MYBPC3 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
We typically dispatch products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributors.
Synonyms
C protein cardiac muscle isoform antibody; C-protein antibody; cardiac muscle isoform antibody; Cardiac MyBP C antibody; Cardiac MyBP-C antibody; Cardiac myosin binding protein C antibody; cardiac-type antibody; CMH4 antibody; FHC antibody; MYBP C antibody; MYBPC antibody; MYBPC3 antibody; Myosin binding protein C cardiac antibody; Myosin binding protein C cardiac-type antibody; Myosin-binding protein C antibody; myosin-binding protein C cardiac type antibody; MYPC3_HUMAN antibody
Target Names
MYBPC3
Uniprot No.

Target Background

Function
MYBPC3 Antibody targets a thick filament-associated protein situated in the crossbridge region of vertebrate striated muscle A-bands. In vitro, it interacts with myosin heavy chain (MHC), F-actin, and native thin filaments. Furthermore, it modulates the activity of actin-activated myosin ATPase. MYBPC3 potentially influences muscle contraction or may play a more structural role.
Gene References Into Functions
  1. Among 52 hypertrophic cardiomyopathy patients, 11 (21.2%) exhibited MYBPC3 variants. PMID: 29386531
  2. MYBPC3 does not appear to be involved in anthracycline-induced cardiotoxicity. PMID: 29716714
  3. A mutation in MYBPC3 has been identified as a causative factor for Restrictive Cardiomyopathy. PMID: 27339502
  4. Pathogenic gene mutations in LMNA and MYBPC3 alter RNA splicing and may contribute to heart disease. PMID: 28679633
  5. MYBPC3 mutation carriers displayed a high incidence of ventricular arrhythmia and syncope. The absence of a family history of sudden death (SD) and a previous history of syncope are valuable prognostic indicators in patients with hypertrophic cardiomyopathy. MYH7 and MYBPC3 mutations did not significantly impact prognosis compared to non-carriers. Patients with the MYBPC3 mutation should be closely monitored for the potential risk of SD. PMID: 27885498
  6. Replacement Fibrosis was most prevalent in hearts with a MYBPC3 mutation that initially presented with Hypertrophic Cardiomyopathy. PMID: 28365402
  7. Mutations in the gene MYBPC3, which encodes cardiac myosin-binding protein-C (cMyBP-C), a multi-domain protein, are the most frequent cause of hypertrophic cardiomyopathy. This molecular insight suggests that critical HCM-causing mutations may significantly alter the native affinity required for the assembly of the domains in cMyBP-C, which is essential for normal cardiac function. PMID: 27267291
  8. Five out of 19 patients (26.3%) exhibited either a pathogenic variant or a likely pathogenic variant in MYBPC3 (n=1), MYH7 (n=1), RYR2 (n=2), or TNNT2 (n=1). All five variants were missense variants previously reported in patients with channelopathies or cardiomyopathies. PMID: 28202948
  9. Double heterozygotes for mutations in DSP and MYBPC3 presented with a variable clinical presentation of arrhythmogenic cardiomyopathy and hypertrophic cardiomyopathy. PMID: 28699631
  10. Data indicates that MYBPC3 mutations are the primary cause of hypertrophic cardiomyopathy (HCM) and are phenotypically indistinguishable from HCM caused by MYH7 mutations. PMID: 29121657
  11. In children and young adults, a 2-parameter 12-lead A-ECG score is retrospectively significantly more sensitive and specific than pooled, age-specific conventional ECG criteria for detecting MYBPC3-HCM and distinguishing such patients from healthy controls, including endurance-trained athletes. PMID: 27061026
  12. Mutations are associated with a reduced super-relaxed state in hypertrophic cardiomyopathy. PMID: 28658286
