ACTB Antibody

Actin Beta, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Definition and Types of ACTB Antibodies

ACTB antibodies are immunoglobulins designed to bind specifically to beta-actin, a 42 kDa protein encoded by the ACTB gene. They are classified as:

  • Monoclonal antibodies: Engineered to target specific epitopes (e.g., clone AC-15, 6D6, 8-7A5).

  • Polyclonal antibodies: Recognize multiple epitopes on the ACTB protein (e.g., GTX110564, 20536-1-AP).

These antibodies are validated for use in Western blotting (WB), immunocytochemistry/immunofluorescence (ICC/IF), and immunohistochemistry (IHC) .

Applications of ACTB Antibodies

ACTB antibodies serve as critical reagents in diverse experimental workflows:

ApplicationKey UsesAntibody Examples
Western BlottingLoading control; quantification of protein expression.AC-15 (Abcam), GTX110564 (GeneTex)
ICC/IFVisualization of cytoskeletal dynamics in live or fixed cells.6D6 (PMC study), 8-7A5 (DSHB)
IHCDetection of ACTB in tissue sections, including tumor samples.Sigma-Aldrich clone, 20536-1-AP (Proteintech)
Knockout ValidationConfirmation of ACTB depletion in CRISPR/Cas9 or siRNA experiments.AC-15 (validated with ACTB KO cell lines)

Heterogeneous Expression of ACTB

Recent studies highlight variable ACTB expression across cell types and cancer subtypes:

Cancer TypeACTB Expression PatternRelevance
Bladder CancerEpithelial cells show strong staining, while stromal/endothelial cells exhibit weak reactivity .Potential diagnostic/prognostic marker.
General CancersDifferential expression in tumor vs. healthy tissue correlates with survival outcomes .Biomarker for cancer progression.

The 6D6 monoclonal antibody demonstrated high specificity for epithelial ACTB in bladder cancer (BLCA), with weak binding to non-epithelial cells . This heterogeneity underscores the need for clone-specific optimization.

Role in Cell Migration and Gene Regulation

Beta-actin regulates:

  • Membrane protrusion dynamics: Loss of β-actin in MEFs reduces migration velocity and lamellipodia protrusion .

  • G-actin/F-actin balance: β-actin depletion alters the G-actin pool, impacting gene expression linked to cell cycle and motility .

Antibody Validation and Specificity

Key validation strategies include:

  1. Knockout Cell Lines: AC-15 (Abcam) and 20536-1-AP (Proteintech) show no signal in ACTB KO cells, confirming specificity .

  2. Epitope Mapping: Sigma-Aldrich’s β-actin antibody targets a fixation-resistant epitope, enabling use with formalin-fixed paraffin-embedded (FFPE) samples .

  3. Species Reactivity: Most antibodies (e.g., AC-15, 20536-1-AP) cross-react with human, mouse, and rat ACTB, while others (e.g., 8-7A5) show rat/buffalo specificity .

Challenges and Considerations

  • Heterogeneous Expression: ACTB levels vary between cell types, necessitating careful interpretation in cancer studies .

  • Fixation Sensitivity: Optimal results require compatibility with fixatives (e.g., Sigma-Aldrich’s antibody works with formalin, ethanol, or methacarn) .

  • Clone Selection: Monoclonal antibodies (e.g., 6D6, AC-15) offer epitope specificity, while polyclonal antibodies provide broader reactivity .

Product Specs

Introduction

Belonging to a family of six different isoforms, Actin Beta (ACTB) is one of two nonmuscle cytoskeletal actins found in humans. These actins are highly conserved proteins essential for cell motility, structure, and integrity. Alpha actins are crucial for the contractile apparatus.

Physical Appearance

The product is a clear and colorless solution that has been sterilized through filtration.

Formulation

This solution has a concentration of 1mg/ml and contains the following components: PBS (pH 7.4), 10% Glycerol, and 0.02% Sodium Azide.

