ACTA1 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Production

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 .

Target Details

  • Molecular Weight: 42 kDa (observed)

  • Cellular Localization: Cytoplasm, cytoskeleton

  • Gene ID: 58 (Human), 11459 (Mouse), 29437 (Rat)

  • UniProt ID: P68133 (Human), P68134 (Mouse), P68136 (Rat)

Muscle Disease Studies

ACTA1 mutations are linked to nemaline myopathy, congenital fiber-type disproportion, and core myopathy . These antibodies facilitate:

  • Detection of ACTA1 expression in muscle biopsies .

  • Analysis of structural defects in myopathies via immunofluorescence .

Cellular Mechanisms

  • Muscle Contraction: Visualizing actin-myosin interactions in skeletal muscle .

  • Cytoskeletal Dynamics: Tracking ACTA1 in cell motility assays .

Validation and Performance

  • 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 .

Advancements in Production

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 .

Associated Diseases and Tissues

Disease/TissueResearch FocusKey Publications
Nemaline MyopathyACTA1 aggregation in muscle fibers
Cardiovascular DiseaseACTA1 expression in hypertrophic myocardium
Muscle RegenerationRole in injury repair mechanisms

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

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.

Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery time may vary depending on the chosen method of purchase or location. Please consult your local distributor for specific delivery time information.
Synonyms
Actin, alpha skeletal muscle (Alpha-actin-1) [Cleaved into: Actin, alpha skeletal muscle, intermediate form], ACTA1, ACTA
Target Names
Uniprot No.