  13. Studies suggest that CACNB2 is a potential candidate hypertrophy-modifying gene contributing to disease variability in MYBPC3-associated familial hypertrophic cardiomyopathy. PMID: 28614222
  14. Researchers demonstrate myosin tail (S2)-dependent functional regulation of actin-activated human beta-cardiac myosin ATPase. Additionally, they show that both S2 and MyBP-C bind to S1 and that phosphorylation of either S1 or MyBP-C weakens these interactions. PMID: 28481356
  15. Research supports that mutations in MYH7 and MYBPC3 should be the primary focus of molecular genetic analysis in HCM, and that mutations in TNNT2 have a low prevalence in the Brazilian population. All detected mutations were missense mutations, with two mutations in MYH7 being previously unreported. PMID: 27737317
  16. These findings highlight the crucial role of MYBPC3 during sarcomere assembly in cardiac myocyte differentiation and suggest developmental influences of MYBPC3 truncating mutations on the mature hypertrophic phenotype. PMID: 27620334
  17. The p.Pro108Alafs*9 mutation is associated with HCM, high penetrance, and disease onset in middle age. PMID: 28029522
  18. Studies reveal a lack of phenotypic differences between MYH7- and MYBPC3-associated hypertrophic cardiomyopathy when assessed by cardiac magnetic resonance imaging. PMID: 28193612
  19. MYBPC3 and MYH7 were the most commonly mutated genes, accounting for 27% of all Hypertrophic Cardiomyopathy patients and 83% of the putative mutations in the main sarcomeric genes. PMID: 27574918
  20. MYBPC3 gene mutation is associated with Early-Onset Hypertrophic Cardiomyopathy. PMID: 27483260
  21. Research demonstrates that the MYBPC3 25-bp deletion polymorphism was significantly associated with an elevated risk of left ventricular dysfunction (LVD), while TTN 18 bp I/D, TNNT2 5 bp I/D, and myospryn K2906N polymorphisms did not exhibit any significant association with LVD. PMID: 27350668
  22. Researchers report a patient presenting with a complex phenotype consisting of severe, adult-onset, dilated cardiomyopathy, hearing loss, and developmental delay, in which exome sequencing revealed two genetic variants inherited from a healthy mother: a variant, in MYBPC3, that is associated with hereditary cardiomyopathy. PMID: 27173948
  23. Five out of 102 (4.9%) athletes carried mutations: a heterozygous MYH7 Glu935Lys mutation, a heterozygous MYBPC3 Arg160Trp mutation, and another heterozygous MYBPC3 Thr1046Met mutation, all of which have been reported as HCM-associated mutations. PMID: 26178432
  24. The phosphorylation pattern of sMyBP-C is differentially regulated in response to age and disease, suggesting that phosphorylation plays significant roles in these processes. PMID: 26287277
  25. MYBPC3 gene mutations play a significant role in Hypertrophic cardiomyopathy (HCM) disease and can be utilized for pre-symptomatic diagnosis of at-risk family members of affected individuals. PMID: 27348999
  26. Atrial fibrillation occurred in 74 patients with hypertrophic cardiomyopathy (31%), but with no difference among genotype groups (31% in MYBPC3, 37% in MYH7, and 18% in other genotypes, p = 0.15). PMID: 26869393
  27. The detection of MYBPC3 mutation, particularly the PTC mutation and double-mutation, can serve as a molecular marker for clinical risk stratification of HCM. PMID: 26090888
  28. Studies demonstrate that MYBPC3 gene mutations, revealed by next-generation sequencing, were associated with familial and sporadic restrictive cardiomyopathy phenotypes in patients. PMID: 26163040