Storage Procedures

For short-term storage (up to 1 month), keep the product refrigerated at 4°C. For long-term storage, it is recommended to store at -20°C. Avoid repeated freeze-thaw cycles.

Stability / Shelf Life

The product remains stable for 12 months when stored at -20°C and for 1 month at 4°C.

Applications

This ACTB antibody has undergone rigorous testing using ELISA, Western blot analysis, and ICC/IF techniques to ensure its specificity and reactivity. However, optimal results may require titration depending on the specific application. Therefore, users are encouraged to optimize the reagent concentration for their particular experimental conditions.

Synonyms

Actin Beta, PS1TP5-Binding Protein 1, Beta Cytoskeletal Actin, Actin, Cytoplasmic 1, Beta-Actin, PS1TP5BP1, BRWS1, ACTB.

Purification Method

ACTB antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.

Type
Mouse Anti Human Monoclonal.
Clone

PAT4G6AT.

Immunogen

Anti-human ACTB mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human ACTB amino acids 1-375 purified from E. coli.

Ig Subclass

Mouse IgG1 heavy chain and κ light chain.

Q&A

What is ACTB and why are ACTB antibodies significant in research?

ACTB refers to β-actin, a highly conserved cytoskeletal protein essential for cellular structure, motility, and intracellular transport mechanisms. ACTB antibodies are significant in research for several key reasons. First, they serve as critical tools for studying cytoskeletal dynamics across diverse cell types. Second, they function as essential loading controls in Western blot experiments due to β-actin's consistent expression in most cell types. Third, they enable visualization of cytoskeletal arrangements through immunofluorescence techniques. Finally, as recent research demonstrates, anti-ACTB antibodies may serve as potential biomarkers for neurodegenerative conditions such as amyotrophic lateral sclerosis (ALS), where serum levels are significantly elevated compared to both normal controls and Parkinson's disease patients . This dual role as both research tool and potential clinical biomarker makes ACTB antibodies particularly valuable across multiple scientific disciplines.

What detection methods provide optimal sensitivity for ACTB antibodies?

The optimal detection method for ACTB antibodies depends on the specific research context and sensitivity requirements. For quantitative analysis of serum anti-ACTB antibody levels, the amplified luminescent proximity homogeneous assay-linked immunosorbent assay (AlphaLISA) has demonstrated excellent sensitivity and reproducibility . This method offers advantages over traditional ELISA by requiring smaller sample volumes and providing enhanced signal-to-noise ratios. For qualitative detection in tissues or cells, immunohistochemistry and immunofluorescence using fluorophore-conjugated secondary antibodies remain standard approaches. Western blotting utilizing chemiluminescence detection systems provides reliable semi-quantitative analysis for ACTB in protein extracts. For more precise binding kinetics assessment, surface plasmon resonance (SPR) and bio-layer interferometry enable real-time analysis of antibody-antigen interactions without labels . When selecting a detection method, researchers should consider the required sensitivity threshold, sample type, available instrumentation, and whether qualitative or quantitative data is needed for their specific experimental objectives.

How should researchers validate ACTB antibodies for experimental applications?

Comprehensive validation of ACTB antibodies requires a multi-faceted approach to ensure specificity, sensitivity, and reproducibility. Begin with positive and negative control experiments using tissues or cell lines with known ACTB expression patterns. For monoclonal antibodies, compare results across multiple clones targeting different epitopes of β-actin. Perform competitive binding assays with purified ACTB protein to confirm specificity. Consider knockdown or knockout validation where cells with reduced or eliminated ACTB expression should show corresponding decreases in antibody signal. Validate across multiple techniques (Western blot, immunohistochemistry, flow cytometry) to confirm consistent performance across applications. For quantitative assays, establish standard curves using recombinant ACTB at known concentrations. Cross-reactivity testing against related proteins (such as other actin isoforms) is essential, particularly when working across species. Finally, inter-laboratory validation through collaboration can significantly strengthen confidence in antibody performance. Thorough documentation of validation procedures should accompany all experimental results to ensure reproducibility and reliability of findings.