Target Background

Function
Actins are highly conserved proteins that are involved in various types of cell motility and are ubiquitously expressed in all eukaryotic cells.
Gene References Into Functions
  1. Our study demonstrates that elevated ActA levels are an independent prognostic factor for survival in cancer patients. ActA appears to influence survival by contributing to the development of cachexia and loss of skeletal muscle mass, suggesting a role beyond simply indicating the severity of the neoplastic disease or inflammatory process. PMID: 28712119
  2. The study confirmed ACTA1 mutations in four patients, including one with intranuclear rods, one with large intracytoplasmic aggregates, and two with nemaline intracytoplasmic rods. PMID: 28780987
  3. The study shows that clinically severe ACTA1-related myopathy can present with muscle morphological findings suggestive of cytoplasmic body myopathy in the absence of definitive nemaline rods. PMID: 28416349
  4. Shorter than normal thin filament length contributes to the impaired force generation in patients with thin filament myopathy, but only in those who harbor specific mutations in NEB or ACTA1. PMID: 27074222
  5. Over-expression of TNC, SMA, and vimentin were significantly correlated with the lower overall survival in prostate cancer patients. PMID: 28341124
  6. ANA and ASMA evaluation in patients with liver transplantation and no history of autoimmune disease has no clinical relevance, since it varies in time and is not related to any risk factors or liver injury. Routine autoimmunity evaluation should be avoided. PMID: 28337446
  7. Upon actin engagement, the N-terminal "strap" and helix 1 are displaced from the vinculin tail helical bundle to mediate actin bundling. PMID: 26493222
  8. This study reported new information on the frequency and phenotypes of congenital myopathy caused by ACTA1 mutations in subjects >/=5 years of age. PMID: 26172852
  9. The authors propose that Lpd delivers Ena/VASP proteins to growing barbed ends and increases their actin polymerase activity by tethering them to actin filaments. PMID: 26295568
  10. Mutations in ACTA1 can cause pathologic features consistent with myofibrillar myopathy. PMID: 25913210
  11. TIMP-1 significantly increased levels of alpha-SMA. PMID: 25895516
  12. A novel homozygous recessive missense variant (c.460G>C, P.(Val154Leu)) was found in two brothers with infantile-onset congenital muscular dystrophy with rigid spine. PMID: 25182138
  13. Myopathy associated with zebra bodies is part of the spectrum of myopathies associated with the ACTA1 gene. PMID: 25747004
  14. A new scapuloperoneal phenotype associated with an ACTA1 mutation. PMID: 25938801
  15. Antisynthetase syndrome-associated myositis is characterized by distinctive myonuclear actin filament inclusions. PMID: 25746564
  16. The predominant pathway mediated by Australian bat lyssavirus G envelope for internalization into HEK293T cells is clathrin-and actin-dependent, also requiring Rab5. PMID: 24576301
  17. Data show that the width and speed of the waves depend counter-intuitively on parameters, negative feedback, and the F-actin time scale. PMID: 23831272
  18. Although most cases of the severe infantile form of nemaline myopathy caused by ACTA1 mutations are sporadic and have no family history PMID: 24313005
  19. These findings suggest that the direct contact of Dengue virus 2 E protein with 43 kDa actin protein may have a crucial function in Dengue virus 2 infection of ECV304 cells. PMID: 23376163
  20. Myotonia in adult human skeletal actin transgenic mice may be explained on the basis of a mosaic expression of ClC-1 channels in different fibres and/or on alterations of other conductances. PMID: 23247112
  21. Respiratory muscles from an animal model of nemaline myopathy with an ACTA1 mutation show distinct muscle fiber weakness compared to limb muscles. PMID: 23656990
  22. The objective was to evaluate the presence and distribution of the lubricating and anti-adhesion glycoprotein lubricin and cells containing the contractile isoform smooth muscle alpha-actin (SMA) in pseudomembranes around loose hip prostheses. PMID: 23174700
  23. Analysis of the pattern of evolutionarily conserved basic and acidic residues that constitutes the binding interface of actin-tropomyosin PMID: 23420843
  24. Data show that GnRHR activation affected several cellular markers of locomotion, including actin organization and polymerization as well as active RhoA-GTP levels. PMID: 23176180
  25. Data indicate that actin nucleation on Chlamydia inclusions exhibited moderate depolymerization dynamics. PMID: 23071671
  26. The organization of actin and cytokeratin cytoskeleton and the expression of TCTP, p53, cyclin A, RhoA and actin in HIO180 non-transformed ovarian epithelial cells, and OVCAR3 and SKOV3 ovarian epithelial cancer cells, was studied. PMID: 23042265
  27. Data show that the urinary messenger RNA (mRNA) levels of alpha-smooth muscle actin (alpha-SMA), fibronectin, and matrix metalloproteinase-9 (MMP-9) were significantly higher in the diabetic nephropathy (DN) group, and mRNA levels increased with DN progression. PMID: 21824169
  28. High alpha-actin is associated with colorectal carcinogenesis. PMID: 21912905
  29. Over-expression of Nkx2.5 and/or cardiac alpha-actin inhibit the contraction ability of adipose tissue-derived stromal cells-derived cardiomyocytes. PMID: 21691712
  30. Platelets gal-1 forms an intracellular complex with monomeric actin. PMID: 22081313
  31. Herpesviruses exploit actin and actin-associated myosin motors for viral entry and virion egress. [review] PMID: 21994736
  32. CK2 regulates vaccinia virus dissemination and actin tail formation. PMID: 22209233
  33. Actin plays differential roles in trafficking of Epstein-Barr virus in B cells and epithelial cells. PMID: 22031939
  34. A link between extramuscular expression of alpha-skeletal muscle actin and clinical symptoms in non-skeletal muscle tissues of patients with ACTA1 mutations, and probably a functional role of alpha-skeletal muscle actin during fetal development PMID: 21514153
  35. The filopodium: a stable structure with highly regulated repetitive cycles of elongation and persistence depending on the actin cross-linker fascin PMID: 21975552
  36. Actin accumulates throughout the activated immunological synapse. PMID: 21931536
  37. Actin-EGFP and ezrin-EGFP accumulated below pilus-coated beads when force was applied. PMID: 21340023
  38. Data show that concurrent phosphorylation of cortactin by ERK1/2 and tyrosine kinases enables cells with the ability to regulate actin dynamics. PMID: 21079800
  39. BCL2 interaction with actin in vitro may inhibit cell motility by enhancing actin polymerization PMID: 20716950
  40. Results suggest that actin polymerization and bundling by VASP are critical for spine formation, expansion, and modulating synaptic strength. PMID: 20826790
  41. Centrosomal actin was detected with the anti-actin antibody 1C7 that recognizes antiparallel ("lower dimer") actin dimers. PMID: 21108927
  42. These findings demonstrated that PI3K-mediated actin rearrangements are required for Cronobacter sakazakii invasion of human brain microvascular endothelial cells. PMID: 20809254
  43. Results confirm that the DNaseI-binding loop (D-loop) is involved in stabilization of skeletal muscle actin structure, both as monomers and filaments; actin is stabilized by ligands (by phalloidin, aluminum fluoride, & ATP [and to lesser extent by ADP]). PMID: 20718862
  44. Simultaneous tracking of both TCR clusters and GFP-actin speckles reveals their dynamic association and individual flow patterns. Actin retrograde flow directs the inward transport of TCR clusters PMID: 20686692
  45. We conclude that the Drosophila indirect flight muscles provide a good model system for studying ACTA1 mutations PMID: 20452215
  46. Data demonstrate for the first time that L-plastin contributes to the fine-tuning of actin turn-over, an activity which is regulated by Ser5 phosphorylation promoting its high affinity binding to the cytoskeleton. PMID: 20169155
  47. Podocyte BK(Ca) channels are regulated by synaptopodin, Rho, and actin microfilaments. PMID: 20630939
  48. These data identify NHS as a new regulator of actin remodeling. PMID: 20332100
  49. ACTA1 mutations are involved in fiber size disproportion in congenital myotonic dystrophy PMID: 20179953
  50. First insights of S100A10 function as a regulator of the filamentous actin network. PMID: 20100475

