MYOZ2 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery time may vary depending on the purchase method and location. For specific delivery times, please consult your local distributors.
Synonyms
C4orf5 antibody; Calcineurin binding protein calsarcin 1 antibody; Calsarcin 1 antibody; Calsarcin-1 antibody; Calsarcin1 antibody; CMH16 antibody; CS 1 antibody; CS1 antibody; FATZ related protein 2 antibody; FATZ-related protein 2 antibody; Muscle specific protein antibody; MYOZ 2 antibody; MYOZ2 antibody; MYOZ2_HUMAN antibody; Myozenin-2 antibody; Myozenin2 antibody
Target Names
MYOZ2
Uniprot No.

Target Background

Function
Myozenins are intracellular binding proteins that link Z line proteins such as alpha-actinin, gamma-filamin, TCAP/telethonin, LDB3/ZASP, and localize calcineurin signaling to the sarcomere. They play a crucial role in modulating calcineurin signaling and may contribute to myofibrillogenesis.
Gene References Into Functions
  1. Myozenin may play a modifying role in hypertrophic cardiomyopathy by affecting the penetrance or degree of performance of the MYH7 gene. PMID: 28296734
  2. The cardiac phenotype in hypertrophic cardiomyopathy caused by MYOZ2 mutations might be independent of calcineurin activity in the heart. PMID: 22987565
  3. Two missense mutations, S48P substitution and I246M affecting highly conserved amino acids, were linked to hereditary Hypertrophic cardiomyopathy characterized by early onset of symptoms, pronounced cardiac hypertrophy, and cardiac arrhythmias. PMID: 17347475
  4. Mutations in MYOZ1 and MYOZ2 are at least very rare events as an underlying disease mechanism for idiopathic or familial DCM. PMID: 17434779
  5. Observational study of genotype prevalence. (HuGE Navigator) PMID: 17347475

Show More

Hide All

Database Links

HGNC: 1330

OMIM: 605602

KEGG: hsa:51778

STRING: 9606.ENSP00000306997

UniGene: Hs.732122

Involvement In Disease
Cardiomyopathy, familial hypertrophic 16 (CMH16)
Protein Families
Myozenin family
Subcellular Location
Cytoplasm, myofibril, sarcomere, Z line.
Tissue Specificity
Expressed specifically in heart and skeletal muscle.

Q&A

What is MYOZ2 and why is it important in muscle research?

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) .

Which applications are MYOZ2 antibodies most suitable for?

Based on validation data from multiple sources, MYOZ2 antibodies have been successfully employed in the following applications:

ApplicationRecommended DilutionNotes
Western Blot (WB)1:500-1:12000Most widely validated application
Immunohistochemistry (IHC-P)1:50-1:500Particularly effective in cardiac and skeletal muscle tissues
Immunocytochemistry/Immunofluorescence (ICC/IF)1:100-1:500Useful for cellular localization studies
ELISAVaries by manufacturerLess common but viable application

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 .

How should I validate MYOZ2 antibody specificity in my experimental system?

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.

What are the optimal protocols for extracting and preserving MYOZ2 in muscle samples?

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:

    • Use RIPA buffer supplemented with protease inhibitors

    • Homogenize tissue thoroughly at 4°C

    • Centrifuge at 12,000g for 15 minutes at 4°C

    • Collect supernatant and determine protein concentration

    • Store aliquots at -80°C to prevent freeze-thaw cycles

  • For IHC preparation:

    • Fix tissues in 4% paraformaldehyde

    • For paraffin embedding, suggested antigen retrieval with TE buffer pH 9.0 or alternatively, citrate buffer pH 6.0

    • For frozen sections, fix briefly (10 minutes) in cold acetone

  • Cell culture preservation:

    • For myoblasts/myotubes, fix with 4% paraformaldehyde for 15 minutes at room temperature

    • Permeabilize with 0.1% Triton X-100 for 5 minutes

How do I optimize MYOZ2 antibody staining in different muscle types?

Optimization strategies differ between cardiac and skeletal muscle tissues:

For cardiac muscle:

  • 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

For skeletal muscle:

  • 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

Optimization parameters to test:

  • 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

How can I investigate the relationship between MEF2A and MYOZ2 in myoblast differentiation?

Research has demonstrated that MEF2A promotes myoblast proliferation and regulates MYOZ2 during myoblast differentiation . To investigate this relationship:

  • ChIP assay protocol:

    • Crosslink cells with 1% formaldehyde for 10 minutes at room temperature

    • Lyse cells and sonicate chromatin to 200-500 bp fragments

    • Immunoprecipitate with anti-MEF2A antibody

    • Analyze binding to the MYOZ2 promoter region by qPCR

    • Include IgG controls and positive control regions

  • Reporter gene assay:

    • Clone the MYOZ2 proximal promoter into a luciferase reporter vector

    • Co-transfect with MEF2A expression plasmid

    • Measure luciferase activity 24-48 hours post-transfection

    • Include promoter deletion/mutation constructs to identify precise binding sites

  • Co-expression analysis:

    • Perform RT-qPCR to monitor MEF2A and MYOZ2 expression during myoblast differentiation

    • Compare with myogenic markers (MyoD, Myf5, myogenin)

    • Western blot to confirm protein level changes

    • Use immunofluorescence to visualize co-localization patterns

  • Functional studies:

    • Overexpress or knockdown MEF2A and measure effects on MYOZ2 expression

    • Assess impact on myoblast proliferation using EdU incorporation assays

    • Evaluate differentiation potential through fusion index quantification and myogenic marker expression

What methodologies should I use to study MYOZ2's role in myoblast proliferation versus differentiation?

