MYOCD 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 orders within 1-3 business days of receipt. Delivery times may vary depending on the purchase method and location. Please consult your local distributors for specific delivery times.
Synonyms
MYCD antibody; MYCD_HUMAN antibody; Myocardin antibody; Myocd antibody
Target Names
MYOCD
Uniprot No.

Target Background

Function
Myocardin is a smooth muscle cell (SMC) and cardiac muscle cell-specific transcriptional factor that binds to the canonical single or multiple CArG boxes DNA sequence. It acts as a cofactor for serum response factor (SRF), potentially modulating SRF-target genes. Myocardin plays a crucial role in cardiogenesis, urinary bladder development, and the differentiation of the smooth muscle cell lineage (myogenesis).
Gene References Into Functions
  1. Research indicates that vascular endothelial growth factor A stimulates STAT3 activity through nitrosylation of myocardin, thereby regulating the expression of vascular smooth muscle cell differentiation markers. PMID: 28572685
  2. Conditional knockout models have revealed a critical role for myocardin during cardiac chamber maturation, and a surprising function in regulating cardiomyocyte proliferation, cell death, and possibly mitochondrial function. PMID: 28631251
  3. YAP negatively regulates the differentiation of vascular smooth muscle cells (VSMCs) derived from cardiovascular progenitor cells (CVPC) by decreasing myocardin transcription in a NKX2.5-dependent manner. PMID: 27571517
  4. Myocardin inhibits estrogen receptor alpha-mediated proliferation of human breast cancer cells by regulating MicroRNA expression. PMID: 27156566
  5. Myocardin plays a key role in the modulation of MEF2C-MYOCD-LMOD1 signaling through miR-214. PMID: 27144530
  6. Studies demonstrate that myocardin indirectly down-regulates Cx43 through its repressive action on miR-206, thereby regulating vascular smooth muscle cell phenotypic switch. PMID: 28342807
  7. Research has revealed the roles of atrogin-1 in regulating smooth muscle contractility through enhancement of myocardin ubiquitylation/degradation and its transcriptional activity. PMID: 27403897
  8. Research suggests that Ang II downregulates miR-145 to regulate Klf4 and myocardin expression in HCASMCs under high glucose conditions. Ang II plays a critical role in regulating miR-145 under hyperglycemic conditions. PMID: 26181633
  9. Human expression data indicate correlations of MYOCD with CAV1 in a majority of human tissues and in the heart, while correlation with MKL2 (MRTF-B) was observed. PMID: 26244347
  10. Inhibition of GSK-3beta reduces myocardin transcriptional activity, suggesting a role for GSK-3beta in myocardin transcriptional activity and smooth muscle differentiation. PMID: 26129946
  11. STAT3 protein regulates vascular smooth muscle cell phenotypic switch through interaction with myocardin and SRF. PMID: 26100622
  12. TNFalpha differentially regulates myocardin expression and activity. PMID: 25384061
  13. Myocardin acts as a guardian of the contractile, noninflammatory VSMC phenotype. Loss of myocardin represents a critical permissive step in the process of phenotypic transition and inflammatory activation, at the onset of vascular disease. PMID: 25614278
  14. miR135b promotes proliferation, invasion and migration of osteosarcoma cells by degrading myocardin. PMID: 25190111
  15. Early cardiac marker gene myocardin levels in peripheral blood mononuclear cells reflect severity in stable coronary artery disease. PMID: 24681789
  16. Myocardin mediates apoptosis in breast cancer through affecting maspin re-expression and epigenetic modification. PMID: 24607789
  17. TEAD1 is a novel general repressor of smooth muscle-specific gene expression by interfering with myocardin binding to SRF. PMID: 24344135
  18. ChIP analysis indicates that PDGF-BB-induced repression of Myocd gene expression is likely regulated by enhanced binding of Klf4 and Klf5, to a lesser extent, to the PRR of PrmM. PMID: 24060351
  19. Myocardin enhances the development and maturation of smooth muscle cell-like cells from human embryonic stem cells, although it may not activate the full repertoire of smooth muscle cell genes. PMID: 22937150
  20. Hypertension negates the activity of myocardin in vascular smooth muscle cells. PMID: 22843699
