Myoglobin Antibody

Myoglobin, Mouse Anti Human
Shipped with Ice Packs
In Stock

Product Specs

Introduction
Myoglobin, a member of the globin superfamily found in skeletal and cardiac muscles, is a haemoprotein crucial for intracellular oxygen storage and facilitated oxygen diffusion between cells. This single-chain globular protein, consisting of 153 amino acids, features a heme prosthetic group (an iron-containing porphyrin) at its core, around which the remaining apoprotein folds. With 8 alpha helices and a hydrophobic core, myoglobin, with a molecular weight of 16.7 kDa, serves as the primary oxygen-carrying pigment in muscle tissues. Its oxygen binding mechanism, characterized by a hyperbolic oxygen dissociation curve often described as having "instant binding tenacity," differs from the cooperative binding observed in hemoglobin, a multimeric protein. This distinction arises from the independent nature of oxygen binding to myoglobin, unaffected by surrounding oxygen pressure. High myoglobin concentrations in the muscle cells of certain organisms contribute to their extended breath-holding capabilities. The red pigmentation of meat is attributed to myoglobin, with the iron atom's charge and the attached oxygen influencing the meat's color. Found in Type I, Type II A, and Type II B muscle fibers, myoglobin is generally considered absent in smooth muscle. Damaged muscle tissue (rhabdomyolysis) releases myoglobin, present in high concentrations within these tissues. Despite being filtered by the kidneys, the released myoglobin exhibits toxicity to renal tubular epithelium, potentially leading to acute renal failure.
Formulation
The antibody is supplied in a solution containing 1mg/ml in PBS at a pH of 7.4, with 0.1% sodium azide added as a preservative.
Storage Procedures
For short-term storage of up to 1 month, keep the antibody at a refrigerated temperature of 4°C. For longer storage durations, it is recommended to store the antibody at -20°C. Avoid repeated freeze-thaw cycles to maintain antibody stability and activity.
Applications
This antibody has undergone rigorous testing using ELISA and Western blot analysis to confirm its specificity and reactivity. However, as specific experimental conditions may vary, it is crucial to optimize the antibody concentration for each application to achieve optimal results. For Western blot analysis, a dilution range of 1:1000 is recommended as a starting point.
Synonyms
Myoglobin, MB, PVALB, MGC13548.
Purification Method
Myoglobin antibody was purified from mouse ascitic fluids by protein-G affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT6E10.
Immunogen
Anti-human Myoglobin, is derived from hybridization of mouse FO myeloma cells with spleen cells from BALB/c mice immunized with recombinant human Myoglobin amino acids 1-154 purified from E. coli.
Ig Subclass
Mouse IgG1 heavy chain and k light chain.

Q&A

What is myoglobin and why are myoglobin antibodies important in research?

Myoglobin is a cytosolic oxygen-binding hemoprotein primarily expressed in cardiac and skeletal muscle tissue, where it plays a crucial role in oxygen storage, diffusion, and transport within myocytes. The protein is essential for maintaining mitochondrial respiration during intense and prolonged muscle activity, facilitating oxygen transport from erythrocytes to mitochondria . Myoglobin antibodies are vital research tools that enable the detection, localization, and quantification of myoglobin in various experimental contexts. They allow researchers to investigate myoglobin's expression patterns in different tissues, its role in oxygen metabolism, and its relevance as a biomarker for various pathological conditions, particularly cardiac and skeletal muscle damage .

What types of myoglobin antibodies are available for research applications?

Researchers can access several types of myoglobin antibodies with different properties suitable for various experimental approaches:

  • Monoclonal antibodies: Mouse-derived monoclonal antibodies (such as A-6 and A-9) recognize specific epitopes of myoglobin with high specificity. These are available in multiple formats including non-conjugated and conjugated forms (HRP, PE, FITC, and various Alexa Fluor® conjugates) .

  • Polyclonal antibodies: Rabbit-derived polyclonal antibodies (such as 16048-1-AP) recognize multiple epitopes on the myoglobin protein, potentially offering enhanced sensitivity for certain applications .

  • Recombinant antibodies: Novel recombinant rabbit anti-myoglobin monoclonal antibodies have been developed with extraordinarily high affinity (KD = 1.21 pM) and broad species reactivity, offering advantages for specific research applications .

