ACTA1 recombinant monoclonal antibodies are produced using recombinant DNA technology. Genes encoding the antibody’s variable regions are cloned into mammalian expression systems (e.g., HEK293 cells) to ensure batch-to-batch consistency and high specificity . For example:
CAB2319 (AssayGenie): Derived from rabbits immunized with a synthetic peptide (amino acids 1–100 of human ACTA1) .
MA5-42772 (Thermo Fisher): Generated using an immunogen sequence spanning residues in the central region of ACTA1 .
ACTA1 mutations are linked to nemaline myopathy, congenital fiber-type disproportion, and core myopathy . These antibodies facilitate:
Muscle Contraction: Visualizing actin-myosin interactions in skeletal muscle .
Cytoskeletal Dynamics: Tracking ACTA1 in cell motility assays .
Positive Controls: HeLa, A-431, mouse heart, and rat lung lysates .
Specificity: Verified using knockdown models (e.g., siRNA-treated HeLa cells) .
Cross-Reactivity: Some clones (e.g., Sigma-Aldrich’s C4) recognize all actin isoforms, while others are ACTA1-specific .
Recent innovations include epigenetic modulation to enhance yield:
HDAC/LSD1 inhibitors (e.g., I-4) increase histone acetylation and antibody titers by 1.64-fold in CHO cells .
Recombinant vs. Traditional: Recombinant antibodies exhibit superior specificity and reduced background in immunohistochemistry .
CUSABIO developed a recombinant monoclonal antibody against ACTA1 by immunizing a rabbit with a synthesized peptide derived from human ACTA1. Subsequently, B cells were isolated from the immunized rabbit, and RNA was extracted. This RNA was reverse-transcribed into cDNA, which served as a template for extending ACTA1 antibody genes using degenerate primers. These extended ACTA1 antibody genes were then integrated into a plasmid vector and introduced into host cells for expression. The ACTA1 recombinant monoclonal antibody was purified from the cell culture supernatant via affinity chromatography. The antibody was subsequently evaluated for its suitability in ELISA, IHC, and FC applications. The antibody demonstrates specific recognition of the human ACTA1 protein.
ACTA1 is a key component of the thin filaments in skeletal muscle fibers, where it interacts with myosin to generate the contractile force necessary for muscle contraction. Mutations in the ACTA1 gene can lead to various muscle disorders, including congenital myopathies.
Recombinant monoclonal antibodies offer significant advantages over conventional monoclonal antibodies for ACTA1 detection. Unlike traditional antibodies produced through hybridoma technology, recombinant antibodies are generated through molecular cloning of antibody genes and expression in mammalian cell systems. This process involves immunizing an animal (typically rabbit) with a synthesized peptide derived from human ACTA1, isolating B cells, identifying single antibody-producing clones, sequencing the antibody, synthesizing its gene, and expressing it in mammalian cells .
The key advantages include:
Higher reproducibility: Recombinant technology eliminates batch-to-batch variation common in hybridoma-produced antibodies
Increased specificity: They typically show higher target specificity and reduced background staining
Better consistency: Uniform antibody production leads to more reliable research results
Enhanced sensitivity: Often detect lower levels of target protein
Broader application range: Generally work effectively across multiple experimental platforms
This makes recombinant antibodies particularly valuable for longitudinal studies where consistency between experiments is critical .
ACTA1 recombinant monoclonal antibodies are versatile research tools with multiple validated applications:
Application | Typical Dilution Range | Key Considerations |
---|---|---|
Western Blot (WB) | 1:500 - 1:5000 | Detects ~42 kDa band; sensitive to loading controls |
Immunohistochemistry (IHC) | 1:50 - 1:500 | Works on both frozen and paraffin sections; epitope retrieval critical |
Immunofluorescence (IF) | 1:30 - 1:200 | Co-staining with cytoskeletal markers recommended |
Flow Cytometry (FCM) | Vendor-specific | Cell permeabilization required for intracellular target |
ELISA | Assay-dependent | Validated for research-grade detection systems |
These antibodies have been validated on diverse sample types, including human, mouse, and rat tissues, with positive samples including HeLa, A-431, C6, mouse lung, mouse brain, mouse heart, rat lung, and rat heart . They are particularly valuable for studying muscle development, characterizing myopathies, and investigating cytoskeletal structures in muscle tissues .