How can computational approaches enhance prediction of ACTB antibody-antigen interactions?

Computational approaches have revolutionized the prediction and optimization of antibody-antigen interactions, including those involving ACTB. Current computational tools like Rosetta, FoldX, and SAAMBE-3D can perform mutational scanning of the antibody-antigen interface to identify key residues critical for binding interactions . Rosetta and FoldX employ built-in energy functions to compute energetic effects of mutations, while SAAMBE-3D utilizes machine learning algorithms trained on experimental binding energy databases to quickly calculate changes in binding energy for amino acid substitutions . When crystal structures of ACTB-antibody complexes are unavailable, docking protocols such as HDOCK, ZDOCK, and RosettaDock can predict complex formation from individual structures . For optimal results, researchers should integrate computational predictions with experimental validation through techniques like alanine scanning mutagenesis or hydrogen-deuterium exchange mass spectrometry. The computational approach becomes particularly valuable when designing antibodies with improved specificity for distinct ACTB conformations or post-translational modifications. As machine learning algorithms continue to improve with expanded training datasets, these computational tools will become increasingly accurate and powerful for rational design of high-performance ACTB antibodies with precisely engineered binding properties.

What are the implications of anti-ACTB autoantibodies as biomarkers for neurodegenerative diseases?

Recent research has revealed significant implications for anti-ACTB autoantibodies as potential biomarkers in neurodegenerative diseases, particularly amyotrophic lateral sclerosis (ALS). Studies employing serological analysis of recombinant cDNA expression libraries (SEREX) have identified elevated anti-ACTB antibody levels in ALS patients compared to both normal controls and Parkinson's disease patients (p < 0.001 and p = 0.001, respectively) . These antibodies demonstrate important clinical correlations: they are significantly higher in advanced-stage ALS patients (Stage 4 on King's ALS clinical staging) compared to early-stage patients (Stage 2) . Furthermore, anti-ACTB antibody levels negatively correlate with ALS Functional Rating Scale revised (ALSFRS-R) scores (ρ = −0.409, p = 0.001) and positively correlate with clinical disease stage (ρ = 0.355, p = 0.003) and disease duration (ρ = 0.294, p = 0.014) . These findings suggest anti-ACTB antibodies may serve as both diagnostic and prognostic biomarkers. For researchers, this underscores the importance of developing standardized, highly sensitive assays for anti-ACTB antibody detection in clinical samples. Future investigations should explore the mechanistic relationship between ACTB autoimmunity and neurodegeneration, determine whether these antibodies contribute to pathogenesis or represent secondary responses to tissue damage, and evaluate their utility in monitoring therapeutic responses in clinical trials.

What methodologies enable precise quantification of ACTB antibody binding kinetics?

Precise quantification of ACTB antibody binding kinetics requires sophisticated methodologies that capture the dynamic nature of these molecular interactions. Surface plasmon resonance (SPR) represents the gold standard, providing real-time, label-free measurements of association (k₁) and dissociation (k₋₁) rates, from which equilibrium dissociation constants (KD) can be calculated . Bio-layer interferometry (BLI) offers similar kinetic information with the advantage of higher throughput. Isothermal titration calorimetry (ITC) provides comprehensive thermodynamic profiles including enthalpy (ΔH) and entropy (ΔS) changes accompanying binding. For solution-phase measurements, microscale thermophoresis (MST) detects binding-induced changes in molecular movement within temperature gradients, requiring minimal sample volumes. When studying ACTB antibody interactions in cellular contexts, Förster resonance energy transfer (FRET) or bioluminescence resonance energy transfer (BRET) can provide spatial and temporal binding information within living cells. For high-throughput screening applications, the amplified luminescent proximity homogeneous assay (AlphaLISA) offers excellent sensitivity while allowing measurement of relative binding affinities across multiple antibody candidates . Integration of multiple complementary techniques provides the most comprehensive characterization of binding kinetics, which is essential for understanding the pharmacological and pharmacodynamic profiles of therapeutic antibodies targeting ACTB or for developing optimized research reagents.