Show More

Hide All

Database Links

HGNC: 129

OMIM: 102610

KEGG: hsa:58

STRING: 9606.ENSP00000355645

UniGene: Hs.1288

Involvement In Disease
Nemaline myopathy 3 (NEM3); Myopathy, actin, congenital, with excess of thin myofilaments (MPCETM); Myopathy, congenital, with fiber-type disproportion (CFTD); Myopathy, scapulohumeroperoneal (SHPM)
Protein Families
Actin family
Subcellular Location
Cytoplasm, cytoskeleton.

Q&A

What is the difference between conventional and recombinant monoclonal antibodies against ACTA1?

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 .

What are the common applications for ACTA1 recombinant monoclonal antibodies in research?

ACTA1 recombinant monoclonal antibodies are versatile research tools with multiple validated applications:

ApplicationTypical Dilution RangeKey Considerations
Western Blot (WB)1:500 - 1:5000Detects ~42 kDa band; sensitive to loading controls
Immunohistochemistry (IHC)1:50 - 1:500Works on both frozen and paraffin sections; epitope retrieval critical
Immunofluorescence (IF)1:30 - 1:200Co-staining with cytoskeletal markers recommended
Flow Cytometry (FCM)Vendor-specificCell permeabilization required for intracellular target
ELISAAssay-dependentValidated 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 .

How should I design experiments to validate ACTA1 antibody specificity in my research model?

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:

    • Western blot: Confirm single band at expected 42 kDa molecular weight

    • Immunohistochemistry: Compare staining pattern with established ACTA1 distribution

    • Peptide competition assay: Pre-incubate antibody with immunizing peptide to block specific binding

  • 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 .

What considerations should be taken when using ACTA1 antibodies for studying ACTA1 gene mutations in myopathies?