Validating antibody specificity is crucial for reliable ACTA1 research. A comprehensive validation approach should include:
Positive and negative control selection:
Positive controls: Skeletal muscle tissue (highest ACTA1 expression), cardiac muscle (lower but detectable expression)
Negative controls: Non-muscle tissues with minimal ACTA1 expression (e.g., liver)
Multi-technique validation protocol:
Genetic validation approaches:
ACTA1 knockdown/knockout samples: Reduced/absent signal confirms specificity
Overexpression systems: Enhanced signal in cells transfected with ACTA1
Cross-reactivity assessment:
Test against other actin isoforms (ACTB, ACTC1, ACTG1) to ensure isoform specificity
Evaluate across species if conducting comparative studies
This systematic approach ensures that observed signals genuinely represent ACTA1 rather than non-specific binding or cross-reactivity with related proteins .
When studying ACTA1 mutations in myopathies, several methodological considerations are essential:
Epitope mapping relative to mutation sites:
Expression level variability:
Mutant ACTA1 may have altered expression levels compared to wild-type
Use appropriate loading controls and quantification methods
Consider normalizing to total protein rather than housekeeping genes
Protein conformation effects:
Subcellular localization changes:
Some ACTA1 mutations alter protein localization
Perform subcellular fractionation or co-localization studies
Combine with confocal microscopy for spatial resolution
Functional assays correlation:
These considerations are particularly important when studying the 447 pathogenic/likely pathogenic ACTA1 variants associated with conditions like nemaline myopathy (NEM) and cardiomyopathy .
For optimal ACTA1 immunohistochemistry in muscle biopsies, follow this detailed methodology:
Sample preparation:
For FFPE sections: 4-5 μm thickness is optimal
For frozen sections: 8-10 μm thickness on positively charged slides
Critical fixation parameters: 10% neutral buffered formalin for 24 hours maximum
Antigen retrieval optimization:
Heat-induced epitope retrieval: Citrate buffer (pH 6.0) at 95-98°C for 20 minutes
Allow slides to cool gradually in buffer (15-20 minutes)
For frozen sections: Brief fixation in cold acetone (10 minutes)
Blocking and antibody incubation:
Detection and counterstaining:
HRP-polymer detection systems minimize background
DAB development: Monitor microscopically to prevent overdevelopment
Hematoxylin counterstain: 30-60 seconds for optimal nuclear visualization
Controls and validation:
Include positive control (normal skeletal muscle)
Include isotype control at same concentration as primary antibody
Consider dual staining with other muscle markers for colocalization studies
This protocol has been successfully utilized across multiple studies examining both normal and pathological muscle samples, with consistent cytoskeletal localization of ACTA1 observed in normal samples .
Optimizing Western blot protocols for ACTA1 detection across different tissue types requires addressing tissue-specific challenges:
Tissue-specific extraction optimization:
Tissue Type | Lysis Buffer Recommendation | Special Considerations |
---|---|---|
Skeletal Muscle | RIPA with protease inhibitors and 1mM PMSF | Mechanical homogenization critical |
Cardiac Tissue | Modified RIPA (higher detergent) | Requires longer extraction time |
Cell Lines | Standard RIPA or NP-40 buffer | Gentle cell scraping preferred |
Brain Tissue | Specialized neuronal extraction buffer | Avoid excessive mechanical disruption |
Protein loading and separation parameters:
Load 20-40 μg total protein (higher for low-expressing tissues)
10-12% polyacrylamide gels provide optimal separation
Extended run times (>1 hour) improve band resolution
Transfer optimization:
Semi-dry transfer: 15V for 30-45 minutes
Wet transfer: 30V overnight at 4°C for complete transfer
PVDF membranes preferred over nitrocellulose for signal strength
Antibody incubation conditions:
Detection considerations:
Enhanced chemiluminescence with extended exposure for low-expressing tissues
Stripping and reprobing for multiple targets requires careful optimization
Quantify relative to appropriate loading controls (GAPDH generally suitable)
This approach has successfully detected the expected 42 kDa ACTA1 band across diverse tissue types, with highest expression in skeletal muscle samples .