The selection of antibody type should be based on experimental requirements, including detection method, sensitivity needs, and species compatibility.

What applications are myoglobin antibodies validated for in research settings?

Myoglobin antibodies have been validated for multiple research applications:

ApplicationDescriptionTypical Dilution Ranges
Western Blotting (WB)Detection of denatured myoglobin in protein lysates1:2000-1:12000
Immunoprecipitation (IP)Isolation of myoglobin from complex protein mixtures0.5-4.0 μg for 1.0-3.0 mg protein lysate
Immunofluorescence (IF)Localization of myoglobin in fixed cells/tissuesVaries by antibody
Immunohistochemistry (IHC)Detection of myoglobin in tissue sections1:20-1:200
ELISAQuantitative measurement of myoglobinVaries by antibody and kit

When designing experiments, researchers should consider that optimal dilutions and conditions may vary across different antibody preparations and experimental systems .

What species reactivity should researchers consider when selecting myoglobin antibodies?

Most commercial myoglobin antibodies demonstrate cross-reactivity with myoglobin from multiple species. For example:

  • Mouse monoclonal antibodies (A-6 and A-9) typically detect myoglobin from mouse, rat, and human sources .

  • Polyclonal antibodies like 16048-1-AP have confirmed reactivity with human, mouse, and rat samples, with reported reactivity in pig samples .

  • Novel recombinant rabbit monoclonal antibodies have demonstrated broad species reactivity including mouse, rat, human, and horse myoglobin .

Researchers should carefully verify the documented species reactivity of their selected antibody and consider validating cross-reactivity when working with less common research models. Interestingly, research has shown that the antibody response to myoglobin is largely independent of the immunized species, which contributes to the wide cross-reactivity observed in many myoglobin antibodies .

How can researchers optimize myoglobin antibody performance for challenging samples?

Optimizing myoglobin antibody performance in challenging samples requires systematic approach to troubleshooting:

  • Sample preparation optimization: For muscle tissues with high myoglobin content, titrate lysis buffer compositions to ensure complete protein extraction while maintaining antibody epitopes. Consider using specialized extraction buffers containing protease inhibitors to prevent degradation of myoglobin during sample preparation.

  • Antigen retrieval methods: For fixed tissues in IHC applications, comparative testing of different antigen retrieval methods is recommended. Evidence suggests that TE buffer (pH 9.0) may provide optimal results for some antibodies, while citrate buffer (pH 6.0) works better for others .

  • Signal amplification strategies: For low-abundance samples, consider employing signal amplification techniques such as tyramide signal amplification (TSA) or polymer-based detection systems rather than simply increasing antibody concentration, which can lead to higher background.

  • Cross-linking fixatives consideration: When working with formalin-fixed samples, recognize that excessive fixation can mask epitopes. Systematic testing of fixation times or alternative gentle fixatives may improve detection in difficult samples.

  • Blocking optimization: For tissues with high background, systematic comparison of different blocking agents (BSA, normal serum, commercial blockers) at various concentrations can significantly improve signal-to-noise ratio.

What methodological approaches best address potential cross-reactivity issues with myoglobin antibodies?

Cross-reactivity can complicate the interpretation of myoglobin antibody results, particularly when working with complex tissue samples containing multiple muscle proteins. Researchers should implement these methodological approaches:

  • Validation with knockout/knockdown controls: Where possible, validate antibody specificity using samples where myoglobin expression has been genetically eliminated or reduced.

  • Pre-absorption controls: Perform pre-absorption experiments by incubating the antibody with purified myoglobin protein before application to samples. Disappearance of signal confirms specificity.

  • Multi-antibody validation: Use multiple antibodies targeting different epitopes of myoglobin to confirm findings, particularly for novel or controversial results.

  • Mass spectrometry correlation: For critical findings, confirm antibody-based detection with orthogonal techniques such as mass spectrometry identification of immunoprecipitated proteins.

  • Tissue-specific expression patterns: Leverage known expression patterns of myoglobin (high in cardiac and skeletal muscle, low/absent in most other tissues) as internal controls within experimental designs .

Research has demonstrated that although myoglobin antibodies show considerable cross-species reactivity, their specificity for myoglobin versus other heme-containing proteins remains high when properly validated .