MYOZ2 exhibits distinct functions during different stages of myogenesis . To investigate these roles:

For proliferation studies:

  • MYOZ2 manipulation:

    • Transfect myoblasts with MYOZ2 overexpression plasmid or siRNA

    • Confirm expression changes via RT-qPCR and Western blot (60,000× increase for overexpression, 0.04× for knockdown)

  • Proliferation assays:

    • Measure cell viability using CCK8 assay 24-48 hours post-transfection

    • Quantify proliferation via EdU incorporation assay

    • Analyze cell cycle distribution using flow cytometry

  • Molecular markers:

    • Assess PAX7 expression (proliferation marker)

    • Monitor MYOD levels (differentiation determining factor)

    • Investigate CDK2 protein expression (cell cycle regulator)

For differentiation studies:

  • Differentiation induction:

    • Culture myoblasts to 80-90% confluence

    • Switch to differentiation medium (2% horse serum)

    • Maintain MYOZ2 overexpression or knockdown during differentiation

  • Fusion assessment:

    • Quantify myotube formation after 3-5 days

    • Calculate fusion index (nuclei in myotubes/total nuclei)

    • Categorize myotubes by number of nuclei (2-4, 5-10, >10)

  • Differentiation markers:

    • Measure early markers (MYF5) and late markers (MYOG)

    • Perform immunofluorescence for muscle-specific proteins like myosin and desmin

How can I design experiments to examine MYOZ2's role in muscle fiber integrity and sarcomere organization?

To investigate MYOZ2's structural functions:

  • Ultrastructural analysis:

    • Process muscle samples for transmission electron microscopy (TEM)

    • Fix cells with 2.5% glutaraldehyde in 0.1M sodium cacodylate buffer

    • Post-fix with 1% osmium tetroxide and 1.5% potassium ferrocyanide

    • Measure Z-line width and sarcomere organization

  • Co-immunoprecipitation studies:

    • Lyse muscle samples in non-denaturing buffer

    • Immunoprecipitate with anti-MYOZ2 antibody

    • Probe for interaction partners (alpha-actinin, gamma-filamin, TCAP/telethonin, LDB3/ZASP)

    • Confirm reciprocal interactions

  • Immunofluorescence co-localization:

    • Double-stain muscle sections or myotubes with MYOZ2 and Z-line markers

    • Acquire high-resolution confocal images

    • Perform quantitative co-localization analysis

    • Use super-resolution microscopy for detailed structural arrangements

  • 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

Why might MYOZ2 antibody staining show inconsistent results between experiments?

Several factors can contribute to inconsistent MYOZ2 staining:

  • Tissue fixation variations:

    • Overfixation can mask epitopes

    • Underfixation may compromise tissue morphology

    • Standardize fixation time (10-24 hours for tissues, 10-15 minutes for cells)

  • Antibody specificity issues:

    • Different antibodies recognize distinct epitopes within MYOZ2

    • Confirm the immunogen region (full-length vs. fragment-specific antibodies)

    • Use antibodies validated for your specific application and species

  • Sample-specific factors:

    • MYOZ2 expression varies between muscle types and developmental stages

    • Expression changes during myoblast differentiation (decreases initially then increases)

    • Expression may be altered in disease states or after experimental interventions

  • Technical considerations:

    • Antigen retrieval method significantly impacts staining (compare TE buffer pH 9.0 vs. citrate buffer pH 6.0)

    • Secondary antibody cross-reactivity

    • Batch-to-batch variations of antibodies

How should I design experiments to study the epigenetic regulation of MYOZ2?

Based on research showing DNA methylation involvement in MYOZ2 regulation :

  • Methylation analysis protocols:

    • Use MethPrimer to predict CpG islands in the MYOZ2 promoter

    • Design bisulfite sequencing PCR (BSP) primers

    • Perform bisulfite conversion of DNA

    • Conduct Sanger sequencing to analyze methylation patterns

  • 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:

    • Investigate the role of DNMT1 in MYOZ2 regulation

    • Study lncRNAs that may influence MYOZ2 methylation (e.g., lncMYOZ2)

    • Perform knockdown/overexpression of candidate regulatory factors

  • 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

What are the best approaches for studying MYOZ2 in the context of muscle pathologies?

To investigate MYOZ2 in disease contexts:

  • Patient sample analysis:

    • Compare MYOZ2 expression in healthy vs. diseased muscle biopsies

    • Use IHC to examine localization changes in cardiomyopathy samples

    • Perform genetic screening for MYOZ2 mutations in familial cases

  • 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

How can advanced imaging techniques enhance our understanding of MYOZ2 dynamics in muscle cells?

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:

    • Integrate fluorescence and electron microscopy data

    • Use serial block-face scanning electron microscopy for 3D reconstruction

    • Implement focused ion beam-scanning electron microscopy for ultrastructural context

  • 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

What are the most promising techniques for investigating MYOZ2's role in cellular metabolism and signaling?

Recent research has revealed MYOZ2's potential involvement in metabolic regulation :

  • Metabolic phenotyping approaches:

    • Measure oxygen consumption rate (OCR) using Seahorse XF Analyzer in MYOZ2-modified cells

    • Assess mitochondrial function parameters (basal respiration, ATP production, maximum capacity)

    • Analyze glycolytic capacity through extracellular acidification rate (ECAR)

  • 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

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.