Myosin Heavy Chain Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to Myosin Heavy Chain Monoclonal Antibodies

Myosin heavy chain (MHC) proteins are critical components of the myosin II motor complex, driving muscle contraction through actin-based motility . Monoclonal antibodies targeting MHC isoforms have become indispensable tools in muscle biology research, enabling precise detection of specific MHC variants in skeletal, cardiac, and smooth muscle tissues. These antibodies are engineered to recognize epitopes unique to distinct MHC isoforms, facilitating studies on muscle development, differentiation, and pathology .

Structure and Function of Myosin Heavy Chain

MHC is a 200–250 kDa protein forming the backbone of the myosin hexamer (two heavy chains, four light chains). It contains ATPase domains for energy transduction and actin-binding regions for contraction . Key isoforms include:

  • Skeletal muscle MHC (fast/slow): Fast MHCs (e.g., MHC IIa, IIb) enable rapid contraction, while slow MHC (MHC I) supports endurance .

  • Cardiac MHC: Expressed in atrial and ventricular myocardium, with isoform-specific antibodies like MF20 (pan-cardiac) and B1 (atrial-specific) .

  • Smooth muscle MHC: Found in vascular and visceral smooth muscle, detectable via antibodies such as BSB-17 .

Skeletal Muscle Antibodies

CloneTargetApplicationsSpecies ReactivitySource
MF20Pan-skeletal MHCImmunofluorescence, IHC, WBHuman, Mouse, Rat
AB01-2/4F2MHC10 (human)WB, IHCHuman, Mouse, Rat
BA-D5MHC IIF, IHC, WBBovine, Human, Rodent

Key Findings:

  • MF20 detects MHC in iPSC-derived cardiomyocytes and neurons, aiding studies on myotube differentiation and CTG repeat instability in myotonic dystrophy .

  • BA-D5 identifies slow MHC in myotubes, distinguishing slow/fast isoforms during muscle regeneration .

Cardiac Muscle Antibodies

CloneTargetSpecificitySpeciesSource
MF20Atrial + Ventricular MHCPan-cardiacChicken, Human
B1Atrial MHCAtrial-specificChicken

Research Insights:

  • MF20 and B1 reveal embryonic cardiac MHC expression patterns, with atrial-specific B1 binding appearing later than pan-cardiac MF20 .

  • In chicken embryonic hearts, MF20 detects MHC in presumptive ventricles at stage 10, while B1 marks atrial precursors at stage 15 .

Smooth Muscle Antibodies

CloneTargetClinical UseSpeciesSource
BSB-17Smooth MHCDistinguishes benign breast lesions from carcinomasHuman

Applications:

  • BSB-17 strongly stains myoepithelial cells in benign sclerosing lesions but is absent in invasive carcinomas, aiding diagnostic pathology .

Muscle Differentiation and Disease Modeling

  • iPSC-Derived Myocytes: MF20 immunostaining confirms MHC expression in myocytes derived from Duchenne muscular dystrophy patient iPSCs, enabling disease modeling .

  • Myotube Fusion: Antibodies like MF20 quantify myotube fusion indices in C2C12 cultures, linking differentiation kinetics to MHC expression .

Diagnostic and Pathological Uses

  • Breast Pathology: BSB-17 highlights myoepithelial layers in benign lesions, providing a diagnostic marker for distinguishing fibroadenomas from phyllodes tumors .

  • Cardiac Hypertrophy: MF20 detects MHC upregulation in hypertrophic cardiomyopathy models, correlating with contractile dysfunction .

Western Blotting and Immunofluorescence

AntibodyWB DetectionIF/ICCNotes
MF20520 kDa bandCytoplasmic MHC stainingValidated in HEK293 and C2C12 cells .
AB01-2/4F2248 kDa MHC10Tissue sectionsDetects human MHC10 in vascular smooth muscle .

