MYO7A Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Structure and Production

The MYO7A recombinant monoclonal antibody is engineered to bind specifically to the MYO7A protein. Key structural and production features include:

FeatureDetailsSource
ImmunogenSynthetic peptide derived from human MYO7A (e.g., clone 31A12, AFCH-13)
Host CellsHEK293F suspension cells (transiently transfected) or rabbit/mouse systems
ClonalityRecombinant monoclonal (e.g., rabbit IgG, mouse IgG2a)
PurificationAffinity chromatography (e.g., Protein A/G)
ConjugationUnconjugated (standard) or tagged (e.g., HA, FLAG)

Recombinant production bypasses traditional hybridoma techniques, enabling precise control over epitope specificity and reducing batch variability.

Applications in Research and Diagnostics

The antibody is validated for multiple techniques:

Enzyme-Linked Immunosorbent Assay (ELISA)

  • Purpose: Quantitative detection of MYO7A in serum or cell lysates.

  • Reactivity: Human, with cross-reactivity in mouse and rat models .

  • Optimal Dilution: 1:50–1:200 (varies by clone) .

Western Blot (WB)

  • Purpose: Confirmation of MYO7A expression in protein lysates.

  • Key Clones:

    • 31A12: Detects full-length MYO7A (~254 kDa) in Y79 retinoblastoma cells .

    • AFCH-13: Validates MYO7A in Y79 lysates at 1:500–1:2000 dilution .

  • Observed Band: ~254 kDa (matches MYO7A’s molecular weight) .

Immunohistochemistry (IHC)

  • Tissue Specificity:

    • Human: Lung cancer, stomach cancer .

    • Mouse/Rat: Retinal and cochlear tissues (e.g., hair cells, photoreceptors) .

  • Protocol: Antigen retrieval with EDTA buffer (pH 8.0), blocking with 10% goat serum, and HRP-based detection .

Flow Cytometry (FC)

  • Applications: Quantification of MYO7A in cell suspensions (e.g., retinal pigment epithelial cells).

  • Reactivity: Human, mouse, rat .

Immunofluorescence (IF/ICC)

  • Applications: Localization of MYO7A in Y79 cells or cochlear hair cells.

  • Example: Clone EPR7498 (Abcam) detects MYO7A in Y79 cells at 1:100 dilution .

Usher Syndrome and Hearing Loss

  • Role in USH1B: MYO7A mutations disrupt stereocilia organization in cochlear hair cells, leading to hearing loss. Antibodies validate gene therapy approaches, such as dual-AAV vectors delivering MYO7A, which restore inner hair cell (IHC) survival in shaker-1 mice .

  • Cochlear Transduction: MYO7A interacts with USH1C, USH1G, and CDH23 to mediate mechanotransduction .

Retinal Degeneration

  • Melanosome Transport: MYO7A regulates melanosome distribution in retinal pigment epithelial cells. Antibodies track its role in photoreceptor disk renewal .

  • Gene Therapy: Subretinal injection of MYO7A-loaded dual-AAV vectors improves photoreceptor function in preclinical models .

Table 1: MYO7A Antibody Clones and Applications

CloneHostApplicationsReactivitySource
31A12Rabbit IgGELISA, FC, WBHuman
AFCH-13Rabbit IgGWBHuman, Mouse, Rat
EPR7498Rabbit IgGWB, ICC/IFHuman
M03915Rabbit IgGIHC, WBHuman, Mouse, Rat

Research Findings and Validation

  1. Dual-AAV Gene Therapy: Subretinal injection of dual-AAV8(Y733F)-MYO7A vectors restored MYO7A expression in shaker-1 mice, preserving 77% of IHCs at the cochlear apex and 82% of outer hair cells (OHCs) .

  2. IHC Validation: Clone M03915 detected MYO7A in human lung cancer, mouse/rat eye, and human stomach cancer tissues .

  3. WB Specificity: Clone 31A12 showed no cross-reactivity with superfamily members in WB .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

The MYO7A recombinant monoclonal antibody is generated through a process that begins with the insertion of the antibody-encoding gene into expression vectors. These vectors are then introduced into host cells via polyethyleneimine-mediated transfection. The host cells containing these vectors are cultured to produce and secrete the antibodies. Following affinity chromatography purification, the antibodies undergo rigorous testing, including ELISA and FC assays, to confirm their ability to recognize the human MYO7A protein.