When studying ACTA1 mutations in myopathies, several methodological considerations are essential:

  • Epitope mapping relative to mutation sites:

    • Select antibodies whose epitopes are distant from mutation sites to avoid false negatives

    • For comprehensive studies, use multiple antibodies targeting different ACTA1 regions

    • Consider the specific amino acid sequence recognized by the antibody (e.g., within amino acids 1-100 of human ACTA1)

  • 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:

    • ACTA1 mutations can alter protein folding and epitope accessibility

    • Native vs. denatured detection methods may yield different results

    • R256H mutation, for example, significantly alters thermal stability (melting temperature 46.1°C vs. 58.9°C for wild-type)

  • 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:

    • Integrate antibody detection with functional assays measuring actin polymerization

    • Consider polymerization kinetics, which can be significantly altered in mutants

These considerations are particularly important when studying the 447 pathogenic/likely pathogenic ACTA1 variants associated with conditions like nemaline myopathy (NEM) and cardiomyopathy .

What is the optimal methodology for using ACTA1 antibodies in immunohistochemistry of muscle biopsies?

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:

    • Block with 5% normal serum from secondary antibody species

    • Primary antibody dilution: Start with 1:100, optimize as needed

    • Incubation: Overnight at 4°C or 1 hour at room temperature

  • 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 .

How can I optimize Western blot protocols for detecting ACTA1 in different tissue types?

Optimizing Western blot protocols for ACTA1 detection across different tissue types requires addressing tissue-specific challenges:

  • Tissue-specific extraction optimization:

    Tissue TypeLysis Buffer RecommendationSpecial Considerations
    Skeletal MuscleRIPA with protease inhibitors and 1mM PMSFMechanical homogenization critical
    Cardiac TissueModified RIPA (higher detergent)Requires longer extraction time
    Cell LinesStandard RIPA or NP-40 bufferGentle cell scraping preferred
    Brain TissueSpecialized neuronal extraction bufferAvoid 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:

    • Primary antibody dilution range: 1:500-1:2000

    • Incubation in 5% non-fat milk or BSA in TBST

    • Overnight incubation at 4°C maximizes specific binding

  • 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 .

What are the most common troubleshooting issues when using ACTA1 antibodies, and how can they be resolved?

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:

      • Standardize fixation protocols (time, temperature, pH)

      • Section thickness optimization (5-8 μm optimal)

      • Store antibody in small aliquots with glycerol to prevent freeze-thaw damage

      • Consider carrier-free formulations for specialized applications

  • Poor reproducibility between experiments:

    • Root causes: Antibody batch variation, protocol inconsistency

    • Solutions:

      • Use recombinant antibodies for batch consistency

      • Document detailed protocols including lot numbers

      • Include internal controls in each experiment

      • Consider validated positive controls (HeLa, A-431, C6 cell lines)

These targeted approaches address specific methodological challenges rather than simply recommending general protocol adjustments.

How should ACTA1 antibodies be stored and handled to maintain optimal activity over time?

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:

    • Store antibody at -20°C to -80°C for maximum stability

    • Add cryoprotectants if buffer-exchanging:

      • 50% glycerol final concentration for -20°C storage

      • Trehalose (5-10%) provides additional stability

    • Avoid antibody formulations containing only PBS for long-term storage

  • Working stock preparation:

    • For frequent use, store small aliquots (10-20 μL) at 4°C for up to one month

    • Return original stock to freezer immediately after aliquoting

    • Add sterile-filtered preservative (0.02% sodium azide) to working stocks

    • Document date of thawing and aliquoting for each vial

  • Freeze-thaw cycle management:

    • Minimize freeze-thaw cycles (ideally ≤5 total)

    • Thaw on ice rather than at room temperature

    • Quick-freeze aliquots in dry ice/ethanol bath before returning to freezer

    • Consider specialized freezer boxes that maintain consistent temperature

  • 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.

How can ACTA1 recombinant monoclonal antibodies be utilized in molecular dynamics studies of ACTA1 mutations?