  21. Nuclear uPAR associates with myocardin, which is then recruited from the promoters of serum response factor target genes and undergoes proteasomal degradation. PMID: 22075245
  22. Transforming growth factor-beta1-induced transcript 1 protein, a novel marker for smooth muscle contractile phenotype, is regulated by serum response factor/myocardin protein. PMID: 21984848
  23. Vascular smooth muscle proteasomal degradation of myocardin is required for its transcriptional activity. PMID: 21506120
  24. Epigenetic inactivation of Myocardin is a frequent and tumor-specific event in nasopharyngeal carcinoma. PMID: 20848417
  25. Induction of microRNA-1 by myocardin in smooth muscle cells inhibits cell proliferation. PMID: 21051663
  26. A positive association was observed between response to methadone and two variants in the genes MYOCD and GRM6. PMID: 20560679
  27. These findings suggest a role of microRNA-1 in the negative feedback loop in the regulation of smooth muscle contractility induced by myocardin. PMID: 20458751
  28. Research shows the relative expression and activities of the major myocardin isoforms across different species. PMID: 20385216
  29. Research indicates that the downregulation of myocardin expression facilitates cell cycle progression through the reduction of p21 expression in human leimyosarcomas. PMID: 20068148
  30. The evaluation of myocardin expression is useful in distinguishing uterine smooth muscle tumors from endometrial stromal sarcomas. PMID: 19952936
  31. Myocardin was identified as a novel gene upregulated in human cervical ripening. PMID: 19883264
  32. Research reveals the first ion channel subunit as a direct target of SRF-MYOCD transactivation, providing further insight into the role of MYOCD as a master regulator of the SMC contractile phenotype. PMID: 19801679
  33. Myocardin is a critical serum response factor cofactor regulating smooth muscle cell differentiation; forced expression of myocardin activated expression of SM22 alpha, smooth muscle alpha-actin, and calponin-h1 genes in mouse ES cells. PMID: 12640126
  34. HERP1 and myocardin expression was localized to smooth muscle cells in the neointima in human coronary atherosclerotic lesions. PMID: 16151017
  35. Myocardin-MYOCD overexpression in small cerebral arteries appears to initiate independently of amyloid beta-peptide, a pathogenic pathway mediating arterial hypercontractility. PMID: 17215356
  36. Myocardin is frequently repressed during malignant transformation, contributing to a differentiation defect. PMID: 17292825
  37. The SRC3-myocardin interaction is a site of convergence for nuclear hormone receptor-mediated and VSMC-specific gene regulation. PMID: 17360478
  38. Data show that fibroblasts from human postmyocardial infarction scars acquire properties of cardiomyocytes after transduction with a recombinant myocardin gene. PMID: 17579192
  39. Forced expression of the myocardin (MyoC) gene in human ventricular scar fibroblasts leads to MyoC-dependent activation of genes that encode connexins, strongly enforcing intercellular electrical coupling. PMID: 17938287
  40. Data suggest that functional natural myocardin promoter variation might be a genetic factor contributing to inter-individual differences in the development of cardiac hypertrophy. PMID: 18028454
  41. Myocd is sufficient for the establishment of a SMC-like contractile phenotype. PMID: 18451334
  42. Foxo3a could negatively regulate myocardin expression levels through up-regulating catalase and the consequent reduction of ROS levels. PMID: 18772130
  43. A rare human sequence variant reveals myocardin autoinhibition. PMID: 18852265
  44. SRF and MYOCD function as a transcriptional switch, controlling Abeta cerebrovascular clearance and progression of AD. PMID: 19098903
  45. Analysis indicates that human retroperitoneal LMS differentiation is dependent on MYOCD amplification/overexpression, suggesting that in these well-differentiated LMS, differentiation could be a consequence of an acquired genomic alteration. PMID: 19276386
  46. Analysis of a novel mode of modulation of SM gene transcription by ERK1/2 through a direct phosphorylation of myocardin. PMID: 19776005
  47. MYOCD can discriminate among several juxtaposed CArG elements, presumably through its novel partnership with NKX3.1, to optimally transactivate the human ACTG2 promoter. PMID: 19797053