MYO7A is a versatile protein playing critical roles in various cellular processes. Its primary functions include involvement in auditory and visual sensory functions, intracellular transport, maintenance of microvilli, cellular adhesion, vesicle trafficking, and melanosome transport. Mutations in the MYO7A gene can result in a range of disorders affecting hearing, vision, and other cellular processes.

Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. The delivery time may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributors.
Synonyms
Unconventional myosin-VIIa, MYO7A, USH1B
Target Names
MYO7A
Uniprot No.

Target Background

Function
Myosins are actin-based motor molecules exhibiting ATPase activity. Unconventional myosins are involved in intracellular movements. Their highly divergent tails bind to membranous compartments, which are then moved relative to actin filaments. In the retina, MYO7A plays a critical role in the renewal of the outer photoreceptor disks. It is also essential for the distribution and migration of retinal pigment epithelial (RPE) melanosomes and phagosomes, as well as in the regulation of opsin transport in retinal photoreceptors. In the inner ear, MYO7A plays a significant role in the differentiation, morphogenesis, and organization of cochlear hair cell bundles. Additionally, it is involved in hair-cell vesicle trafficking of aminoglycosides, which are known to induce ototoxicity. This motor protein is an integral part of the functional network formed by USH1C, USH1G, CDH23, and MYO7A, mediating mechanotransduction in cochlear hair cells. MYO7A is essential for normal hearing.
Gene References Into Functions
  1. Pathogenic mutation c.2011G>A identified in Chinese family with autosomal dominant hearing loss PMID: 29400105
  2. This study reports novel homozygous mutations in various genes causing Usher syndrome, expanding the spectrum of causative mutations. The study also validates combined sequencing techniques as valuable tools for identifying novel disease-causing mutations. This is the largest report of a genetic analysis of Israeli and Palestinian families (n = 74) with different Usher syndrome subtypes. PMID: 29490346
  3. This study extends the known spectrum of MYO7A mutations and demonstrates the utility of next-generation sequencing in the molecular diagnosis of Usher syndrome. PMID: 29605349
  4. These findings broaden the phenotypic spectrum of the MYO7A gene and may facilitate understanding the molecular pathogenesis of the disease, as well as provide genetic counseling for families. PMID: 28731162
  5. A novel stop-gained variant c.4513G > T (p.Glu1505Ter) in MYO7A was identified in an Iranian pedigree with two affected members with Usher syndrome type 1. PMID: 29287847
  6. Two pathogenic variations (c.849+2T>C and c.5994G>A) in MYO7A were successfully identified and individually separated from parents. PMID: 29287864
  7. This study reveals remarkable genetic heterogeneity in the studied families with Usher syndrome type 1, with a variety of mutations, including three novel ones. These novel mutations will be included in the NADf mutation screening chip, enabling higher diagnostic efficiency for this extremely genetically heterogeneous disease. PMID: 27440999
  8. Patients carrying mutations in MYO7A exhibited an earlier onset of hearing and visual impairments compared to those with mutations in USH2A, leading to an earlier diagnosis in the former group. PMID: 27828912
  9. The study suggests that the newly identified mutation named c.6079_6081del (p.H2027del) is the primary cause of deaf-blindness found in this family, clinically diagnosed as Usher syndrome type 1B. PMID: 26864046
  10. Results demonstrate that the structures of Myo7a IQ5-SAH (single a helix) lever arm extension in complex with apo- and Ca2+-CaM, respectively, reveal an extended and rigid lever arm in the motor at low Ca2+ concentration conditions. Increased cellular Ca2+ concentration induces conformational flexibility of the motor, thus regulating its activity. PMID: 28262393
  11. A novel mutation c.3847_3848insTCTG (p. N1285LfsX24) in compound heterozygosity with c.2239_2240delAG in the MYO7A gene is the primary cause of Usher syndrome type 1 in the proband. PMID: 28688563
  12. This study reports the results of genetic analyses performed on Moroccan families with autosomal recessive non-syndromic hearing loss, identifying two families with compound heterozygous MYO7A mutations. PMID: 28472130
  13. Myosin VIIa movement appears to be suitable for translocating USH1 proteins on stereocilia actin bundles in inner-ear hair cells. PMID: 28507101
  14. In USH1B-MYO7A, the constriction rate of EZ extent depends on the initial eccentricity of the transition. Ellipsoid zone edges in the macula correspond to large local changes in cone vision, but extramacular EZ edges show more pronounced losses on rod-based vision tests. PMID: 27409480
  15. An unreported splice site mutation c.3924+1G > C compound with c.6028G > A in the MYO7A gene were detected to cosegregate with Usher syndrome type 2 in a Han Chinese family. PMID: 27729122
  16. There are more than 39 deafness genes reported to cause non-syndromic hereditary hearing loss (HHL) in Iran, with the most prevalent causative genes including GJB2, SLC26A4, MYO15A, and MYO7A. This review highlights some of the more common genetic causes of syndromic HHL in Iran. PMID: 27743438
  17. This study demonstrates that Mendelian sensorineural hearing loss exhibits vestibular dysfunction, including DFNA9, DFNA11, DFNA15, and DFNA28. PMID: 27083884
  18. The novel compound heterozygous mutations (c.3671C>A and c.390_391insC) in the MYO7A gene identified in this study were responsible for the autosomal recessive sensorineural hearing loss of this Chinese family. PMID: 26968074
