ACTA1 antibodies target skeletal alpha-actin, a 42 kDa protein essential for muscle contraction and cytoskeletal organization . The antibody binds to specific epitopes on the actin protein, enabling its visualization or quantification in biological samples.
ACTA1 antibodies are produced via immunization with synthetic peptides or recombinant proteins. They are available in monoclonal or polyclonal forms, depending on their production method.
CAB2319: Synthetic peptide corresponding to amino acids 1–100 of human ACTA1 .
17521-1-AP: Peptide immunogen (exact sequence unspecified) .
ACTA1 antibodies are employed in diverse experimental and clinical contexts.
ACTA1 mutations are linked to severe congenital myopathies, including:
Intranuclear Rod Myopathy: Features nuclear actin accumulation .
Fiber-Type Disproportion: Altered muscle fiber composition .
ACTA, Actin, actin, alpha 1, skeletal muscle, alpha skeletal muscle actin, alpha skeletal muscle, alpha-actin-1, ASMA, CFTD, CFTDM, MPFD, NEM1, NEM2, NEM3.
ACTA1 antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
PAT2F5AT.
Anti-human ACTA1 mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human ACTA1 amino acids 13-377 purified from E. coli.
Mouse IgG2a heavy chain and κ light chain.
ACTA1 (actin alpha 1, skeletal muscle) is a 42 kDa protein encoded by the human ACTA1 gene . It functions as a major component of muscle fibers and plays an essential role in skeletal muscle contraction. The protein is primarily localized in the cytoplasm and cytoskeleton of muscle cells, where it contributes to the structural integrity and contractile properties of skeletal muscle .
ACTA1 is particularly significant in research because mutations in the ACTA1 gene have been linked to various congenital myopathies and muscle disorders, making it a key target for studies in muscle development and disease mechanisms . Understanding ACTA1 function and regulation provides insights into fundamental muscle biology and potential therapeutic targets for muscle-related conditions.
Based on current research tools, ACTA1 antibodies have been validated for multiple applications:
Application | Recommended Dilution | Common Uses |
---|---|---|
Western Blot (WB) | 1:500 - 1:2000 | Protein quantification, molecular weight confirmation |
Immunohistochemistry (IHC) | Varies by antibody | Tissue localization, pattern analysis |
Immunofluorescence (IF/ICC) | 1:50 - 1:200 | Subcellular localization, co-localization studies |
ELISA | Antibody-dependent | Quantitative protein detection |
These applications enable researchers to investigate ACTA1 expression, localization, and interactions in various experimental contexts . The selection of application should be guided by specific research questions and available sample types.
Commercial ACTA1 antibodies demonstrate reactivity primarily with human, mouse, and rat samples . This cross-reactivity stems from the high sequence conservation of ACTA1 across mammalian species. Researchers have successfully used antibodies like CAB2319 and RP1070 with samples from these three species in applications including Western blot, immunohistochemistry, and immunofluorescence .
Cross-reactivity with other species such as canine samples may be possible but requires experimental validation. When working with non-validated species, researchers should conduct preliminary tests comparing tissues with known ACTA1 expression patterns across species to confirm antibody performance .
Distinguishing between actin isoforms (ACTA1, ACTA2, ACTB, ACTC, etc.) presents a significant challenge due to high sequence homology. Effective isoform differentiation requires:
Epitope selection: Choose antibodies raised against sequences unique to ACTA1. For example, the C-terminal region (around aa 359-377) may provide better isoform specificity than highly conserved regions .
Validation strategy:
Peptide competition assays using isoform-specific peptides
Western blot comparison using tissues with known differential isoform expression
Parallel analysis with isoform-specific mRNA quantification
Control selection:
Positive controls: Skeletal muscle (high ACTA1 expression)
Negative controls: Tissues expressing other actin isoforms but minimal ACTA1
Complementary approaches: Use multiple antibodies targeting different epitopes to confirm specificity and consider orthogonal techniques like mass spectrometry for definitive isoform identification .