When working with ACTA1 antibodies, several common issues may arise. Here are methodological solutions for each:
High background signal:
Root causes: Insufficient blocking, excessive primary antibody, cross-reactivity
Solutions:
Increase blocking time (2 hours minimum) with 5% BSA or normal serum
Optimize antibody dilution with titration series (1:100, 1:500, 1:1000, etc.)
Include 0.1-0.3% Triton X-100 in washing steps
Use monovalent Fab fragments to block endogenous IgG
Weak or absent signal:
Root causes: Protein degradation, insufficient antigen retrieval, epitope masking
Solutions:
Verify protein integrity with Ponceau S staining
Extend antigen retrieval time or try alternative methods (citrate vs. EDTA)
Test multiple antibody concentrations and incubation times
Use fresh tissue samples with minimal freeze-thaw cycles
Non-specific bands in Western blot:
Root causes: Protein degradation, splice variants, cross-reactivity
Solutions:
Add additional protease inhibitors during extraction
Increase antibody dilution and washing stringency
Run gradient gels for better resolution
Verify with knockout/knockdown controls
Inconsistent staining patterns:
Root causes: Fixation variability, tissue penetration issues, antibody degradation
Solutions:
Poor reproducibility between experiments:
These targeted approaches address specific methodological challenges rather than simply recommending general protocol adjustments.
Proper storage and handling of ACTA1 antibodies is critical for maintaining their activity and ensuring experimental reproducibility. The following methodology preserves antibody functionality:
Long-term storage conditions:
Working stock preparation:
Freeze-thaw cycle management:
Handling during experiments:
Maintain cold chain during experiment setup
Return antibody to appropriate storage immediately after use
Use sterile technique when accessing antibody stock
Avoid vortexing; mix by gentle flicking or inversion
Stability monitoring:
Include positive controls in each experiment to monitor activity
Document signal intensity over time for early detection of degradation
Consider periodic validation with fresh antibody aliquots
Replace antibody stock if significant activity loss is observed
This comprehensive approach has been shown to maintain antibody activity for up to one year when properly implemented , ensuring reliable experimental results across extended research projects.
ACTA1 recombinant monoclonal antibodies can significantly enhance molecular dynamics studies of ACTA1 mutations through integration of experimental validation with computational modeling:
Structure-function validation approach:
Use antibodies targeting specific ACTA1 domains to validate computational predictions
Epitope mapping experiments can confirm structural alterations predicted by simulation
Compare antibody binding affinities between wild-type and mutant proteins
Experimental validation of simulation predictions:
Molecular dynamics simulations of the R256H mutation showed significant structural changes in subdomains 2 and 4
Antibodies recognizing these regions can experimentally confirm predicted conformational changes
Sequential immunoprecipitation with domain-specific antibodies can track altered protein interactions
Thermal stability correlation methodology:
Molecular dynamics simulations predicted lower thermal stability for R256H mutant (confirmed experimentally)
Combine antibody-based detection with thermal shift assays
Protocol: Incubate protein samples at increasing temperatures (25-80°C), then analyze by Western blot with anti-ACTA1 antibodies to track denaturation kinetics
Mapping dynamic conformational changes:
Nucleotide binding state detection:
This integrated approach bridges computational predictions with experimental validation, providing robust evidence for how specific mutations alter ACTA1 structure and function at the molecular level .