  19. MYO7A binds to and impinges on CASPASE-8, revealing a new regulatory axis affecting RIPK1>CASPASE-8 signaling. Results expose a conserved role for unconventional myosins in transducing caspase-dependent regulation of kinases. PMID: 26960254
  20. Genetic correction of the MYO7A mutation resulted in morphological and functional recovery of hair cell-like cells derived from induced pluripotent stem cells from a deaf patient. PMID: 27013738
  21. The c.6377delC mutation is found in a significant proportion of Usher syndrome cases among indigenous South Africans. PMID: 26469752
  22. Ten variants in the MYO7A gene and 34 variants in the USH2A gene were detected in Italian patients with Usher syndrome, exhibiting a high detection rate. PMID: 25558175
  23. The study concluded that Usher syndrome type 1 in this family was caused by compound heterozygous mutations in MYO7A. PMID: 25080338
  24. Electron microscopy revealed that myosin VIIA is a monomer where the tail domain bends back toward the head-neck domain, forming a compact structure. This compact structure is extended at high ionic strength or in the presence of Ca(2+). PMID: 26001786
  25. Clinical phenotypes of Usher syndrome associated with MYO7A mutations. PMID: 24831256
  26. The MYO7A gene is responsible for two distinct diseases and provides evidence that the p.P1887L mutation in a homozygous state may be responsible for nonsyndromic hearing loss. PMID: 24194196
  27. New variants in genes such as MYO7A are associated with nonsyndromic deafness and vestibular dysfunction. PMID: 24275721
  28. Data indicate that CIB2 localizes to stereocilia and interacts with the USH proteins myosin VIIa and whirlin, suggesting that CIB2 is a Ca2+-buffering protein essential for calcium homeostasis in the mechanosensory stereocilia of inner ear hair cells. PMID: 24022220
  29. Data show that AAV-mediated hMYO7A gene transfer to sh1(-/-) mice retina is effective. PMID: 23991031
  30. MYO7A-related ocular disease is variable. Central vision typically remains preserved at least until the third decade of life, with 50% of affected individuals reaching legal blindness by 40 years of age. PMID: 24199935
  31. Hearing loss was found to co-segregate with locus-specific STR markers for MYO7A in 32 Pakistani families. PMID: 23770805
  32. Results demonstrate that MYO7A therapy with AAV2 or AAV5 single vectors is efficacious; however, the dual AAV2 approach proved to be less effective. PMID: 23344065
  33. Possible digenism could not be excluded in two families segregating genomic variations in both MYO7A and USH2A, and two families with CLRN1 and USH2A. PMID: 22681893
  34. Two novel mutations, c.3742G>A (p.E1248K) and c.6051+1G>A (donor splice site mutation in intron 44), of MYO7A in a Chinese non-consanguineous family with Usher syndrome type 1, were identified. PMID: 23559863
  35. A new missense mutation (Arg668His) in the motor head domain of myosin VIIA in a family with autosomal dominant hearing impairment (DFNA11). PMID: 23383098
  36. A study of consanguineous patient families with Leber congenital amaurosis identified five putative disease-causing mutations, including four novel alleles, in six families. These five mutations are located in four genes: ALMS1, IQCB1, CNGA3, and MYO7A. PMID: 21901789
  37. A previously reported mutation (c.52C>T; p.Q18X) in the myosin VIIA (MYO7A) gene was found in the homozygous state in the affected siblings. PMID: 22690115
  38. Pathogenic mutations in MYO7A, USH1C, and USH1G have been found in four consanguineous Israeli Arab families with Usher syndrome type 1. PMID: 22219650
  39. Results suggest that in a cellular environment, compartment-specific fluctuations in free Mg2+ ions can mediate the conditional switching of myosin-7a between cargo moving and tension bearing modes. PMID: 21687988
  40. The authors speculate that null MYO7A alleles could be associated with milder dysfunction and fewer photoreceptor structural losses at ages when other genotypes show more severe phenotypes. PMID: 21873662
  41. Significant linkage in DFNA11 markers was found in hereditary deafness. PMID: 17702415
  42. A single nucleotide polymorphism (SNP) T/C at position -4128 in the wild-type MYO7A promoter allele that sorts with the degree of hearing loss severity in the pedigree. PMID: 21378158
  43. Association of myosin VIIA monomers with the membrane via the MyRip/Rab27a complex facilitates the cargo-transporting activity of myosin VIIA, achieved by cluster formation on the membrane. PMID: 21482763
  44. Novel missense mutations in MYO7A underlying postlingual high- or low-frequency non-syndromic hearing impairment in two large families. PMID: 21150918
  45. Five mutations (three in MYO7A and two in CDH23) were identified in four of five unrelated patients with Usher syndrome type 1. PMID: 20844544
  46. The molecular determinant of a mild form of retinopathy in association with a subtle splicing modulation of MYO7A mRNA was investigated. PMID: 21031134
  47. Variable hearing impairment in a DFNB2 family with a novel MYO7A missense mutation. PMID: 20132242
  48. The shaping of the hair bundle relies on a functional unit composed of myosin VIIa, harmonin b, and cadherin 23, essential for ensuring the cohesion of the stereocilia. PMID: 12485990
  49. Expressed in the retina. Domain structure. Carrier proteins determine cellular function. PMID: 15180257
  50. A new heterozygous missense mutation (c.2557C>T; p.R853C) was found in autosomal dominant non-syndromic hearing loss, changing an invariant residue of the fifth IQ motif, a putative calmodulin (CaM) binding domain. PMID: 15300860