Show More

Hide All

Database Links

HGNC: 16067

OMIM: 606127

KEGG: hsa:93649

STRING: 9606.ENSP00000401678

UniGene: Hs.462257

Subcellular Location
Nucleus.
Tissue Specificity
Expressed in heart, aorta, and in smooth muscle cell-containing tissues: stomach, bladder, small intestine, colon, lung, placenta and uterus. Very faint expression in prostate and skeletal muscle.

Q&A

What are the most reliable methods for detecting endogenous MYOCD protein?

Due to the limitations of conventional antibodies, epitope tagging approaches have emerged as the most reliable methods for MYOCD detection. CRISPR-Cas9 genome editing has been successfully used to knock-in epitope tags at the C-terminal end of the myocd locus . This approach allows for unambiguous detection of the protein without relying on antibodies that may cross-react with other proteins. For researchers without access to CRISPR-modified cell lines or animals, careful validation of commercial antibodies is essential. Validation should include positive controls (cells known to express MYOCD, such as smooth muscle cells) and negative controls (cells with MYOCD knockdown or tissues known not to express the protein) . Western blotting under reducing conditions using Immunoblot Buffer Group 1 has shown specific detection of MYOCD at approximately 105 kDa when using validated antibodies .

Which cell lines are appropriate positive and negative controls for MYOCD antibody validation?

Based on the literature, several cell lines have been identified as suitable controls for MYOCD antibody validation:

Cell LineTypeMYOCD ExpressionUse as Control
MCF-7Human breast cancerPositivePositive control
RajiHuman Burkitt's lymphomaPositivePositive control
HeLaHuman cervical epithelial carcinomaPositivePositive control
C2C12Mouse myoblastVariable (can be induced)Experimental control
PAC1Smooth muscle/skeletal muscle hybridHighPositive control
H9c2Rat cardiomyocytesInducible with Ang-II or TGF-βInducible system

Validation experiments should demonstrate that the antibody detects a band of appropriate molecular weight in positive control cells that is absent or reduced in negative control cells or knockdown experiments .

How can I optimize western blot protocols for reliable MYOCD detection?

Western blot optimization for MYOCD detection requires careful attention to several parameters:

  • Sample preparation: Use PVDF membranes and reducing conditions with Immunoblot Buffer Group 1 for optimal results .

  • Antibody concentration: A concentration of 1 μg/mL of MYOCD monoclonal antibody has been shown to be effective when followed by HRP-conjugated anti-mouse IgG secondary antibody .

  • Molecular weight expectation: Look for specific bands at approximately 105-150 kDa, depending on the antibody and protein isoform .

  • Positive controls: Include lysates from cells known to express MYOCD such as smooth muscle cells or cardiomyocytes .

  • Loading controls: Use appropriate housekeeping genes such as β-actin (Actb) for normalization .

The discrepancy between the expected (150 kDa) and often observed (105 kDa) molecular weights necessitates careful interpretation of results and thorough validation of antibody specificity .

What are the recommended protocols for immunofluorescence detection of MYOCD?

For immunofluorescence detection of MYOCD, the following protocol has been effectively employed in multiple studies:

  • Fixation: Fix tissues or cells in 4% paraformaldehyde .

  • Antigen retrieval: For paraffin-embedded sections, perform antigen retrieval by boiling deparaffinized and rehydrated tissue sections in Tris-EDTA buffer (pH 9.0) for 30 minutes .

  • Blocking: Block non-specific binding using 10% donkey serum for 1 hour at room temperature .

  • Primary antibody incubation: Incubate with MYOCD antibody at optimized dilution (typically 1:200 to 1:600 based on antibody source) overnight at 4°C .

  • Secondary antibody: After washing, incubate with appropriate secondary antibodies conjugated with fluorophores (Alexa Fluor 488, 594, or 647) at 1:600 dilution for 1 hour .

  • Counterstaining: Counterstain nuclei with DAPI in mounting medium .

  • Imaging: Use confocal microscopy for high-resolution imaging of nuclear MYOCD localization .

Since MYOCD is predominantly a nuclear protein, nuclear localization of the signal should be observed in positive cells, which serves as an internal validation of specificity .

How can I distinguish between MYOCD and other myocardin family members in my experiments?

Distinguishing MYOCD from other myocardin family members requires specific approaches:

  • Epitope-tagged MYOCD: CRISPR-Cas9-mediated epitope tagging provides the most definitive approach for specific detection of MYOCD without cross-reactivity with other family members .

  • Antibody validation: Perform rigorous antibody validation in cell lines with confirmed expression or knockdown of MYOCD, including testing in multiple cell types to ensure the antibody doesn't recognize other myocardin family proteins .

  • Cell-type specificity analysis: MYOCD is predominantly expressed in smooth muscle cells and cardiomyocytes, whereas other myocardin family members have different tissue distributions. Confirm expression patterns align with expected tissue specificity .