When investigating myopathies associated with ACTA1 mutations, researchers should consider several methodological factors:
Epitope accessibility: ACTA1 mutations may alter protein conformation or aggregation state, affecting antibody binding. Multiple antibodies targeting different epitopes may be necessary to ensure detection .
Fixation optimization: Standard paraformaldehyde (PFA) fixation is recommended for most applications, but modifications may be necessary for tissues with protein aggregates or structural abnormalities .
Control selection:
Age and sex-matched healthy controls
Non-affected tissues from the same patient
Related myopathies with different molecular causes
Complementary techniques: Combine antibody-based detection with genetic analysis, protein function assays, and structural studies to comprehensively characterize mutation effects.
Quantitative analysis: Develop standardized scoring systems for abnormal ACTA1 distribution patterns to enable objective comparison between samples and studies .
When extending ACTA1 antibody use to new models, a systematic validation approach is essential:
Sequence alignment analysis: Compare the immunogen sequence with the target species ACTA1 sequence to predict cross-reactivity potential. For example, the RP1070 antibody immunogen (C-terminal peptide ITKQEYDEAGPSIVHRKCF) should be compared across species .
Stepwise validation protocol:
Begin with Western blot to confirm target molecular weight (42 kDa for ACTA1)
Progress to fixed-cell immunostaining to assess subcellular localization
Validate in tissue sections with appropriate controls
Sensitivity optimization:
Test multiple antibody concentrations (starting with manufacturer recommendations)
Evaluate different detection systems and signal amplification methods
Optimize blocking conditions to minimize background in the specific model
Document validation data thoroughly for publication, including positive and negative controls, optimization steps, and any limitations identified .
Successful ACTA1 detection depends on appropriate tissue preparation:
For challenging samples such as diseased muscle with altered architecture, modifications to standard protocols may be necessary. Researchers should systematically test multiple conditions when establishing protocols for new tissue types or disease states .
Achieving optimal signal-to-noise ratio for ACTA1 Western blot requires attention to several technical aspects:
Sample preparation:
Include protease inhibitors during extraction
Fresh tissue samples yield better results than archived samples
For skeletal muscle, specialized extraction buffers with higher salt concentrations may improve solubilization
Antibody optimization:
Blocking and washing:
Extend blocking time (1-2 hours) for high background issues
Use 5% BSA instead of milk for phospho-sensitive applications
Implement stringent washing protocols (4-5 washes of 10 minutes each)
Controls:
For detecting low abundance ACTA1, researchers can employ several sensitivity-enhancing strategies:
Sample enrichment:
Concentrate protein samples through immunoprecipitation
For tissue sections, optimize fixation to preserve antigen availability
Signal amplification methods:
Tyramide signal amplification (TSA) can increase sensitivity 10-100 fold for IHC/IF
High-sensitivity chemiluminescent substrates for Western blot
Consider biotin-streptavidin systems for additional amplification
Detection optimization:
Image acquisition:
Interpreting ACTA1 staining pattern variations requires careful consideration of multiple factors:
Normal pattern characteristics:
Regular, striated pattern reflecting sarcomeric organization
Consistent intensity across similar fiber types
Uniform subcellular distribution
Pathological pattern indicators:
Sarcomeric disorganization: irregular, fragmented staining
Protein aggregation: focal intense staining
Fiber type conversion: altered intensity across fiber populations
Necrosis/regeneration: absence or intensification of staining
Quantitative assessment methodology:
Measure staining intensity across multiple fields (minimum 5-10 fields per sample)
Compare matched anatomical regions between patient and control samples
Account for muscle fiber type composition differences
Complementary analyses:
While actin is commonly used as a loading control, ACTA1 specifically presents unique considerations:
Tissue-specific expression profile:
ACTA1 is predominantly expressed in skeletal muscle
Expression is limited in non-muscle tissues, making it unsuitable as a universal loading control
For skeletal muscle studies, ACTA1 may be appropriate but could be affected by experimental conditions
Potential confounding factors:
ACTA1 expression may be altered in muscle development, regeneration, or disease states
Mechanical loading, denervation, and exercise can all affect ACTA1 levels