  29. Data suggest that homozygous or compound heterozygous truncating pathogenic myosin binding protein C (MYBPC3) mutations cause severe neonatal cardiomyopathy with features of left ventricular noncompaction and septal defects. PMID: 25335496
  30. Case Report: double cMyBP-C mutation in a patient with end-stage hypertrophic cardiomyopathy. PMID: 25971843
  31. Mutations in the MYBPC3 and CASQ2 genes and six combinations between loci in the MYBPC3, MYH7, and CASQ2 genes were responsible for cardiomyopathy risk in a studied cohort. PMID: 25892673
  32. While females with MYBPC3 mutations exhibited a later onset of hypertrophic cardiomyopathy, female patients were more symptomatic at diagnosis and experienced more frequent heart failure events once hypertrophy developed. PMID: 25123604
  33. A founder MYBPC3 mutation, arising over 550 years ago, is the predominant cause of hypertrophic cardiomyopathy in Iceland. PMID: 25740977
  34. Mutations in MYBPC3 are associated with cardiomyopathy. [Review] PMID: 26358504
  35. Characterization of novel splicing mutations in cardiomyopathy genes MYBPC3 and TNNT2. PMID: 25849606
  36. Gene-specific severity of cardiac abnormalities may underlie differences in disease onset and suggests that early initiation of metabolic treatment may be beneficial, particularly in myosin heavy chain (MYH7) mutation carriers. PMID: 24835277
  37. Results show that the N-terminal region of MyBP-C stabilizes the ON state of thin filaments and the OFF state of thick filaments, leading to a novel hypothesis for the physiological role of MyBP-C in regulating cardiac contractility. PMID: 25512492
  38. This review summarizes evidence that phosphorylation of MyBP-C is a key regulator of cardiac force and contraction. PMID: 25552695
  39. At the cMyBP-C expression levels observed in hypertrophic cardiomyopathy patients, cross-bridge kinetics are preserved, indicating that the depressed maximal force development is not explained by perturbation of cross-bridge kinetics. PMID: 24186209
  40. A founder MYBPC3 mutation, originating over 550 years ago, is the primary cause of hypertrophic cardiomyopathy in Iceland. PMID: 25078086
  41. Research indicates that MYBPC3 Val762Asp may be associated with unfavorable hypertrophic cardiomyopathy phenotypes. PMID: 25281569
  42. A comprehensive review of MYBPC3 mutations associated with hypertrophic cardiomyopathy. PMID: 24337823
  43. The structural features of the R502W mutation of myosin bindin protein C are described and discussed. PMID: 25058872
  44. This review explores the known roles of cMyBP-C as a regulator of contraction and its involvement in HCM. [review] PMID: 24240729
  45. cMyBP-C is a key regulator of cardiac contractility. Although mutations in the gene encoding cMyBP-C are a leading cause of hypertrophic cardiomyopathy, limited understanding exists regarding the molecular mechanisms underlying the disease process. [review] PMID: 24327208
  46. Alterations in the ability of cMyBP-C to bind cardiac actin-modified filaments may contribute to the development of the disease. PMID: 24736382
  47. The G263X mutation of MYBPC3 was identified in 7 patients with hypertrophic cardiomyopathy in Asturias, Spain. PMID: 23870641
  48. Hypertrophic cardiomyopathy patients with MYBPC3 mutations exhibit a distinct miRNA expression profile. PMID: 24083979
  49. The N-terminal fragment of cardiac myosin-binding protein C (cMyBP-C) impairs myofilament function in human myocardium. PMID: 24509847
  50. cMyBP-C is released into the bloodstream rapidly after cardiac damage, making it a potential biomarker for the onset of myocardial infarction. PMID: 24337456