Show More

Hide All

Database Links

HGNC: 7606

OMIM: 276900

KEGG: hsa:4647

STRING: 9606.ENSP00000386331

UniGene: Hs.370421

Involvement In Disease
Usher syndrome 1B (USH1B); Deafness, autosomal recessive, 2 (DFNB2); Deafness, autosomal dominant, 11 (DFNA11)
Protein Families
TRAFAC class myosin-kinesin ATPase superfamily, Myosin family
Subcellular Location
Cytoplasm. Cytoplasm, cell cortex. Cytoplasm, cytoskeleton. Cell junction, synapse.
Tissue Specificity
Expressed in the pigment epithelium and the photoreceptor cells of the retina. Also found in kidney, liver, testis, cochlea, lymphocytes. Not expressed in brain.

Q&A

What is MYO7A and what are its primary functions in cellular processes?

MYO7A is a large, multifunctional motor protein (calculated molecular weight of 254,390 Da) that plays diverse roles in cellular processes. Its primary functions include:

  • Maintenance of stereocilia structure and function in inner ear hair cells

  • Visual sensory functions in the retina

  • Intracellular transport and trafficking

  • Maintenance of microvilli and cellular adhesion

  • Vesicle trafficking and melanosome transport

  • Formation of protein complexes with other sensory proteins

MYO7A is particularly important in auditory and visual pathways, where mutations can lead to disorders affecting hearing, vision, and other cellular processes . The protein forms complexes with partners like PDZD7 (a paralog of USH1C and DFNB31) and is associated with membranes in various cell types .

How are MYO7A recombinant monoclonal antibodies generated?

The production of MYO7A recombinant monoclonal antibodies follows a sophisticated molecular biology process:

  • The MYO7A antibody-encoding gene is inserted into expression vectors

  • These vectors are delivered into host cells using polyethyleneimine-mediated transfection

  • Host cells containing these vectors are cultured to produce and secrete the antibodies

  • The antibodies undergo affinity chromatography purification

  • Quality control includes ELISA and flow cytometry (FC) tests to confirm target recognition

  • The antibodies are validated for specific reactivity to human MYO7A protein

This recombinant approach provides advantages over traditional hybridoma-based monoclonal antibody production, including better batch-to-batch consistency and the ability to engineer specific binding properties.