Cytoskeletal-targeting treatments may alter ACTA1 expression or stability
Technical limitations:
High abundance can lead to signal saturation
Dynamic range limitations may mask subtle changes in target proteins
Stripping and reprobing membranes may affect ACTA1 detection
Recommended alternatives:
Investigating ACTA1 in dynamic processes (development, adaptation, regeneration) requires thoughtful experimental design:
Temporal considerations:
Establish appropriate time points based on the process being studied
For development: embryonic, neonatal, juvenile, and adult stages
For regeneration: early (0-3 days), middle (4-7 days), and late (8+ days) phases
Spatial mapping:
Compare multiple muscle groups with different fiber type compositions
Examine regional differences within the same muscle
Consider three-dimensional reconstruction for complex architectural changes
Functional correlation:
Combine ACTA1 detection with functional measurements (force production, contractility)
Correlate protein expression with mechanical properties
Integrate with electrophysiological measurements where relevant
Molecular context:
Adapting ACTA1 antibody applications to high-throughput screening requires:
Assay miniaturization:
Microplate-based immunoassays (ELISA, AlphaLISA)
High-content imaging of cell arrays
Tissue microarrays for parallel analysis of multiple samples
Automation considerations:
Robotics-compatible protocols with minimal manual intervention
Standardized reagents with batch-to-batch consistency
Optimized antibody concentrations for consistent performance
Data acquisition and analysis:
Automated image analysis algorithms for pattern recognition
Machine learning approaches for classification of staining patterns
Integrated data management systems for large-scale experiments
Validation strategy:
Integrating ACTA1 antibody-based detection with complementary techniques enhances research insights:
Combining with genetic approaches:
CRISPR/Cas9 modification of ACTA1 requires validation of antibody recognition for mutated proteins
RNA interference experiments should correlate protein reduction with mRNA knockdown
Transgenic models expressing tagged ACTA1 need validation with endogenous protein detection
Integration with proteomics:
Immunoprecipitation followed by mass spectrometry to identify interaction partners
Orthogonal validation of proteomic findings using antibody-based methods
Assessment of post-translational modifications identified in proteomic screens
Live-cell applications:
Considerations for antibody fragments or nanobodies for intracellular applications
Assessment of antibody effects on ACTA1 function when used in living systems
Correlation between fixed-tissue staining patterns and live dynamics
Super-resolution microscopy:
When faced with contradictory results from different ACTA1 antibodies, researchers should implement a systematic investigation:
Characterize antibody parameters:
Compare immunogen sequences and epitope locations
Assess antibody types (monoclonal vs. polyclonal) and host species
Review validation data provided by manufacturers
Technical assessment:
Test antibodies side-by-side under identical conditions
Evaluate concentration-dependent effects on staining patterns
Assess impacts of different detection methods on results
Biological validation:
Use genetically modified systems with known ACTA1 status
Correlate antibody staining with mRNA expression
Perform peptide competition assays to confirm specificity
Resolution strategy:
Prioritize antibodies with most comprehensive validation
Report discrepancies transparently in publications
Consider that different antibodies may reveal different aspects of protein biology (conformation, complexes, modifications)
Implement orthogonal approaches to resolve persistent contradictions
In mammals, there are six known isoforms of actin:
The β- and γ-actin isoforms, known as cytoplasmic actins, are highly homologous and differ by only four amino acids. These isoforms are involved in maintaining cell structure and motility .
Mouse anti-human actin antibodies are monoclonal antibodies produced by immunizing mice with synthetic peptides corresponding to human actin sequences. These antibodies are widely used in research to detect and study actin in various applications, including Western blotting, immunohistochemistry, immunofluorescence, and flow cytometry.
One commonly used mouse anti-human actin antibody is the β-Actin (8H10D10) Mouse mAb. This antibody detects endogenous levels of total β-actin protein and may cross-react with cytoplasmic γ-actin due to the high sequence identity between these isoforms .
Mouse anti-human actin antibodies are valuable tools in cell biology research. They are used to:
These antibodies are specific to human, mouse, and rat actin and do not cross-react with other actin isoforms such as α-skeletal, α-cardiac, or α-smooth muscle actin .