Show More

Hide All

Database Links

HGNC: 7551

OMIM: 115197

KEGG: hsa:4607

STRING: 9606.ENSP00000442795

UniGene: Hs.524906

Involvement In Disease
Cardiomyopathy, familial hypertrophic 4 (CMH4); Cardiomyopathy, dilated 1MM (CMD1MM); Left ventricular non-compaction 10 (LVNC10)
Protein Families
Immunoglobulin superfamily, MyBP family

Q&A

What are the primary applications for MYBPC3 antibodies?

MYBPC3 antibodies are versatile tools that can be utilized in multiple experimental techniques, with varying recommended dilutions for optimal results:

ApplicationCommon Dilution Ranges
Western Blot (WB)1:500-1:3000
Immunohistochemistry (IHC)1:50-1:500
Immunofluorescence (IF-P)1:50-1:500
Immunofluorescence (IF/ICC)1:50-1:500
ELISAApplication-specific

These antibodies have been validated across multiple tissue types, with positive Western blot detection confirmed in mouse, rat, and human heart tissue samples . It is recommended to titrate antibodies in each specific testing system to obtain optimal results, as requirements may be sample-dependent .

What species reactivity can be expected with commercially available MYBPC3 antibodies?

Most commercial MYBPC3 antibodies demonstrate reactivity with samples from multiple species:

  • Human samples: Validated in heart tissue and relevant cell lines

  • Mouse samples: Particularly heart tissue and specialized cardiomyocyte models

  • Rat samples: Primarily heart tissue samples

When selecting a MYBPC3 antibody, review both tested reactivity (experimentally confirmed by the manufacturer) and cited reactivity (reported in published literature) to ensure compatibility with your experimental model . The species reactivity is determined by the conservation of the epitope sequence across species and the specific immunogen used to generate the antibody.

What is the optimal storage protocol for MYBPC3 antibodies?

For maximum shelf life and maintained reactivity:

  • Store unopened antibodies at -20°C

  • Most formulations remain stable for at least one year after shipment when stored properly

  • For many MYBPC3 antibodies, aliquoting is unnecessary for -20°C storage

  • Most commercial preparations are supplied in PBS with 0.02% sodium azide and 50% glycerol at pH 7.3

  • Some smaller antibody volumes (e.g., 20μl sizes) may contain 0.1% BSA as a stabilizer

Avoid repeated freeze-thaw cycles as they can compromise antibody performance. Always follow manufacturer-specific recommendations, as formulations may vary between suppliers.

How should tissue samples be prepared for optimal MYBPC3 detection?

For immunohistochemistry applications:

  • For mouse heart tissue, antigen retrieval with TE buffer at pH 9.0 is typically recommended

  • Alternatively, antigen retrieval may be performed with citrate buffer at pH 6.0

  • For MYBPC3 detection in fixed tissue samples, a 1:50-1:500 dilution range is typically effective

When working with formalin-fixed, paraffin-embedded samples, thorough deparaffinization and hydration are required before antigen retrieval steps to ensure accessibility of the epitope.

How can MYBPC3 antibodies be used to investigate truncating mutations associated with hypertrophic cardiomyopathy?

MYBPC3 mutations are the most common genetic cause of hypertrophic cardiomyopathy (HCM), with truncating variants accounting for approximately 91% of pathogenic MYBPC3 variants . When designing experiments to investigate these mutations:

  • Utilize antibodies targeting different epitopes (N-terminal vs. C-terminal) to distinguish between truncated and full-length proteins

  • Implement comparative analysis between patient-derived samples and controls

  • Consider combinatorial approaches with genetic testing validation

Research has shown that truncating variants are evenly dispersed throughout the gene, and hypertrophy severity and outcomes are not associated with variant location . This suggests that experimental designs should focus on detecting the presence of truncation rather than its specific location.

What methodological considerations are important when using MYBPC3 antibodies in iPSC-derived cardiomyocyte models?

When employing MYBPC3 antibodies in induced pluripotent stem cell (iPSC) cardiomyocyte models:

  • Validation steps should include immunofluorescence confirmation in both heterozygous and homozygous MYBPC3 knockout models

  • A homozygous promoter deletion iPSC line can serve as a negative control, as it completely lacks MyBP-C expression by immunofluorescence

  • When analyzing frameshift mutations, consider that nonsense-mediated decay (NMD) removes approximately 66-70% of mutant MYBPC3 transcripts in both cellular models and human heart tissue

  • For quantitative analysis, normalize protein expression to appropriate loading controls and compare to wild-type reference samples

These considerations ensure reliable interpretation of results when studying MYBPC3 mutations in cellular models.