What applications are MYO7A antibodies validated for in research settings?

MYO7A antibodies have been validated for multiple research applications, with varying recommended dilutions based on the specific assay:

ApplicationRecommended DilutionValidation Techniques
Flow Cytometry (FC)1:50-1:200Target cell analysis
Western Blot (WB)1:50Protein size confirmation
ELISAVaries by assayProtein quantification
ImmunoprecipitationNot specified in sourcesComplex isolation

Researchers should note that specific antibodies like the one described in source react with both human and mouse MYO7A, making them versatile for cross-species studies. Each application requires optimization of conditions for specific experimental systems.

How can MYO7A antibodies be utilized to study protein complexes in stereocilia?

MYO7A forms critical complexes with other proteins in stereocilia, making antibody-based approaches valuable for studying these interactions. A methodological approach includes:

  • Stabilize protein complexes using chemical crosslinking with membrane-impermeant crosslinker DTSSP, which enhances detection of low-abundance membrane protein interactions

  • Use stringent extraction and washing conditions to minimize dissociation of protein complexes that include extracellular domains

  • Employ immunoaffinity purification using anti-MYO7A antibodies (such as the 138-1 monoclonal antibody for chicken MYO7A)

  • Remove nonspecific proteins by first passing extracts over control beads constructed with purified IgG

  • Apply the unbound material to anti-MYO7A antibody-conjugated beads

  • After thorough washing, elute with SDS and analyze by shotgun and targeted mass spectrometry

This approach has successfully identified MYO7A complexes with PDZD7, USH1C, and CDH23, suggesting a functional network in stereocilia . The high enrichment (approximately 10,000-fold) of MYO7A through this method allows detection of even rare interacting partners.

What strategies should be employed when validating MYO7A antibody specificity for studies in mutant models?

When validating MYO7A antibody specificity in mutant models, researchers should implement a multi-tiered approach:

  • Genetic controls: Use MYO7A-null models (such as shaker-1 Myo7a^-/-^ mice or Myo7a^4626SB/4626SB^ mutants) as negative controls to confirm antibody specificity

  • Epitope mapping: Verify antibody recognition using antibodies directed against different portions of MYO7A (N-terminal, middle, and C-terminal regions)

  • Cross-reactivity assessment: Test antibody reactivity against related myosin family members

  • Tagged protein validation: Compare staining patterns between antibodies recognizing native MYO7A and those recognizing epitope tags (such as HA) in tagged transgene expression systems

  • Immunoblotting validation: Confirm the detection of appropriately sized bands in wild-type but not mutant tissues

Research has demonstrated that proper antibody validation is critical when studying MYO7A, as demonstrated in studies using shaker-1 mice where antibodies detecting the middle portion of MYO7A only detected expression when both 5′ and 3′ vectors were used to infect cells .

What considerations are important when designing experiments to study MYO7A-mediated functions in sensory cells?

Designing experiments to study MYO7A-mediated functions requires careful consideration of multiple factors:

  • Tissue-specific expression patterns: MYO7A functions differ between auditory hair cells, vestibular hair cells, and retinal cells, necessitating tissue-specific experimental designs

  • Subcellular localization: Consider the distribution of MYO7A within different cellular compartments (stereocilia, cell body, synapses)

  • Temporal dynamics: Account for developmental timing and age-related changes in MYO7A expression and function

  • Genetic background effects: Control for genetic background influences on phenotypic expression in MYO7A mutant models

  • Redundancy with other myosins: Consider potential functional overlap with other unconventional myosins

  • Protein complex formation: Design experiments that capture MYO7A's participation in different protein complexes

Research has shown that MYO7A is multifunctional, interacting with several different protein complexes in stereocilia and carrying out various transport and functional roles . Therefore, experimental designs must account for these multiple functions rather than assuming a single role.

What technical challenges arise when using MYO7A antibodies for immunoprecipitation of protein complexes?

Immunoprecipitation of MYO7A protein complexes presents several technical challenges:

  • Low abundance: MYO7A and its complexes are rare within stereocilia, requiring approximately 10^6-fold enrichment from dissected inner ears for definitive detection

  • Complex integrity preservation: Maintaining intact protein complexes during extraction and purification is challenging, often requiring chemical crosslinking

  • Nonspecific binding: Extensive washing is required to reduce nonspecific binding, which biases analysis toward high-affinity interactions

  • Starting material limitations: Large-scale purification requires substantial starting material (hundreds of ear-equivalents)

  • Sequential purification requirements: Multiple purification steps may be needed to achieve sufficient enrichment

Research suggests that future studies should employ a second antibody purification step following initial precipitation with anti-MYO7A antibodies to generate purer complexes. This might involve using antibodies against known interacting partners like anti-PDZD7 .