Cross-Species Reactivity and Limitations

AntibodyHostReactivityLimitationsSource
MF20MouseHuman, Mouse, RatNo cross-reactivity with smooth muscle MHC
CAB4963RabbitHuman, Mouse, RatRequires optimization for IHC-P
BA-D5MouseBovine, Human, RodentLimited to slow MHC isoforms

Key Considerations:

  • Specificity: Smooth muscle antibodies (e.g., BSB-17) show no cross-reactivity with skeletal or cardiac MHC .

  • Sensitivity: Rabbit monoclonals (e.g., CAB4963) may require higher dilutions (1:200) for IHC compared to mouse IgG (1:50) .

Product Specs

Buffer
Phosphate Buffered Saline (PBS), pH 7.4, containing 0.02% sodium azide as a preservative and 50% glycerol.
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery times may vary depending on the purchase method and location. For specific delivery times, please contact your local distributor.

Q&A

What is the molecular structure and function of Myosin Heavy Chain?

Myosin Heavy Chain (MHC) is an asymmetric protein characterized by a globular head domain and an α-helical coiled-coil rod structure that facilitates the assembly of myosin into filaments. The protein functions as a major component of the contractile apparatus in muscle cells, where it interacts with actin to generate force during muscle contraction. In smooth muscle cells, MHC constitutes a critical cytoplasmic structural protein essential for cellular motility and contractile function . The globular head domain contains ATPase activity, which hydrolyzes ATP to generate the energy needed for conformational changes during the cross-bridge cycling process that drives muscle contraction .

How do Myosin Heavy Chain antibodies like MF20 and SMMS-1 differ in their specificity?

MF20 is a general Myosin Heavy Chain antibody that recognizes multiple MHC isoforms in skeletal and cardiac muscle. It typically detects MHC in the sarcoplasm of muscle tissues and is widely used to study muscle development and differentiation . In contrast, SMMS-1 is specifically designed to recognize Smooth Muscle Myosin Heavy Chain (SMHC) and selectively reacts with human visceral and vascular smooth muscle cells, as well as myoepithelial cells . This differential specificity makes SMMS-1 particularly valuable in diagnostic applications where distinguishing between smooth muscle and other muscle types is crucial, such as in distinguishing between benign sclerosing breast lesions and infiltrating carcinomas .

What are the primary applications of Myosin Heavy Chain antibodies in muscle biology research?

Myosin Heavy Chain antibodies serve multiple critical functions in muscle biology research, including:

  • Assessment of myogenic differentiation through qualitative and quantitative analysis of MHC expression in cell culture models, such as C2C12 myoblasts undergoing differentiation

  • Identification and characterization of muscle tissues in histological specimens through immunohistochemistry, enabling researchers to study muscle development, regeneration, and pathology

  • Evaluation of contractile protein expression in various physiological and pathological states, including monitoring changes in fiber-type distribution based on MHC isoform expression patterns

  • Analysis of myoepithelial cell layers in tissue specimens, which is particularly valuable in breast pathology research to distinguish between benign and malignant lesions

How do different serum concentrations impact MHC expression during myogenic differentiation?

Research demonstrates that serum concentration significantly influences MHC expression during myogenic differentiation, though it appears to operate independently of certain regulatory transcription factors. In studies with LHCN-M2 cells, increasing serum concentrations from serum-free to 0.5% and 2% resulted in progressively higher MHC area coverage (11.92 ± 0.85%, 23.10 ± 5.82%, and 43.94 ± 8.92%, respectively) . Interestingly, these changes in MHC expression occurred without corresponding alterations in the mRNA expression of myogenic regulatory factors MYOD1 and MYF5, which showed consistent patterns of expression (MYOD1 increased 6.58 ± 1.33-fold by day 5, while MYF5 was suppressed to 0.21 ± 0.11-fold) regardless of serum concentration . This suggests that while serum factors enhance myotube formation and MHC protein expression, they may act through post-transcriptional mechanisms or alternative signaling pathways rather than directly modulating primary myogenic transcription factors.

What role does Myosin Heavy Chain expression play in disease modeling using induced pluripotent stem cells?