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:

    • ACTA1 mutations alter ADP binding and subdomains 2-4 interactions

    • Use conformation-sensitive antibodies to detect these states

    • Develop ELISA-based assays with antibodies recognizing native vs. altered conformations

  • Nucleotide binding state detection:

    • Various ACTA1 mutations affect nucleotide binding and retention

    • Use antibodies recognizing ACTA1 in different nucleotide-bound states

    • Combine with competitive nucleotide binding assays to correlate simulation with experimental data

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 .

What are the advanced methods for studying ACTA1 mutations in cardiomyopathy using recombinant antibodies?

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:

    • For dilated cardiomyopathy-associated R256H mutation :

      • Generate phospho-specific or conformation-specific antibodies recognizing mutant forms

      • Validate specificity using recombinant wild-type and mutant proteins

      • Apply to tissue sections for spatial distribution analysis

  • Contractile dynamics assessment:

    • Protocol for in vitro motility assays with ACTA1 antibodies:

      • Reconstruct thin filaments with varying ratios of wild-type:mutant ACTA1

      • Measure filament velocity at different calcium concentrations

      • R256H mutation showed speed-pCa curves with unchanged pCa50 but decreased maximum speed

  • 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 .

How do ACTA1 antibodies compare to antibodies against other actin isoforms for studying cytoskeletal dynamics?

When studying cytoskeletal dynamics, researchers must carefully select between actin isoform-specific antibodies. This comparative analysis guides methodological selection:

ParameterACTA1 (Skeletal Muscle)ACTC1 (Cardiac)ACTB (Cytoplasmic β)ACTG1 (Cytoplasmic γ)
Expression PatternPrimarily skeletal muscle; some cardiac expressionPredominantly cardiac muscleUbiquitous, highest in non-muscle cellsUbiquitous with tissue-specific variation
Subcellular DistributionSarcomeric thin filamentsSarcomeric thin filamentsCortical cytoskeleton, stress fibersCortical cytoskeleton, specialized structures
Dynamics DetectionSlower turnover, stable filamentsModerately stable filamentsRapid turnover, highly dynamicIntermediate dynamics
Experimental ApplicationsMuscle development, myopathiesCardiomyopathies, heart developmentCell migration, divisionSpecialized functions
Antibody Cross-Reactivity ConcernModerate (with ACTC1)Moderate (with ACTA1)LowLow

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:

    • ACTA1 antibodies are superior for studying nemaline myopathy (74% of ACTA1 mutations)

    • For cardiomyopathies, consider both ACTA1 and ACTC1 antibodies

    • Use epitope mapping to ensure antibody binding site is distinct from mutation site

  • For dynamic filament studies:

    • Live-cell imaging: Combine fluorescently tagged actin with fixed-cell validation using antibodies

    • For turnover studies: Use FRAP combined with immunofluorescence validation

    • For polymerization studies: Use pyrene-actin assays with antibody validation

This comparative approach ensures selection of the appropriate actin isoform antibody based on the specific biological question being addressed .

What are the methodological differences when using ACTA1 antibodies from different host species and clone types?

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:

    ParameterRabbit MonoclonalMouse Monoclonal (e.g., 5C5 clone)
    Epitope RecognitionOften N-terminal region (amino acids 1-100) Mid-region or C-terminal epitopes
    Background in Murine TissuesLower (reduced anti-mouse cross-reactivity)Higher (secondary antibodies may detect endogenous mouse IgG)
    Multiplexing CapabilityCompatible with mouse primary antibodiesRequires same-species detection strategies
    Reported SensitivityHigher (especially recombinant versions) Good but generally lower than rabbit
    ApplicationsSuperior for IHC, IF; excellent for WBHistorical standard for WB; variable IHC performance
  • 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:

    • Recombinant rabbit monoclonals combine advantages:

      • Consistent performance of monoclonals

      • High affinity of rabbit antibodies

      • Eliminated batch variation

      • Defined sequence and glycosylation profile

  • 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 .

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 2024 Thebiotek. All Rights Reserved.