MYOZ2 (Myozenin-2, also known as Calsarcin-1 or FATZ-related protein 2) is a protein primarily expressed in cardiac and skeletal muscle tissues where it plays a crucial role in maintaining the structural integrity of muscle fibers . It functions as an intracellular binding protein that links Z-line proteins such as alpha-actinin, gamma-filamin, TCAP/telethonin, and LDB3/ZASP . MYOZ2 also localizes calcineurin signaling to the sarcomere and modulates this signaling pathway . Recent studies have demonstrated its role in myofibrillogenesis and its potential involvement in muscle growth and development . The significance of MYOZ2 extends to pathology, as defects in the MYOZ2 gene are associated with familial hypertrophic cardiomyopathy type 16 (CMH16) .
Based on validation data from multiple sources, MYOZ2 antibodies have been successfully employed in the following applications:
The selection of the appropriate application should be guided by your specific research question. For protein expression quantification, WB is most suitable, while IHC and ICC/IF are better for examining localization patterns within tissues or cells .
A comprehensive validation strategy for MYOZ2 antibodies should include:
Positive controls: Use tissues known to express high levels of MYOZ2, particularly heart and skeletal muscle samples .
Molecular weight verification: Confirm the detected band appears at the expected molecular weight (approximately 30 kDa, though some antibodies may detect bands between 30-35 kDa) .
Knockdown/overexpression validation: Compare antibody staining in systems with manipulated MYOZ2 expression levels, as demonstrated in studies where MYOZ2 was successfully overexpressed or silenced .
Cross-reactivity assessment: Test the antibody on tissues from different species to ensure it recognizes the intended target specifically. Most commercial MYOZ2 antibodies have been validated with human, mouse, and/or rat samples .
Peptide competition assay: Pre-incubate the antibody with the immunizing peptide to confirm binding specificity.
For effective MYOZ2 extraction and preservation:
Tissue collection: Rapidly harvest muscle tissue and either flash-freeze in liquid nitrogen or immediately process for fixation (if intended for histology) .
Protein extraction for WB:
For IHC preparation:
Cell culture preservation:
Optimization strategies differ between cardiac and skeletal muscle tissues:
Use lower antibody dilutions (1:50-1:100) initially for IHC applications
Extend antigen retrieval time to 20-25 minutes
Paraffin-embedded human heart tissue has shown good results with MYOZ2 antibodies at 1:300 dilution
Higher antibody dilutions (1:100-1:500) are often sufficient
Include longer blocking steps (1-2 hours) with 5% BSA or 10% normal serum
For myotubes, DAPI counterstaining helps visualize multinucleated structures
Primary antibody incubation: Test both overnight at 4°C and 1-2 hours at room temperature
Secondary antibody dilution: Start with 1:500 and adjust as needed
Washing steps: Increase number and duration for high-background samples
Signal amplification: Consider using biotin-streptavidin systems for low-abundance detection
Research has demonstrated that MEF2A promotes myoblast proliferation and regulates MYOZ2 during myoblast differentiation . To investigate this relationship:
ChIP assay protocol:
Reporter gene assay:
Co-expression analysis:
Functional studies:
MYOZ2 exhibits distinct functions during different stages of myogenesis . To investigate these roles:
MYOZ2 manipulation:
Proliferation assays:
Molecular markers:
Differentiation induction:
Fusion assessment:
Differentiation markers:
To investigate MYOZ2's structural functions:
Ultrastructural analysis:
Co-immunoprecipitation studies:
Immunofluorescence co-localization:
Functional disruption assays:
Generate MYOZ2 truncation mutants to identify critical domains
Transfect into myotubes and assess effects on sarcomere organization
Measure contractile function in engineered muscle tissues
Develop MYOZ2 knock-in models with domain-specific mutations
Several factors can contribute to inconsistent MYOZ2 staining:
Tissue fixation variations:
Antibody specificity issues:
Sample-specific factors:
Technical considerations:
Based on research showing DNA methylation involvement in MYOZ2 regulation :
Methylation analysis protocols:
Functional validation approaches:
Treat cells with DNA methyltransferase inhibitors (e.g., 5-azacytidine)
Measure effects on MYOZ2 expression via RT-qPCR and Western blot
Correlate methylation status with expression levels
Regulatory factor identification:
Chromatin organization studies:
Conduct ChIP assays to examine histone modifications at the MYOZ2 locus
Implement ATAC-seq to assess chromatin accessibility
Use 3C/4C/Hi-C techniques to identify long-range chromatin interactions
To investigate MYOZ2 in disease contexts:
Patient sample analysis:
Disease modeling approaches:
Generate MYOZ2 mutations associated with cardiomyopathy
Develop patient-derived iPSCs for differentiation into cardiomyocytes
Analyze structural and functional consequences in 2D and 3D models
Functional genomics strategies:
Perform transcriptomic analysis of MYOZ2-deficient or mutant cells
Study the impact of MYOZ2 on calcineurin-NFAT signaling in disease models
Examine interaction of MYOZ2 with other Z-line components in pathological states
Therapeutic investigation platforms:
Test compounds that modulate Z-line protein interactions
Explore approaches to normalize MYOZ2 expression in deficient systems
Develop gene therapy strategies for MYOZ2-related disorders
Cutting-edge imaging approaches for MYOZ2 research:
Live-cell imaging methodologies:
Generate MYOZ2-GFP fusion constructs for real-time visualization
Utilize FRAP (Fluorescence Recovery After Photobleaching) to measure MYOZ2 mobility
Implement TIRF microscopy for Z-line association dynamics
Super-resolution techniques:
Apply STORM or PALM imaging to resolve MYOZ2 nanoscale organization
Use SIM for improved visualization of MYOZ2 within the sarcomeric structure
Combine with proximity ligation assays to detect protein-protein interactions in situ
Correlative microscopy approaches:
Quantitative image analysis:
Develop automated algorithms for sarcomere organization assessment
Implement deep learning for pattern recognition in MYOZ2 distribution
Quantify co-localization coefficients with Z-line markers
Recent research has revealed MYOZ2's potential involvement in metabolic regulation :
Metabolic phenotyping approaches:
Signaling pathway analysis:
Investigate MYOZ2's impact on calcineurin-NFAT signaling
Examine phosphorylation status of downstream targets
Employ CRISPR-Cas9 screening to identify synthetic interactions
Protein-protein interaction mapping:
Perform BioID or APEX proximity labeling to identify MYOZ2 interactome
Validate key interactions through co-immunoprecipitation and FRET
Develop domain-specific mutants to disrupt specific interactions
Translational approaches:
Study metabolic changes in muscle-specific MYOZ2 knockout models
Investigate MYOZ2's role in insulin response and muscle adaptation
Examine potential therapeutic targeting of MYOZ2-mediated pathways