Myosin Heavy Chain antibodies have become instrumental in disease modeling studies using induced pluripotent stem cells (iPSCs), particularly for muscular disorders. In research on myotonic dystrophy type 1 (DM1), patient-derived iPSCs can be differentiated into cardiomyocytes or transfected with MyoD1 vectors to generate myocytes for disease modeling . MHC antibodies allow researchers to confirm successful myogenic differentiation and assess the structural integrity of the contractile apparatus in these model systems. The expression patterns of MHC in DM1 patient-derived myocytes can reveal disruptions in myofibrillogenesis and sarcomere organization that characterize the disease phenotype, providing insights into pathogenic mechanisms and potential therapeutic targets . This approach enables longitudinal studies of disease progression and the evaluation of CTG repeat length instability in different cellular contexts.

How can differential expression of fast and slow Myosin Heavy Chain isoforms be leveraged in muscle fiber-type research?

Differential expression of fast (MYH1/MYH2) and slow (MYH7) Myosin Heavy Chain isoforms provides a powerful tool for investigating muscle fiber-type distribution and plasticity in various physiological and pathological conditions. Researchers can employ isoform-specific antibodies to quantitatively assess fiber-type proportions in muscle tissues and to monitor fiber-type switching in response to interventions such as exercise, electrical stimulation, or disease processes .

What are the optimal fixation and retrieval protocols for Myosin Heavy Chain detection in tissue sections?

For optimal detection of Myosin Heavy Chain in tissue sections, the fixation and retrieval protocols must be carefully selected based on the specific antibody clone and target tissue. For paraffin-embedded tissues, the following protocol has demonstrated reliable results:

  • Fixation in 10% neutral buffered formalin for 24-48 hours

  • Heat-induced epitope retrieval using an alkaline buffer (Antigen Retrieval Reagent-Basic, pH 9.0)

  • For the MF20 antibody clone specifically, incubation at 5 μg/mL for 1 hour at room temperature followed by detection using an appropriate secondary detection system such as Anti-Mouse IgG VisUCyte HRP Polymer Antibody

For frozen sections, optimal results have been achieved with:

  • Perfusion fixation with 4% paraformaldehyde

  • Cryoprotection in 30% sucrose

  • Sectioning at 8-10 μm thickness

  • Brief post-fixation in acetone for 10 minutes

  • Similar antibody incubation and detection protocols as for paraffin sections

These protocols maintain the structural integrity of the tissue while ensuring adequate epitope exposure for antibody binding, resulting in specific staining localized to the sarcoplasm of muscle fibers or myoepithelial cells depending on the target tissue.

What are the recommended protocols for assessing myogenic differentiation using MHC antibodies in cell culture models?

The assessment of myogenic differentiation using MHC antibodies in cell culture models requires a standardized protocol to ensure reproducible and quantifiable results. Based on established methodologies, the following protocol is recommended:

  • Cell Culture and Differentiation:

    • Seed myoblasts (e.g., C2C12 cells) at 70-80% confluence

    • Induce differentiation by switching from growth medium (containing 10-20% FBS) to differentiation medium (containing 0.5-2% serum or serum-free with insulin)

    • Maintain cultures for 5-10 days, with medium changes every 2-3 days

  • Immunofluorescence Staining:

    • Fix cells in 4% paraformaldehyde for 15 minutes at room temperature

    • Permeabilize with 0.2% Triton X-100 for 10 minutes

    • Block with 5% normal serum for 30 minutes

    • Incubate with Mouse Anti-Human Myosin Heavy Chain Monoclonal Antibody (e.g., MF20) at 10 μg/mL for 3 hours at room temperature

    • Apply fluorophore-conjugated secondary antibody (e.g., NorthernLights 557-conjugated Anti-Mouse IgG) and counterstain nuclei with DAPI

  • Quantitative Analysis:

    • Calculate fusion index: (number of nuclei within MHC-positive myotubes/total nuclei) × 100%

    • Determine MHC area: percentage of field of view covered by MHC-positive structures

    • Measure myotube diameter, length, and branching characteristics

This protocol allows for both qualitative assessment of myotube morphology and quantitative evaluation of differentiation efficiency under various experimental conditions.