How can researchers accurately interpret myoglobin detection data in the context of tissue damage biomarker studies?

Interpreting myoglobin data in tissue damage contexts requires consideration of several important factors:

  • Temporal dynamics: Myoglobin is released rapidly into circulation following muscle damage, appearing earlier than other cardiac markers like troponins. In acute myocardial infarction, levels can increase from normal ranges (6-85 ng/ml) to above 250 ng/ml . Researchers should design sampling timepoints accordingly to capture these dynamics.

  • Specificity considerations: Myoglobin lacks cardiac-specific expression, being present in both cardiac and skeletal muscles. Results must be interpreted alongside skeletal muscle damage markers and other cardiac-specific biomarkers (e.g., troponin I) to establish cardiac specificity .

  • Confounding conditions: Renal dysfunction can elevate myoglobin levels independently of muscle damage by reducing clearance. Researchers should measure and account for renal function parameters in biomarker studies .

  • Standardization challenges: Different antibodies and detection methods may yield different absolute values. Researchers should establish method-specific reference ranges and focus on relative changes rather than absolute values when comparing across studies.

  • Statistical analysis approaches: The relationship between myoglobin levels and extent of damage is not always linear. Consider non-linear modeling approaches (such as Logistic function fitting) when analyzing dose-response relationships in detection systems .

What are the key considerations when designing biosensing platforms using myoglobin antibodies?

Designing effective biosensing platforms requires careful attention to several technical aspects:

  • Surface chemistry optimization: The functionalization protocol significantly impacts antibody orientation and activity. Research indicates that APTMS (3-aminopropyltrimethoxysilane) surface modification followed by EDC/NHS coupling chemistry provides effective antibody immobilization while maintaining binding activity .

  • Detection limit enhancement: For achieving clinically relevant detection limits (in the ng/ml range), consider optical techniques utilizing bound states in the continuum (BIC) which have demonstrated enhanced sensitivity for myoglobin detection .

  • Non-specific binding minimization: Implementation of effective blocking strategies and optimized washing protocols is critical, especially for complex biological samples like serum or plasma.

  • Validation across concentration ranges: Comprehensive characterization across physiologically relevant myoglobin concentrations is necessary, spanning from normal levels (6-85 ng/ml) to pathological levels (>250 ng/ml) .

  • Sample matrix effects: Different biological matrices (buffer, serum, plasma) can significantly affect antibody-antigen interactions. Validation should be performed in the intended sample matrix rather than idealized buffer systems alone.

What controls are essential when working with myoglobin antibodies in research applications?

Implementing appropriate controls is crucial for ensuring reliable and interpretable results:

  • Positive controls: Include samples known to express myoglobin at high levels, such as heart or skeletal muscle tissue lysates. Published data confirms strong myoglobin expression in mouse heart tissue, mouse skeletal muscle, rat heart, and rat skeletal muscle .

  • Negative controls: Include tissues known not to express myoglobin (e.g., liver, kidney) to confirm specificity. For IHC/IF, include primary antibody omission controls on positive tissues.

  • Isotype controls: For monoclonal antibodies (particularly in flow cytometry), include matched isotype controls (e.g., mouse IgG1 kappa for A-6 and A-9 antibodies) to distinguish specific binding from Fc receptor interactions .

  • Loading controls: For quantitative western blotting, include appropriate loading controls that reflect the tissue type being analyzed.

  • Antibody validation controls: When possible, include antibody validation using recombinant myoglobin protein, competitive blocking with purified antigen, or myoglobin-deficient samples.

  • Cross-species validation: When applying antibodies to species not explicitly validated by the manufacturer, include step-wise validation showing expected tissue expression patterns and molecular weight confirmation.

What methodological considerations are important for quantitative analysis of myoglobin using antibody-based techniques?

For accurate quantitative analysis of myoglobin levels:

  • Standard curve design: When using ELISA, construct standard curves using purified myoglobin covering the full range of expected concentrations (normal range 6-85 ng/ml to pathological levels >250 ng/ml) .

  • Linear range determination: Establish the linear range of detection for each antibody and application, and ensure that experimental samples fall within this range. When necessary, perform sample dilutions to bring concentrations into the linear detection range.

  • Reference standard inclusion: Include reference standards across multiple experimental runs to normalize inter-assay variability, particularly for longitudinal studies or comparisons across different experimental batches.