How can Fc engineering optimize ACTB antibody performance for specific research applications?

Fc engineering represents a sophisticated approach to enhance ACTB antibody performance beyond conventional variable domain optimization. For research applications requiring extended tissue retention, introducing specific mutations in the Fc region can significantly modify FcRn binding to enhance recycling and prolong antibody half-life . When studying cytoskeletal dynamics in inflammatory environments, engineering the Fc domain to reduce interaction with activating FcγRs while maintaining binding to inhibitory FcγRIIB can minimize unwanted activation of immune effector functions that might alter cellular behavior . For applications requiring controlled receptor clustering, strategic mutations in the CH2 domain can enhance binding to FcγRIIB, which has been shown to increase agonist activity by up to 25-fold compared to wild-type antibodies in some systems . Alternatively, engineering direct Fc-Fc interactions through mutations like T437R and K248E can facilitate antibody crosslinking independent of FcR expression, providing more consistent experimental outcomes across diverse cellular systems . When absolute elimination of Fc effector functions is required, researchers might consider complete removal of the Fc domain and utilization of alternative scaffolds like single-domain antibodies (sdAbs) or designed ankyrin repeat proteins (DARPins) that maintain binding specificity without immunological complications. Each engineering approach should be selected based on the specific requirements of the research application and validated in relevant experimental systems.

What strategies address epitope masking when detecting ACTB in complex with binding partners?

Epitope masking presents a significant challenge when detecting ACTB in complex with its numerous binding partners, as these interactions can obscure antibody recognition sites. Several methodological strategies can effectively address this challenge. Epitope mapping using hydrogen-deuterium exchange mass spectrometry (HDX-MS) or X-ray crystallography enables precise identification of accessible regions on ACTB when bound to partner proteins. This information guides selection or development of antibodies targeting exposed epitopes. Computational docking and molecular dynamics simulations can predict conformational changes in ACTB upon partner binding, informing rational design of antibodies recognizing complex-specific epitopes . Proximity ligation assays (PLA) offer an alternative approach, detecting ACTB and its binding partner simultaneously while generating signal only when both proteins are in close proximity. For dynamic studies, developing conformation-specific antibodies that recognize distinct structural states of ACTB can reveal transition states during complex formation. Cross-linking mass spectrometry (XL-MS) combined with immunoprecipitation using multiple ACTB antibodies targeting different epitopes provides comprehensive mapping of accessibility in various complexes. When analyzing fixed specimens, gentle epitope retrieval techniques using citrate buffer or protease-based methods can expose hidden epitopes without disrupting important protein-protein interactions. Implementation of these strategies requires careful validation to ensure that detection accurately reflects biological reality rather than artifacts of the detection method.

What experimental design elements are critical for accurate quantification of anti-ACTB autoantibodies?

Accurate quantification of anti-ACTB autoantibodies requires meticulous experimental design with several critical elements. First, implement rigorous sample handling protocols, including standardized collection times and processing procedures, as antibody stability can be affected by freeze-thaw cycles and storage conditions. Establish appropriate reference standards using pooled positive sera with known anti-ACTB antibody concentrations to ensure inter-assay comparability. Always include comprehensive controls: healthy individuals, disease-specific controls (as seen with Parkinson's disease patients in ALS studies), and technical controls including blank, non-specific binding, and maximum signal references . When using the recommended AlphaLISA technique, carefully optimize antibody coupling to beads, incubation times, and buffer conditions to maximize signal-to-noise ratios . Implement multiple dilutions of each sample to ensure measurements fall within the linear range of the assay. Statistical analysis should include appropriate normalization methods, accounting for potential batch effects. For clinical investigations, match case and control populations for age, sex, and relevant comorbidities to minimize confounding variables. Document disease parameters using standardized clinical metrics (such as the ALSFRS-R score for ALS studies) to enable meaningful correlation analyses . Finally, validate findings using orthogonal detection methods like ELISA or immunoblotting to confirm specificity of the detected autoantibodies for ACTB rather than related cytoskeletal proteins.