What considerations are important when selecting secondary detection systems for MHC immunostaining?

The selection of appropriate secondary detection systems is crucial for achieving optimal signal-to-noise ratios and specificity in MHC immunostaining. Several key considerations include:

  • Host Species Compatibility: The secondary antibody must be raised against the species in which the primary MHC antibody was produced. For example, Anti-Mouse IgG secondaries are appropriate for mouse monoclonal antibodies like MF20 and SMMS-1 .

  • Application-Specific Requirements:

    • For fluorescence applications: Bright, photostable fluorophores with appropriate excitation/emission properties that avoid spectral overlap with other fluorescent markers in multiplexed experiments

    • For chromogenic detection: Enzyme conjugates (e.g., HRP) with suitable substrates (e.g., DAB) that provide adequate sensitivity and stability for the intended application

  • Signal Amplification Needs: For low-abundance targets or weakly expressed MHC isoforms, polymer-based detection systems (e.g., VisUCyte HRP Polymer) offer enhanced sensitivity compared to conventional secondary antibodies by increasing the number of enzyme molecules per binding event .

  • Background Reduction Strategies: Pre-adsorbed secondary antibodies or those with minimal cross-reactivity to endogenous immunoglobulins should be selected, particularly for tissues with high endogenous immunoglobulin content.

  • Quantification Requirements: For quantitative applications, fluorescent secondaries with linear signal response over a wide dynamic range are preferable to chromogenic detection, which may saturate at high antigen densities.

How can researchers quantitatively analyze MHC expression to assess myogenic differentiation efficiency?

Quantitative analysis of MHC expression provides crucial metrics for assessing myogenic differentiation efficiency. A comprehensive analytical approach includes the following parameters and methodologies:

These quantitative parameters should be analyzed using appropriate statistical methods, including normality testing and selection of parametric or non-parametric comparisons based on data distribution.

What are the common pitfalls in interpreting Myosin Heavy Chain immunostaining results in tissues?

Interpretation of Myosin Heavy Chain immunostaining in tissues presents several potential pitfalls that researchers must navigate carefully:

  • Cross-reactivity with Non-target Isoforms:
    Certain MHC antibodies may detect multiple isoforms, potentially leading to misinterpretation of fiber-type distribution or cell identity. Researchers should validate antibody specificity using appropriate positive and negative controls, and consider using multiple isoform-specific antibodies in parallel to confirm observations .

  • Edge Effects and Fixation Artifacts:
    Peripheral staining artifacts or uneven staining intensity across tissue sections can result from inadequate fixation or suboptimal antigen retrieval. These artifacts may be misinterpreted as biological heterogeneity. Standardized tissue processing protocols and careful examination of staining patterns across the entire section are essential .

  • Developmental and Pathological Variations:
    Myosin isoform expression changes during development and in pathological states. For example, regenerating muscle fibers may temporarily express developmental isoforms that differ from those in mature fibers. Context-specific interpretation considering the developmental stage, disease state, and regenerative status is crucial .

  • Quantification Challenges in Heterogeneous Tissues:
    In tissues with mixed fiber types or variable MHC expression, representative sampling becomes critical. Analysis of multiple fields and standardized selection criteria help avoid sampling bias. Digital image analysis with consistent thresholding parameters improves reproducibility compared to subjective visual assessment .

  • Misinterpretation of Myoepithelial Staining:
    In breast tissue analysis, distinguishing between myoepithelial cells (which express smooth muscle myosin) and myofibroblasts in stromal reactions requires careful morphological correlation. The presence or absence of myoepithelial staining has diagnostic implications in distinguishing benign from malignant lesions .

How can researchers reconcile contradictions between protein-level MHC expression and myogenic transcription factor mRNA levels?