What are the optimal approaches for using MYBPC3 antibodies in gene therapy research applications?

For researchers investigating AAV-based gene therapies for MYBPC3-associated HCM:

  • MYBPC3 antibodies can be used to confirm successful transgene expression following AAV delivery

  • Consider using antibodies specific to wild-type MYBPC3 that do not cross-react with common mutant forms

  • In AAV9 gene therapy models, antibodies can help verify cardiac-specific expression patterns

  • Understand that approximately 72% of MYBPC3-associated HCM patients have AAV9 neutralizing antibody titers ≤1:10, which is relevant for clinical translation

When designing experiments to evaluate gene therapy efficacy, include robust quantification of MYBPC3 protein levels using calibrated Western blot analysis or ELISA techniques with appropriate controls.

How should researchers address epitope-specific considerations when selecting MYBPC3 antibodies?

The structural complexity of MYBPC3 requires careful consideration of epitope location:

  • MYBPC3 contains multiple immunoglobulin and fibronectin domains (C0-C10), with nontruncating pathogenic variants clustering particularly in the C3, C6, and C10 domains

  • Antibodies targeting the C10 domain may show altered binding patterns in certain mutations, as C10 mutant MyBP-C often fails to incorporate into myofilaments and shows accelerated degradation (≈90% faster)

  • In contrast, C3 and C6 mutant MYBPC3 typically incorporates normally with degradation rates similar to wild-type

Therefore, researchers should select antibodies based on the specific domain of interest and the experimental question being addressed. For comprehensive analysis, consider using antibodies targeting different domains simultaneously.

What strategies can resolve weak or absent signal when using MYBPC3 antibodies in Western blot applications?

When facing detection challenges:

  • Optimize protein extraction from cardiac tissue:

    • Use specialized extraction buffers containing protease inhibitors

    • Ensure complete homogenization of fibrous cardiac tissue

    • Consider sonication to improve protein solubilization

  • Adjust antibody conditions:

    • Increase primary antibody concentration (up to 1:500 dilution)

    • Extend primary antibody incubation time (overnight at 4°C)

    • Test different blocking agents (5% milk vs. 5% BSA)

  • Verify protein transfer:

    • MYBPC3 is a large protein (141-150 kDa) that requires extended transfer times

    • Use Ponceau S staining to confirm successful transfer of high molecular weight proteins

    • Consider wet transfer methods for large proteins like MYBPC3

Proper sample preparation is critical, as MYBPC3's association with the cardiac sarcomere can make complete protein extraction challenging.

How can cross-reactivity issues with MYBPC3 antibodies be identified and addressed?

To minimize cross-reactivity concerns:

  • Implement proper controls:

    • Include MYBPC3 knockout samples as negative controls

    • Use purified recombinant MYBPC3 protein as a positive control

    • Test antibody specificity in tissues known not to express MYBPC3

  • Validation approaches:

    • Perform peptide competition assays to confirm specificity

    • Consider using multiple antibodies targeting different MYBPC3 epitopes

    • Compare reactivity patterns across species with predicted sequence homology

  • Experimental design considerations:

    • Increase washing steps duration and frequency

    • Adjust antibody dilution to minimize non-specific binding

    • Pre-absorb antibodies with non-cardiac tissue lysates if non-specific binding persists

These approaches can help distinguish between true MYBPC3 signal and potential cross-reactivity with other MyBP family members or related proteins.

How can MYBPC3 antibodies be utilized in non-invasive biomarker development for hypertrophic cardiomyopathy?