How can dual-AAV vector-mediated MYO7A expression be optimized in inner ear research?

Optimizing dual-AAV vector-mediated MYO7A expression for inner ear research requires attention to several critical factors:

  • Vector design: Use hybrid dual-AAV vectors with recombinogenic sequences (e.g., AP coding sequence) and splice donor/acceptor sites to promote efficient recombination and transcript processing

  • Vector serotype selection: AAV8(Y733F) has shown efficacy for inner ear delivery

  • Promoter choice: The ubiquitous CMV/chicken β-actin (smCBA) promoter has proven effective for MYO7A expression

  • Injection technique: Employ posterior semicircular canal (PSC) injection for efficient cochlear transduction

  • Timing of intervention: Administer therapy early enough to prevent irreversible damage

  • Vector dose optimization: Titrate vector doses to achieve sufficient expression while minimizing toxicity

Studies have demonstrated that hybrid dual-AAV8(Y733F)-MYO7A vectors can restore MYO7A protein expression in transduced shaker-1 inner hair cells (IHCs) and outer hair cells (OHCs), significantly improving hair cell survival compared to untreated mutants .

What protocols are recommended for validating MYO7A antibodies across different species?

When validating MYO7A antibodies for cross-species reactivity, researchers should implement the following protocols:

  • Sequence alignment analysis: Compare MYO7A sequences across target species to identify conserved and variable regions

  • Epitope selection: For recombinant antibodies, target highly conserved regions (the AA range 830-910 has been used successfully for antibodies reacting with both human and mouse MYO7A)

  • Multiple application testing: Validate antibodies in different applications (WB, ELISA, FC, IHC) for each species

  • Positive control samples: Use well-characterized positive control samples from each species

  • Negative control samples: Include MYO7A-null or knockdown samples when available

  • Cross-reactivity panel: Test against closely related myosin family members

Researchers should be aware that antibody validation is application-specific – an antibody that works well for Western blotting may not perform adequately in immunohistochemistry or flow cytometry applications.

How should researchers interpret data from MYO7A localization studies in different sensory cell types?

Interpreting MYO7A localization data across different sensory cell types requires careful consideration of:

  • Cell type-specific distribution patterns: MYO7A localizes differently in various cell types:

    • In hair cells: Concentrated in stereocilia, cuticular plate, and cell body

    • In retinal cells: Associated with connecting cilium and melanosomes

  • Fixation and permeabilization effects: Different protocols can affect epitope accessibility and apparent distribution patterns

  • Antibody specificity considerations: Different antibodies targeting different regions of MYO7A (N-terminal, middle, or C-terminal portions) may show slightly different localization patterns

  • Co-localization analysis: Examine co-distribution with known interacting partners (PDZD7, USH1C, CDH23) to validate functional associations

  • Resolution limitations: Consider the diffraction limit of conventional microscopy versus super-resolution techniques when interpreting apparent co-localization

Research has demonstrated that MYO7A's localization correlates with its diverse functions, including interactions with stereocilia membranes and protein complexes at tip links and ankle links .

What are the key considerations when analyzing MYO7A complex formation in mass spectrometry data?

When analyzing mass spectrometry data for MYO7A complexes, researchers should consider:

  • Enrichment analysis: Compare relative abundance of proteins in immunoprecipitates versus starting material to calculate immunoaffinity enrichment ratios (MYO7A itself should show high enrichment, ~10-fold or greater)

  • Specificity controls: Distinguish proteins detected only in MYO7A immunoprecipitates from those in control samples

  • Known complex components: Verify presence of established MYO7A-interacting proteins (USH1C, USH1G, CDH23) as internal validation

  • Abundance estimates: Consider that even after 10,000-fold purification, MYO7A complex components may be present at low levels

  • Crosslinking effects: Account for potential artificial associations introduced by chemical crosslinking

  • Stoichiometry limitations: Recognize that additional purification steps are required for accurate stoichiometric determination of complex components

Research has shown that despite challenges, large-scale purification of protein complexes from stereocilia can identify interacting components that may not be detected by other strategies .