  • Signal normalization strategies: For western blotting, normalize myoglobin signals to established loading controls appropriate for the tissue type being analyzed. For muscle tissues, consider sarcomeric proteins as appropriate normalizers.

  • Quantitative imaging parameters: For IF/IHC quantification, standardize image acquisition parameters (exposure time, gain, offset) and conduct analysis using calibrated intensity measurements rather than subjective scoring.

  • Statistical approach selection: Apply appropriate statistical methods based on data distribution. For biomarker studies with non-linear relationships, consider Logistic function fitting for dose-response analysis .

How should researchers address reproducibility challenges when working with myoglobin antibodies across different experimental systems?

Ensuring reproducibility requires systematic approaches:

  • Detailed methodology documentation: Record complete antibody information (catalog number, lot number, clone for monoclonals) and all experimental conditions including incubation times, temperatures, buffer compositions, and blocking reagents.

  • Antibody validation across lots: When switching antibody lots, perform validation experiments comparing the new lot to the previously used lot under identical conditions before proceeding with critical experiments.

  • Protocol standardization: Develop and follow standardized protocols, particularly for critical steps like tissue fixation, antigen retrieval, blocking, and washing conditions.

  • Inter-laboratory validation: For multi-center studies, implement centralized antibody validation or exchange of positive control samples to confirm consistent detection across different laboratory settings.

  • Environmental variable control: Monitor and control laboratory environmental factors (temperature, humidity) that can affect antibody performance, particularly for sensitive applications like IHC and IF.

  • Reporting standards adherence: Follow field-specific reporting guidelines such as ARRIVE for animal studies or MIQE for qPCR experiments involving myoglobin detection, ensuring all critical parameters are documented.

How can myoglobin antibodies be effectively utilized in multiplex detection systems?

Multiplex detection systems allow simultaneous measurement of myoglobin alongside other biomarkers:

  • Spectral compatibility planning: For fluorescence-based multiplex systems, select compatible fluorophore conjugates that minimize spectral overlap. Consider the range of available myoglobin antibody conjugates (FITC, PE, various Alexa Fluor® conjugates) .

  • Antibody cross-reactivity testing: Validate that antibody combinations do not exhibit cross-reactivity or steric hindrance when used simultaneously. This is particularly important when combining antibodies raised in the same species.

  • Sequential immunodetection protocols: For challenging multiplexing scenarios, implement sequential detection protocols with careful stripping or blocking steps between rounds of detection.

  • Imaging parameters optimization: For multiplex IF/IHC, systematically optimize exposure settings for each channel to ensure balanced signal capture across all targets without bleed-through.

  • Data normalization strategies: Develop consistent normalization approaches for quantitative multiplex data, accounting for potential differences in antibody affinities and detection efficiencies across different targets.

Recent developments in recombinant myoglobin antibodies with extremely high affinity (KD = 1.21 pM) offer enhanced performance in multiplex systems due to their specificity and sensitivity at low concentrations .

What approaches can improve myoglobin antibody specificity in diagnostic applications?

Improving specificity for diagnostic applications requires specialized approaches:

  • Epitope selection optimization: Targeting unique epitopes of myoglobin that are not conserved in other globin proteins can enhance specificity. Antibodies raised against synthetic peptides representing unique myoglobin regions can offer improved specificity over those raised against the whole protein.

  • Affinity maturation techniques: Advanced recombinant antibody technology allows for affinity maturation to enhance both specificity and sensitivity, as demonstrated in the development of recombinant rabbit anti-myoglobin monoclonal antibodies .

  • Combinatorial detection strategies: Implement multi-antibody approaches targeting different epitopes of myoglobin, requiring concordant results for positive determination.

  • Machine learning integration: Apply machine learning algorithms to analyze complex antibody binding patterns, potentially distinguishing subtle differences between myoglobin forms or sources (cardiac versus skeletal).

  • Competitive binding assays: Employ competitive binding assays with epitope-specific competitors to enhance discrimination between myoglobin and structurally similar proteins.

Recent research has demonstrated that novel recombinant rabbit anti-myoglobin monoclonal antibodies exhibit excellent tissue specificity for skeletal muscle and myocardium, making them valuable tools for diagnostic applications focused on rhabdomyolysis-associated acute kidney injury .