For biomarker development research:

  • Sandwich ELISA approaches:

    • Utilize capture and detector antibodies targeting different MYBPC3 epitopes

    • For human MYBPC3 detection, a capture antibody dilution of 2 µg/mL and detector antibody dilution of 0.5 µg/mL has been validated

    • Consider detecting both full-length and truncated MYBPC3 fragments in circulation

  • Study design considerations:

    • Include appropriate patient cohorts with confirmed MYBPC3 mutations

    • Correlate circulating MYBPC3 levels with clinical parameters and outcomes

    • Establish normal reference ranges from healthy control populations

  • Technical validation:

    • Confirm assay specificity using recombinant MYBPC3 proteins

    • Assess potential interference from other circulating proteins

    • Determine minimum detection limits and dynamic range for clinical utility

This approach leverages MYBPC3 antibodies beyond traditional research applications toward clinical biomarker development for early disease detection and monitoring.

What considerations are important when using MYBPC3 antibodies to investigate posttranslational modifications?

MYBPC3 undergoes significant regulatory phosphorylation, particularly by cAMP-dependent protein kinase (PKA) during adrenergic stimulation . When studying these modifications:

  • Selection of phospho-specific antibodies:

    • Choose antibodies that recognize specific phosphorylation sites

    • Validate specificity using phosphatase treatments

    • Consider the impact of sample preparation on phosphorylation status

  • Experimental design:

    • Include appropriate controls for basal and stimulated phosphorylation states

    • Carefully time sample collection to capture dynamic phosphorylation events

    • Consider phosphorylation status in the context of disease models

  • Technical considerations:

    • Use phosphatase inhibitors during sample preparation

    • Avoid excessive sample heating which may affect phospho-epitopes

    • Consider parallel detection of total and phospho-MYBPC3 for normalization

These approaches enable researchers to investigate how MYBPC3 phosphorylation states relate to cardiac contractility regulation and disease pathogenesis.

How might MYBPC3 antibodies contribute to next-generation gene therapy assessment in cardiomyopathies?

As gene therapy approaches for MYBPC3-associated cardiomyopathies advance:

  • Evaluating therapeutic efficacy:

    • Antibody-based quantification of restoration of normal MYBPC3 levels

    • Assessment of proper sarcomeric incorporation of therapeutic MYBPC3

    • Monitoring long-term stability of gene therapy-derived MYBPC3 expression

  • Emerging research applications:

    • Development of non-invasive monitoring techniques for gene therapy patients

    • Correlation of MYBPC3 restoration with functional cardiac improvements

    • Identification of potential immune responses to introduced wild-type MYBPC3

  • Clinical translation considerations:

    • Understanding that approximately 92% of MYBPC3-associated HCM patients have AAV9 neutralizing antibody titers ≤1:80, indicating potential eligibility for gene therapy

    • Developing companion diagnostics using MYBPC3 antibodies to select appropriate patients

    • Monitoring for immune responses against restored MYBPC3 protein in patients with frameshift mutations

These applications position MYBPC3 antibodies as critical tools in the translational pipeline for novel gene therapies targeting hypertrophic cardiomyopathy.

What role might MYBPC3 antibodies play in understanding the molecular mechanisms of heart failure progression in cardiomyopathy patients?

For investigating disease progression mechanisms:

  • Structural and functional changes:

    • Analyze MYBPC3 distribution patterns in different stages of heart failure

    • Assess correlations between MYBPC3 levels/localization and functional parameters

    • Investigate temporal changes in MYBPC3 expression during disease progression

  • Interaction studies:

    • Use co-immunoprecipitation with MYBPC3 antibodies to identify altered binding partners

    • Investigate changes in MYBPC3 interactions with actin, myosin, and titin

    • Study how these interactions are modified in different stages of cardiomyopathy

  • Mechanistic insights:

    • Determine whether MYBPC3 degradation rates change with disease progression

    • Investigate potential post-translational modifications as disease biomarkers

    • Study compensatory mechanisms in response to MYBPC3 haploinsufficiency

These applications can provide deeper understanding of how MYBPC3 mutations lead to progressive cardiac dysfunction, potentially identifying new therapeutic targets beyond gene replacement.

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.