How can structure-guided methods be applied to develop highly specific ACTB antibodies?

Structure-guided methods offer powerful approaches for developing highly specific ACTB antibodies with precisely engineered binding properties. Begin with high-resolution structural analysis of ACTB using X-ray crystallography or cryo-electron microscopy to identify unique surface features distinguishing it from other actin isoforms. Computational epitope mapping can then identify regions with maximum sequence and structural divergence from related proteins. Next, implement in silico screening using molecular docking to design antibody paratopes complementary to targeted epitopes . Apply energy minimization and molecular dynamics simulations to optimize binding interfaces and predict binding affinities. Directed evolution techniques like yeast surface display or phage display can be guided by structural insights to focus mutation libraries on key binding residues rather than random variation . When crystal structures are unavailable, homology models can be created using computational tools like Rosetta or FoldX, which can then inform docking protocols . For converting existing antibodies to improve specificity, alanine scanning guided by structural data can identify critical binding residues amenable to mutation, as demonstrated in the successful conversion of an antagonistic single-domain antibody to an agonist . High-throughput mutational analysis combined with binding assays can validate computationally predicted improvements. Finally, structural characterization of the antibody-ACTB complex provides feedback for iterative optimization cycles. This integrated approach combines the precision of structural biology with the efficiency of computational design to develop antibodies with exceptional specificity for ACTB.

What are the specific considerations for detecting ACTB antibodies across different biological matrices?

Detection of ACTB antibodies across different biological matrices requires matrix-specific optimization strategies to overcome unique challenges associated with each sample type. In serum or plasma, high protein concentration and abundant immunoglobulins necessitate selective capture approaches; pre-adsorption steps to remove non-specific antibodies and careful dilution series are essential for accurate quantification. For cerebrospinal fluid (CSF) analysis, particularly relevant in neurodegenerative disease studies, the lower protein concentration demands highly sensitive detection methods like AlphaLISA or Single Molecule Array (Simoa) to detect potentially smaller quantities of anti-ACTB antibodies . Tissue homogenates present challenges with cellular debris and proteases; protease inhibitors and optimized extraction buffers are crucial, along with centrifugation steps to remove particulates that might interfere with assay performance. Cell culture supernatants may contain media components that affect antibody stability or detection; media-matched calibration curves are recommended to account for matrix effects. When analyzing saliva or mucosal secretions, researchers must address variable viscosity and enzymes that might degrade antibodies; standardized collection and immediate processing protocols help maintain sample integrity. For all matrices, researchers should determine recovery rates by spiking known quantities of anti-ACTB antibodies into matrix samples and measuring detected levels compared to buffer controls. Matrix-matched standards and controls are essential for accurate quantification, as are validation studies comparing results across different detection platforms to ensure consistency regardless of the biological matrix being analyzed.

How should researchers reconcile contradictory results from different anti-ACTB antibody detection methods?

When confronted with contradictory results from different anti-ACTB antibody detection methods, researchers should implement a systematic reconciliation approach. Begin by examining the fundamental differences between the methodologies, recognizing that each technique measures different aspects of antibody-antigen interactions. AlphaLISA detects proximity-based interactions, while ELISA measures binding under static conditions, and surface plasmon resonance captures real-time kinetics . Next, evaluate each method's specific variables: antibody immobilization strategy, detection tags, buffer compositions, and sample preparation protocols can all influence results. Perform cross-platform validation using identical samples and standards to identify method-specific biases. Consider epitope accessibility variations—conformational epitopes may be preserved differently across methods, explaining apparent discrepancies. Examine sensitivity thresholds and dynamic ranges, as methods with different detection limits naturally produce divergent results with low-abundance samples. Implement dilution linearity studies across all platforms to identify potential matrix effects or hook effects specific to certain methods. Statistical approaches including Bland-Altman plots can quantify systematic differences between methods. When possible, correlate results with functional assays or clinical outcomes to determine which method provides the most biologically relevant measurements . Finally, consider using orthogonal methods in combination rather than relying on a single technique, as this complementary approach often provides more comprehensive insights than any individual method. This systematic reconciliation process transforms contradictory results from a research obstacle into an opportunity for deeper methodological understanding.