How can researchers effectively utilize myoglobin antibodies for studying non-traditional myoglobin expression patterns?

While traditionally associated with muscle tissues, recent studies indicate myoglobin expression in non-muscle tissues as well . Research approaches for these investigations include:

  • Sensitivity enhancement strategies: When investigating tissues with low myoglobin expression, employ signal amplification techniques like tyramide signal amplification or polymer-based detection systems to enhance detection sensitivity.

  • Multiple antibody validation: Confirm unexpected expression patterns using multiple antibodies targeting different epitopes to rule out non-specific binding artifacts.

  • Complementary RNA detection: Correlate protein detection with RNA-level analysis (RT-PCR, RNA-seq, or in situ hybridization) to confirm expression at the transcriptional level.

  • Single-cell analysis techniques: Implement single-cell approaches to identify specific cell populations expressing myoglobin within heterogeneous tissues, distinguishing true expression from contamination.

  • Functional correlation studies: Connect antibody-based detection with functional studies (e.g., oxygen consumption measurements) to establish physiological relevance of myoglobin expression in non-traditional tissues.

These approaches allow researchers to investigate emerging hypotheses regarding myoglobin's potential roles outside of traditional muscle contexts, expanding our understanding of this protein's biological significance.

Product Science Overview

Introduction

Myoglobin is a crucial protein found in muscle tissues, responsible for the storage and transport of oxygen within muscle cells. It is a single-chain globular protein with a molecular weight of approximately 17 kDa and contains a heme prosthetic group that binds oxygen. Myoglobin is particularly abundant in the muscles of vertebrates, including humans, where it plays a vital role in maintaining oxygen supply during muscle contraction.

Myoglobin Structure and Function

Myoglobin belongs to the globin superfamily and shares structural similarities with hemoglobin, the oxygen-carrying protein in red blood cells. However, unlike hemoglobin, which is a tetramer, myoglobin is a monomer. The heme group in myoglobin consists of an iron ion (Fe2+) coordinated to a porphyrin ring, which allows it to bind oxygen molecules reversibly.

The primary function of myoglobin is to facilitate the diffusion of oxygen from the bloodstream to the mitochondria within muscle cells. This process is essential for aerobic respiration and energy production, especially during periods of intense physical activity. Additionally, myoglobin serves as an oxygen reservoir, providing a buffer against oxygen deprivation in muscle tissues.

Myoglobin as a Biomarker

Myoglobin is released into the bloodstream following muscle injury or damage, making it a sensitive biomarker for muscle-related conditions. Elevated levels of myoglobin in the blood can indicate muscle trauma, myocardial infarction (heart attack), or other forms of muscle damage. Due to its rapid release and clearance from the bloodstream, myoglobin is often used in conjunction with other biomarkers, such as troponin, to diagnose and monitor cardiac events.

Mouse Anti-Human Myoglobin Antibodies

Mouse anti-human myoglobin antibodies are monoclonal antibodies specifically designed to recognize and bind to human myoglobin. These antibodies are produced by immunizing mice with human myoglobin, followed by the isolation and cloning of myoglobin-specific B cells. The resulting monoclonal antibodies are highly specific and can be used in various research and diagnostic applications.

Applications of Mouse Anti-Human Myoglobin Antibodies

Mouse anti-human myoglobin antibodies are widely used in scientific research and clinical diagnostics. Some common applications include:

  • Immunohistochemistry (IHC): These antibodies can be used to detect myoglobin in tissue sections, allowing researchers to study its distribution and expression in different muscle types.
  • Western Blotting: Mouse anti-human myoglobin antibodies are employed to identify and quantify myoglobin in protein samples extracted from tissues or cells.
  • Enzyme-Linked Immunosorbent Assay (ELISA): These antibodies are used in ELISA assays to measure myoglobin levels in biological fluids, such as blood or urine, for diagnostic purposes.
  • Immunocytochemistry (ICC): Researchers use these antibodies to visualize myoglobin within cultured cells, providing insights into its cellular localization and function.
  • Immunoprecipitation: Mouse anti-human myoglobin antibodies can be used to isolate myoglobin from complex protein mixtures, facilitating further biochemical analysis.

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 2025 TheBiotek. All Rights Reserved.