Reconciling discrepancies between MHC protein expression and myogenic transcription factor mRNA levels requires consideration of multiple regulatory mechanisms that operate between transcription and the final protein product:

  • Temporal Dynamics and Expression Kinetics:
    Transcription factors like MYOD1 and MYF5 often exhibit transient expression patterns that precede their downstream targets. Peak MYOD1 expression may occur before maximal MHC protein accumulation, creating an apparent disconnect when analyzed at a single time point. Time-course studies capturing the complete differentiation trajectory can reveal the sequential relationship between these markers .

  • Post-transcriptional Regulation:
    Research shows that serum concentration significantly affects MHC protein expression (serum-free: 11.92%, 0.5% serum: 23.10%, 2% serum: 43.94%) without altering MYOD1 or MYF5 mRNA levels . This suggests regulation at the post-transcriptional level, including:

    • mRNA stability differences

    • Translational efficiency modulation

    • Protein stability and turnover regulation

    Analysis of additional markers in the regulatory cascade, such as microRNAs that regulate myogenic transcripts, may help explain these discrepancies.

  • Indirect Regulatory Pathways:
    Serum factors may activate alternative signaling pathways that enhance MHC expression independently of the canonical myogenic transcription factors. For example, mTOR signaling enhances protein synthesis rates without necessarily altering transcription. Investigators should consider:

    • Analyzing phosphorylation status of translation factors

    • Assessing activation of alternative signaling pathways (mTOR, MAPK)

    • Measuring global protein synthesis rates alongside specific protein accumulation

  • Methodological Considerations:
    Technical factors may contribute to apparent discrepancies:

    • Different detection sensitivities between RT-qPCR and immunostaining

    • Non-linear relationship between transcript and protein abundance

    • Subcellular compartmentalization affecting protein detection

Integrative approaches combining transcriptomics, proteomics, and functional assays provide a more complete understanding of the regulatory network governing myogenesis.

What strategies can resolve weak or inconsistent MHC staining in immunohistochemistry?

Weak or inconsistent MHC staining in immunohistochemistry can be addressed through a systematic troubleshooting approach:

  • Optimizing Antigen Retrieval:

    • For formalin-fixed tissues, compare heat-induced epitope retrieval using basic (pH 9.0) versus acidic (pH 6.0) buffers

    • Extend retrieval time incrementally (10, 20, 30 minutes) to determine optimal duration

    • For MF20 antibody specifically, heat-induced epitope retrieval using Antigen Retrieval Reagent-Basic has shown optimal results

  • Adjusting Antibody Concentration and Incubation Parameters:

    • Perform antibody titration (2.5, 5, 10 μg/mL) to determine optimal concentration

    • Compare room temperature incubation versus 4°C overnight incubation

    • For skeletal muscle tissues, 5 μg/mL concentration with 1-hour room temperature incubation has proven effective for the MF20 clone

  • Enhancing Detection Sensitivity:

    • Implement polymer-based detection systems (e.g., VisUCyte HRP Polymer) that offer signal amplification compared to conventional secondary antibodies

    • Consider tyramide signal amplification for extremely low-abundance targets

    • Optimize chromogen development time through timed monitoring of signal emergence

  • Addressing Tissue-Specific Challenges:

    • For fibrous tissues, incorporate extended permeabilization steps

    • For tissues with high background, implement additional blocking steps using bovine serum albumin (BSA) at 1-5% concentration

    • For tissues with high endogenous peroxidase activity, ensure thorough quenching with 3% hydrogen peroxide for 10-15 minutes

  • Controlling Pre-analytical Variables:

    • Standardize fixation time (24-48 hours optimal for most tissues)

    • Minimize cold ischemia time before fixation

    • Ensure consistent processing protocols across specimens to reduce batch effects

Implementation of these strategies in a systematic manner, changing one variable at a time, can help identify and resolve specific factors contributing to suboptimal MHC staining.

How can researchers address non-specific background in MHC immunofluorescence of muscle cultures?