What are the emerging quality control standards for ACTB antibodies in high-impact research?

Emerging quality control standards for ACTB antibodies in high-impact research increasingly emphasize comprehensive validation strategies that ensure reproducibility and reliability. Multi-method validation has become essential, requiring demonstration of antibody performance across different applications (Western blot, immunohistochemistry, flow cytometry) with consistent results. Genetic validation using CRISB/Cas9 knockout or knockdown systems provides definitive specificity verification by demonstrating signal elimination or reduction when ACTB is depleted. Mass spectrometry validation confirms that immunoprecipitated proteins match the expected ACTB sequence and post-translational modification profile. Independent validation by multiple laboratories is increasingly required, with some high-impact journals requesting validation data from separate research groups. Lot-to-lot consistency testing has become standard practice, with researchers expected to document performance across different antibody lots using standardized positive controls. Epitope mapping and cross-reactivity profiling against all actin isoforms ensure specificity for β-actin versus related proteins. For quantitative applications, calibration against absolute standards with defined concentrations enables meaningful cross-study comparisons. Complete methodological transparency in publications has become mandatory, including comprehensive reporting of antibody clone, lot number, dilution, incubation conditions, and validation procedures. Finally, pre-registration of antibody validation protocols before conducting main experiments is emerging as a best practice to prevent selective reporting of favorable validation results. These rigorous standards collectively represent a paradigm shift toward ensuring that antibody-based research findings are robust, reproducible, and reliable.

How might novel antibody formats enhance ACTB antibody functionality in complex research applications?

Novel antibody formats are poised to dramatically enhance ACTB antibody functionality by overcoming limitations of conventional formats in complex research applications. Bispecific antibodies simultaneously targeting ACTB and interacting proteins enable direct visualization of protein complexes in situ without artifacts introduced by traditional co-immunoprecipitation. Nanobodies (VHH fragments) derived from camelid antibodies offer exceptional access to cryptic epitopes on ACTB due to their small size (~15 kDa) and extended CDR3 loops, enabling detection of conformational states inaccessible to conventional antibodies . Intrabodies—antibodies designed for intracellular expression—can track ACTB dynamics in living cells without fixation artifacts, particularly when fused to fluorescent proteins. Antibody-drug conjugates carrying photoactivatable crosslinkers can capture transient ACTB interactions upon light stimulation, freezing dynamic complexes for subsequent analysis. Switchable antibody platforms incorporating light-responsive or small molecule-responsive domains allow temporal control over ACTB detection or perturbation. DNA-conjugated antibodies enable highly multiplexed imaging of ACTB alongside dozens of other proteins using DNA-PAINT or Exchange-PAINT super-resolution microscopy. Aptamer-antibody chimeras combine the target specificity of antibodies with the programmable nature of nucleic acids for advanced detection schemes. Each of these innovative formats addresses specific research challenges, from accessing hidden epitopes to capturing dynamic interactions, ultimately expanding the experimental toolkit available for studying ACTB biology in increasingly sophisticated research contexts.

What computational approaches show promise for predicting anti-ACTB antibody cross-reactivity?