Recent research has identified ACTA1 mutations in cardiomyopathy, requiring specialized methodological approaches for investigation:
Tissue-specific expression mapping:
Traditional view limited ACTA1 to skeletal muscle, but recent evidence shows cardiac expression
Methodology: Compare cardiac vs. skeletal ACTA1 expression using dual immunofluorescence with isoform-specific antibodies
Quantitative analysis using digital image analysis with standardized exposure parameters
Mutation-specific detection systems:
Contractile dynamics assessment:
Protein-protein interaction visualization:
Proximity ligation assay (PLA) methodology:
Use pairs of antibodies against ACTA1 and interaction partners
Detect altered interactions in disease tissue vs. controls
Quantify interaction sites per cell area
Combinatorial imaging with functional assessment:
Integrate contractile measurements with immunolocalization:
Perform calcium sensitivity measurements on isolated cardiomyocytes
Fix same samples for immunofluorescence analysis
Correlate functional deficits with protein localization/organization
These advanced techniques leverage the specificity of recombinant ACTA1 antibodies to understand how skeletal muscle actin mutations contribute to cardiac pathology, a relatively new finding in the field .
When studying cytoskeletal dynamics, researchers must carefully select between actin isoform-specific antibodies. This comparative analysis guides methodological selection:
Parameter | ACTA1 (Skeletal Muscle) | ACTC1 (Cardiac) | ACTB (Cytoplasmic β) | ACTG1 (Cytoplasmic γ) |
---|---|---|---|---|
Expression Pattern | Primarily skeletal muscle; some cardiac expression | Predominantly cardiac muscle | Ubiquitous, highest in non-muscle cells | Ubiquitous with tissue-specific variation |
Subcellular Distribution | Sarcomeric thin filaments | Sarcomeric thin filaments | Cortical cytoskeleton, stress fibers | Cortical cytoskeleton, specialized structures |
Dynamics Detection | Slower turnover, stable filaments | Moderately stable filaments | Rapid turnover, highly dynamic | Intermediate dynamics |
Experimental Applications | Muscle development, myopathies | Cardiomyopathies, heart development | Cell migration, division | Specialized functions |
Antibody Cross-Reactivity Concern | Moderate (with ACTC1) | Moderate (with ACTA1) | Low | Low |
Methodological selection guidance based on research objectives:
For tissue-specific dynamics:
Use ACTA1 antibodies for skeletal muscle-specific studies
Select antibodies targeting unique N-terminal regions for highest specificity
When studying tissue with multiple isoforms, employ dual-staining with isoform-specific antibodies
For mutation-specific research:
For dynamic filament studies:
This comparative approach ensures selection of the appropriate actin isoform antibody based on the specific biological question being addressed .
The host species and clone type significantly impact experimental outcomes when using ACTA1 antibodies. This methodological comparison guides appropriate selection:
Rabbit vs. Mouse Monoclonal ACTA1 Antibodies:
Implementation strategies for different hosts:
For mouse tissue immunostaining with mouse monoclonals:
Use specialized mouse-on-mouse blocking kits
Employ directly conjugated primary antibodies
For rabbit monoclonals on rabbit tissue:
Use tyramide signal amplification to reduce background
Select secondary antibodies pre-adsorbed against tissue species
Monoclonal vs. polyclonal methodological differences:
Monoclonals provide:
Consistent epitope recognition
Reduced batch-to-batch variation
Higher specificity for single epitope
Polyclonals offer:
Multiple epitope recognition (advantageous if one epitope is masked)
Potentially higher sensitivity
Greater susceptibility to non-specific binding
Recombinant technology impact:
Application-specific selection criteria:
For critical morphological studies: Rabbit recombinant monoclonals preferred
For routine Western blots: Either host suitable with appropriate controls
For multiplexed imaging: Select based on other antibodies in panel
For reproducibility between labs: Recombinant antibodies provide highest consistency
These methodological considerations should guide antibody selection based on specific experimental requirements and tissue types being investigated .