various cellular processes, including cell signaling, receptor-mediated membrane ruffling, phagocytosis, and motility. CapG is particularly interesting due to its involvement in the regulation of actin filament dynamics, which is essential for cell movement and stability .
CapG has garnered significant attention for its role in oncogenesis, particularly in the context of cancer cell migration and invasion. Studies have shown that CapG is frequently upregulated in various types of cancer, including oral squamous cell carcinoma (OSCC) and non-small cell lung cancer (NSCLC). The overexpression of CapG in these cancers is associated with advanced disease stages and poor prognosis .
CapG functions by binding to the barbed ends of actin filaments, thereby preventing the addition or loss of actin monomers. This capping activity is crucial for maintaining the stability and organization of the actin cytoskeleton. The regulation of actin dynamics by CapG is essential for various cellular processes, including cell shape maintenance, motility, and division .
The expression levels of CapG have been studied extensively in the context of cancer. In OSCC, CapG expression is significantly higher in tumor tissues compared to normal tissues. This upregulation is correlated with the TNM stage grading of the cancer, suggesting that CapG could serve as a potential biomarker for cancer progression and prognosis . Similarly, in NSCLC, high levels of CapG expression are associated with increased tumor motility and multidrug resistance, further highlighting its potential as a therapeutic target .
The development of mouse anti-human CapG antibodies has facilitated the study of CapG expression in various tissues. These antibodies are used in immunohistochemistry (IHC) and real-time quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) to detect and quantify CapG expression at the protein and mRNA levels, respectively. These techniques have been instrumental in elucidating the role of CapG in cancer and other diseases .