Advanced computational approaches are revolutionizing the prediction of anti-ACTB antibody cross-reactivity, enabling more targeted experimental validation and accelerating antibody development. Sequence-based epitope conservation analysis using multiple sequence alignment algorithms can identify regions of β-actin that share homology with other proteins, particularly other actin isoforms, flagging potential cross-reactivity. Structural bioinformatics approaches employing protein structure databases can detect proteins with similar three-dimensional epitope conformations despite limited sequence homology . Machine learning algorithms trained on existing cross-reactivity data are increasingly capable of predicting novel cross-reactive targets based on epitope features. Molecular dynamics simulations can model antibody binding flexibility, revealing potential for promiscuous binding to structurally similar but distinct epitopes. Fragment-based mapping tools dissect epitopes into smaller segments to identify shared motifs across the proteome. Network analysis approaches examine protein-protein interaction databases to identify ACTB binding partners that might competitively interfere with antibody binding. Improved homology modeling tools like AlphaFold2 now generate highly accurate protein structure predictions, enabling virtual screening of antibody binding against the entire proteome without requiring experimental structures . Quantum mechanical modeling of antibody-antigen binding energetics provides atomic-level insights into binding determinants that drive specificity versus promiscuity. These computational approaches collectively reduce the experimental burden of exhaustive cross-reactivity testing while providing mechanistic insights into the molecular basis of antibody specificity, ultimately leading to more selective anti-ACTB antibodies for research and clinical applications.

How does current ACTB antibody research integrate with broader trends in antibody engineering and validation?

Current ACTB antibody research exemplifies the convergence of multiple transformative trends in antibody engineering and validation. The integration of high-throughput screening with computational design represents a significant paradigm shift, with researchers increasingly employing machine learning algorithms to predict optimal binding configurations before experimental testing . This approach dramatically reduces development timelines while improving antibody performance characteristics. The emphasis on reproducibility parallels broader scientific concerns, with ACTB antibodies now subjected to rigorous multi-platform validation protocols that ensure consistent performance across experimental conditions. Structure-guided antibody engineering reflects the maturation of rational design approaches, with atomic-level understanding of ACTB epitopes informing precise paratope modifications to enhance specificity and affinity . The application of anti-ACTB autoantibodies as biomarkers in neurodegenerative diseases illustrates the expanding role of antibodies beyond traditional research tools to clinical diagnostics and prognostics . The focus on understanding binding kinetics rather than simple endpoint measurements represents a sophistication in characterization methods, providing deeper insights into antibody function in complex biological environments . Finally, the development of antibodies against specific conformational states of ACTB reflects a broader trend toward dynamic protein analysis rather than static detection. These integrated approaches collectively demonstrate how ACTB antibody research has evolved from simple reagent development to sophisticated molecular engineering that precisely controls binding properties, specificity profiles, and functional outcomes—a microcosm of the broader revolution in antibody science.

Product Science Overview

Introduction

Actin Beta (β-Actin) is a highly conserved protein that plays a crucial role in cell motility, structure, and integrity. It is ubiquitously expressed in all eukaryotic cells and is a major component of the cytoskeleton. The use of antibodies targeting β-Actin, such as Mouse Anti-Human β-Actin antibodies, is common in various scientific applications, including Western Blot, Immunohistochemistry, Immunocytochemistry, ELISA, and Flow Cytometry .

Structure and Function of β-Actin

β-Actin is one of the six isoforms of actin found in mammals. It is a cytoplasmic actin isoform, along with γ-actin, and these two isoforms are involved in controlling cell structure and motility . The protein sequence of β-actin is highly homologous to that of γ-actin, differing by only four biochemically similar amino acids . This high sequence identity means that antibodies raised against β-actin may cross-react with γ-actin.

Actin exists mainly as a fibrous polymer (F-actin) and can be depolymerized into its monomeric form (G-actin) in response to cytoskeletal reorganizing signals during processes such as cytokinesis, endocytosis, or stress .

Applications of Mouse Anti-Human β-Actin Antibodies

Mouse Anti-Human β-Actin antibodies are widely used as loading controls in Western Blot experiments due to the consistent expression of β-actin in most cell types . These antibodies are also used in Immunohistochemistry (IHC) and Immunocytochemistry (ICC) to visualize the distribution of β-actin within cells and tissues .

Specificity and Cross-Reactivity

Mouse Anti-Human β-Actin antibodies are designed to detect endogenous levels of total β-actin protein. Due to the high sequence identity between β-actin and γ-actin, these antibodies may cross-react with cytoplasmic γ-actin . However, they do not cross-react with other actin isoforms such as α-skeletal, α-cardiac, α-vascular smooth, or γ-enteric smooth muscle isoforms .

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.