Non-specific background in MHC immunofluorescence of muscle cultures presents a significant challenge for accurate interpretation and quantification. The following comprehensive approach can effectively mitigate this issue:

  • Optimized Blocking Protocol:

    • Implement sequential blocking with:

      • 5% normal serum (from the same species as the secondary antibody) for 30-45 minutes

      • Additional 30-minute incubation with 1% BSA and 0.1% Tween-20

    • For particularly problematic samples, include 0.1-0.3 M glycine to block free aldehyde groups from fixation

  • Antibody Optimization:

    • Titrate primary antibody concentrations (5-15 μg/mL range)

    • For MF20 clone specifically, 10 μg/mL for 3 hours at room temperature has demonstrated optimal signal-to-noise ratio in C2C12 cultures

    • Utilize pre-absorbed secondary antibodies to minimize cross-reactivity with endogenous immunoglobulins

  • Washing Procedure Refinement:

    • Implement extended washing steps (5 washes of 5 minutes each)

    • Include 0.1% Tween-20 in wash buffers to reduce hydrophobic interactions

    • Consider increasing salt concentration (up to 500 mM NaCl) in wash buffers for one wash cycle to disrupt low-affinity non-specific binding

  • Media Component Considerations:

    • Recognize that serum-containing differentiation media may contribute immunoglobulins that interact with secondary antibodies

    • For cultures differentiated in various serum concentrations (0-2%), adjust blocking conditions proportionally to serum content

    • Consider using immunoglobulin-depleted serum for culture maintenance when possible

  • Control Implementation:

    • Include isotype controls at equivalent concentrations to the primary antibody

    • Implement secondary-only controls to distinguish antibody-specific background from autofluorescence

    • Include non-differentiated myoblasts as biological negative controls for MHC expression

Through systematic implementation of these approaches, researchers can achieve clean, specific MHC immunofluorescence staining that enables accurate quantification of myogenic differentiation parameters.

What factors contribute to variability in MHC expression during in vitro myogenic differentiation experiments?

Multiple factors contribute to variability in MHC expression during in vitro myogenic differentiation, necessitating careful experimental design and standardization:

  • Serum Composition and Concentration Effects:

    • Quantitative research demonstrates that increasing serum concentrations from 0% to 2% progressively enhances MHC expression area (11.92 ± 0.85% to 43.94 ± 8.92%)

    • Different serum lots contain variable concentrations of growth factors, hormones, and cytokines that influence myogenic differentiation

    • Mitogenic components in serum can promote proliferation at the expense of differentiation, affecting the timing of MHC expression onset

  • Cell Density and Contact Inhibition:

    • Initial seeding density influences cell-cell contact frequency, a critical factor for myoblast fusion

    • Optimal density typically ranges from 70-80% confluence at differentiation induction

    • Uneven cell distribution creates regions of variable differentiation within cultures, contributing to field-to-field variation in MHC expression

  • Passage Number and Cell Line Heterogeneity:

    • Myoblast cell lines demonstrate progressive loss of differentiation potential with increasing passage number

    • Subpopulations with varying myogenic potential emerge through selective pressures during culture

    • Immortalized lines like C2C12 exhibit greater passage-dependent variability compared to primary myoblasts

  • Temporal Dynamics of Differentiation:

    • MHC expression follows distinct temporal patterns, with expression typically beginning around day 3-4 and increasing through day 10

    • Asynchronous differentiation within cultures creates mixed populations of early and late differentiating cells

    • The timing of analysis significantly impacts observed MHC expression levels and patterns

  • Microenvironmental Factors:

    • Substrate stiffness and coating composition (collagen, laminin, fibronectin) influence cell adhesion and differentiation signaling

    • Temperature and pH fluctuations affect enzymatic activities involved in myogenic regulatory pathways

    • Oxygen tension gradients in culture vessels impact metabolic activity and differentiation progression

Standardization of these variables through detailed protocol documentation, consistent reagent sourcing, and implementation of quality control metrics is essential for reducing experimental variability and enabling meaningful comparisons across studies.

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.