  • Functional validation: Confirm the identity of detected proteins through functional assays, as MYOCD has specific effects on downstream gene expression, including activation of smooth muscle genes and repression of skeletal muscle genes .

  • Molecular weight verification: The true molecular weight of MYOCD is approximately 150 kDa, which is larger than other family members. Careful size analysis on western blots can help distinguish between family members .

What are the optimal ChIP protocols for studying MYOCD interactions with chromatin?

Chromatin immunoprecipitation (ChIP) for MYOCD presents challenges due to antibody specificity issues. The following approach has proven effective:

  • Epitope-tagged MYOCD: Use of CRISPR-Cas9 genome-edited cells expressing epitope-tagged MYOCD allows for highly specific chromatin immunoprecipitation using antibodies against the epitope tag rather than MYOCD itself .

  • Quantitative PCR analysis: ChIP followed by quantitative PCR is the preferred method for analyzing MYOCD binding to specific genomic regions .

  • Control regions: Include known MYOCD target genes (smooth muscle-specific genes containing CArG boxes) as positive controls, and non-CArG containing regions as negative controls .

  • Cross-validation: Confirm MYOCD binding by cross-validating with serum response factor (SRF) ChIP, as MYOCD functions as an SRF coactivator .

  • Functional validation: Correlate ChIP data with gene expression changes in response to MYOCD modulation to establish functional relevance of binding events .

How do I interpret contradictory results when studying MYOCD expression across different tissues?

Contradictory results in MYOCD expression studies can arise from several factors:

  • Antibody specificity: Poor antibody specificity is a primary cause of contradictory results. Validate antibodies thoroughly in appropriate positive and negative control tissues .

  • Molecular weight discrepancies: The actual molecular weight of MYOCD (≈150 kDa) differs from commonly reported values, leading to misinterpretation of western blot data. Ensure you're analyzing the correct molecular weight region .

  • Isoform-specific expression: MYOCD has multiple isoforms with tissue-specific expression patterns. Some antibodies may preferentially detect certain isoforms, leading to apparent contradictions in expression data .

  • Dynamic regulation: MYOCD expression can be dynamically regulated in response to stimuli. For example, angiotensin II and TGF-β can induce MYOCD expression in cardiomyocytes, and MYOCD is upregulated in OVA-challenged lungs in asthma models . Ensure experimental conditions are comparable when comparing results.

  • Technical variations: Differences in sample preparation, protein extraction methods, and detection techniques can contribute to contradictory results. Standardize protocols and use multiple detection methods (qPCR, western blot, immunostaining) when possible .

How can I effectively modulate MYOCD expression in experimental models?

Several approaches have been validated for modulating MYOCD expression:

  • siRNA knockdown: siRNA targeting MYOCD has been successfully employed in both cell culture and in vivo models. For in vivo delivery, conjugation with homing peptides has shown efficacy in targeting cardiac tissue .

  • Conditional knockout models: Doxycycline-inducible Cre-loxP systems have been used to generate tissue-specific MYOCD knockout mice. Administration of doxycycline chow (625 mg/kg) and drinking water (0.5 mg/ml) for 4 weeks effectively induces Cre-mediated Myocd deletion .

  • Adenoviral overexpression: Adenoviral vectors expressing MYOCD have been used successfully to overexpress MYOCD in cell culture models, including C2C12 and BC3H1 skeletal muscle cell lines .

  • Stable transfection: Generation of stable cell lines expressing MYOCD has been achieved in C2C12 cells, allowing for long-term studies of MYOCD effects on cell differentiation and gene expression .

  • CRISPR-Cas9 editing: Beyond epitope tagging, CRISPR-Cas9 can be used for targeted disruption or modification of the MYOCD gene .

What are the key considerations when using MYOCD antibodies in disease models?

When studying MYOCD in disease models, researchers should consider:

  • Disease-specific regulation: MYOCD expression is dynamically regulated in various pathological conditions. For example, MYOCD is upregulated in cardiac hypertrophy and fibrosis models and in asthma airway remodeling .

  • Cell type-specific expression: In disease states, MYOCD expression may be altered in specific cell populations. Use co-staining approaches with cell-type-specific markers (e.g., Acta2 for smooth muscle cells, Cdh1 for epithelial cells) to identify the precise cellular source of MYOCD .

  • Temporal dynamics: MYOCD expression may change throughout disease progression. In chronic allergic asthma models, significant increases in MYOCD mRNA occur after repeated OVA challenges .