How might MYO7A antibodies be utilized in therapeutic development for Usher syndrome?

MYO7A antibodies have significant potential in therapeutic development for Usher syndrome through multiple approaches:

  • Biomarker development: Use antibodies to measure MYO7A expression levels and localization as biomarkers for therapeutic efficacy

  • Vector development assessment: Employ antibodies to evaluate dual-AAV vector-mediated MYO7A expression in preclinical models

  • Gene therapy monitoring: Use antibodies to assess restoration of MYO7A expression following gene therapy in shaker-1 mice or human tissues

  • Drug screening: Apply antibodies in high-throughput screens for compounds that stabilize mutant MYO7A or enhance its function

  • Therapeutic antibody engineering: Develop antibody-based therapeutics that might stabilize MYO7A complexes or enhance their function

Research has demonstrated that dual-AAV vector delivery of human MYO7A cDNA can restore MYO7A expression in mouse models, with significant preservation of inner hair cells compared to untreated mutants , suggesting promising therapeutic avenues.

What roles might MYO7A play in protein complexes beyond the currently established interactions?

Current research suggests MYO7A may participate in additional protein complexes and functions beyond those currently established:

  • Novel binding partners: Further immunoprecipitation and mass spectrometry studies may reveal currently unknown interaction partners

  • Trafficking complexes: MYO7A likely participates in various cargo trafficking complexes beyond those characterized in stereocilia

  • Transcriptional regulation: Potential nuclear functions have not been fully explored

  • Signaling pathway integration: MYO7A may link mechanical stimuli to intracellular signaling cascades

  • Immune system roles: Possible functions in specialized immune cells have not been extensively investigated

Research has indicated that MYO7A is a multifunctional protein interacting with several different protein complexes in stereocilia and carrying out transport and functional roles , suggesting many aspects of its biology remain to be discovered.

What are the optimal protocols for using MYO7A antibodies in immunofluorescence studies of inner ear tissues?

Immunofluorescence studies of MYO7A in inner ear tissues require specialized protocols:

  • Tissue preparation:

    • Fresh dissection or rapid fixation is critical

    • Fixation with 4% paraformaldehyde for 30-60 minutes preserves structure while maintaining epitope accessibility

    • Consider specialized fixatives for transmission electron microscopy studies

  • Decalcification considerations:

    • For intact cochlea, use EDTA-based decalcification to preserve antigenicity

    • Monitor decalcification progress to avoid over-processing

  • Permeabilization optimization:

    • Test different detergents (Triton X-100, saponin) at various concentrations

    • Balance permeabilization for antibody access while preserving delicate stereocilia structures

  • Antibody incubation:

    • Extended incubation times (overnight at 4°C) improve penetration

    • Use antibodies directed against different portions of MYO7A (N-terminal, middle, and C-terminal) to validate findings

  • Controls:

    • Include MYO7A-deficient tissues (e.g., from shaker-1 mice) as negative controls

    • Use dual-labeling with established hair cell markers for reference

These approaches have successfully demonstrated MYO7A expression patterns in both normal and genetically modified inner ear tissues .

How can researchers quantitatively assess MYO7A expression levels in different experimental conditions?

Quantitative assessment of MYO7A expression requires rigorous methodological approaches:

  • Western blot quantification:

    • Use purified recombinant MYO7A standards for absolute quantification

    • Normalize to appropriate housekeeping proteins or total protein

    • Employ fluorescent secondary antibodies for improved linear range

  • ELISA-based quantification:

    • Develop sandwich ELISA using different epitope-targeting antibodies

    • Generate standard curves with recombinant MYO7A

  • Flow cytometry:

    • Optimize cell permeabilization protocols for this large intracellular protein

    • Use recommended antibody dilutions (1:50-1:200) for FC applications

    • Include proper controls for autofluorescence and nonspecific binding

  • Image analysis:

    • Apply consistent acquisition parameters across samples

    • Use automated, threshold-based quantification

    • Normalize to cell number or tissue area

    • Account for regional variations in expression (apex vs. middle turn vs. base of cochlea)

Research has demonstrated regional differences in MYO7A expression following treatment, with measurements showing 14 ± 1.2 cells/136 μm at the apex, 14 ± 1.9 cells/136 μm at the middle turn, and 7.3 ± 1.8 cells/136 μm at the base of the cochlea .

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 2024 Thebiotek. All Rights Reserved.