  • Functional validation: Correlate MYOCD expression with downstream target genes and functional outcomes. For example, in cardiac disease models, monitor expression of hypertrophic markers (ANP, β-MHC) and fibrotic genes (Col 1a, Col 3a, Col 4a, TGF β, CTGF, FGF β) .

  • Intervention studies: When modulating MYOCD expression therapeutically, assess both molecular (gene expression) and functional (e.g., airway hyperresponsiveness in asthma models) endpoints .

How do I design experiments to study MYOCD's role in smooth versus skeletal muscle differentiation?

MYOCD functions as a bifunctional switch in muscle differentiation, promoting smooth muscle differentiation while inhibiting skeletal muscle differentiation. To study this dual role:

  • Cell culture models: Use appropriate cell lines including:

    • C2C12 and BC3H1 (skeletal muscle lines) for studying MYOCD's inhibitory effects on skeletal muscle differentiation

    • PAC1 cells (displaying both smooth and skeletal muscle features) for studying the balance between programs

    • Primary smooth muscle cells for smooth muscle differentiation studies

  • Gene expression analysis: Monitor key marker genes including:

    • Smooth muscle markers: smooth muscle calponin, smooth muscle myosin heavy chain (Myh11)

    • Skeletal muscle markers: Myog (myogenin), myf5, MyoD

  • Functional assays: Assess the functional consequences of MYOCD modulation:

    • For skeletal muscle: myotube formation assays

    • For smooth muscle: contractility assays

  • Molecular mechanism studies: Investigate MYOCD's interactions with:

    • SRF for smooth muscle gene activation

    • MEF2-MyoD interactions for skeletal muscle gene repression

  • DNA binding studies: Use ChIP assays to examine:

    • MYOCD-SRF binding to CArG boxes in smooth muscle genes

    • MYOCD interference with MyoD-E12 binding to E-boxes in skeletal muscle genes

How have CRISPR-based approaches improved MYOCD protein detection and functional studies?

CRISPR-Cas9 genome editing has revolutionized MYOCD research through several mechanisms:

  • Epitope tagging: CRISPR-Cas9-mediated knock-in of epitope tags at the C-terminal end of the endogenous MYOCD locus has enabled unambiguous detection of MYOCD protein at its true molecular weight (≈150 kDa) .

  • Accurate localization: Epitope tagging has permitted facile detection of nuclear MYOCD in cultured cells, confirming its primary localization in the nucleus .

  • Improved ChIP analysis: Epitope-tagged MYOCD allows for more specific chromatin immunoprecipitation, revealing authentic DNA binding patterns without the limitations of potentially cross-reactive antibodies .

  • Protein interaction studies: Tagged MYOCD facilitates more reliable protein-protein interaction studies, enabling better understanding of MYOCD's associations with transcription factors like SRF and MyoD .

  • Isoform-specific analysis: CRISPR approaches can be designed to tag or modify specific MYOCD isoforms, allowing for more precise analysis of isoform-specific functions .

These advances have corrected significant misunderstandings in the field regarding MYOCD protein size and expression patterns, highlighting the importance of accurate protein detection methods in molecular biology research .

What are the emerging applications of MYOCD antibodies in therapeutic research?

Recent research has revealed potential therapeutic applications involving MYOCD:

  • Cardiac hypertrophy and fibrosis: MYOCD inhibition using siRNA conjugated with homing peptides has shown promise in reversing cardiac hypertrophy and fibrosis in RAL (renal artery ligation) rat models. This approach resulted in significant reduction of hypertrophy markers (ANP, β-MHC) and fibrotic genes (Col 1a, Col 3a, Col 4a, TGF β, CTGF, FGF β) .

  • Asthma airway remodeling: MYOCD has been identified as a key transcriptional coactivator involved in asthma airway remodeling. Conditional knockout of MYOCD attenuated airway hyperresponsiveness in OVA-challenged mice, suggesting MYOCD as a potential therapeutic target in asthma .

  • Target validation: Antibodies against MYOCD are increasingly being used to validate the efficacy of therapeutic interventions targeting MYOCD-dependent pathways, with immunostaining and western blotting serving as key analytical methods .

  • Biomarker development: While still emerging, there is potential for using MYOCD detection as a biomarker for cardiovascular and respiratory diseases characterized by aberrant smooth muscle cell behavior .

  • Model systems: Accurate detection of MYOCD using validated antibodies or epitope tagging approaches is crucial for developing and characterizing disease models that can be used for therapeutic screening .

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.