GJB2 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Overview of GJB2 and Its Biological Role

GJB2 encodes Connexin 26, a transmembrane protein critical for forming gap junctions that facilitate intercellular communication. These channels allow the transfer of ions and small molecules (≤1 kDa), including metabolites and signaling molecules, between adjacent cells . Mutations in GJB2 are linked to hereditary deafness, skin disorders, and placental developmental defects .

Definition and Production of Recombinant Monoclonal Antibodies

Recombinant monoclonal antibodies (R-mAbs) are engineered using defined genetic sequences inserted into mammalian expression systems. Unlike traditional hybridoma-derived antibodies, R-mAbs ensure batch-to-batch consistency and eliminate genetic drift risks . The GJB2 R-mAb is typically produced by cloning immunoglobulin variable domains into plasmids and expressing them in host cells (e.g., HEK-293T) .

Inhibitory Effects on Pathogenic Hemichannels

A human-derived GJB2 monoclonal antibody demonstrated reversible inhibition of Cx26 hemichannels without disrupting gap junction communication. This antibody effectively suppressed hyperactive mutants linked to keratinopathies and deafness (KID/HID syndrome) via steric hindrance identified through crystallography .

Role in Developmental Studies

  • Placental Defects: Homozygous Gjb2 knockout mice generated via tetraploid embryo complementation exhibited severe placental dysplasia, underscoring GJB2's role in trophoblast differentiation .

  • Cochlear Function: Immunohistochemical analysis using ab303498 confirmed Cx26 expression in mouse cochlea, supporting its involvement in auditory signaling .

Validation and Technical Performance

  • IHC Optimization: ab303498 showed robust staining in paraffin-embedded liver and cochlea tissues at 1/500 dilution, validated using Leica Bond RX instruments .

  • Cross-Reactivity: Negative controls in skeletal muscle confirmed specificity, with no off-target binding observed .

  • Subclass Switching: Engineered IgG subclass variants (e.g., IgG1/IgG2a) enable multiplex labeling without altering binding affinity .

Future Directions

  • Therapeutic Potential: Engineered R-mAbs could treat Cx26-related disorders by targeting hyperactive hemichannels .

  • Diagnostic Tools: High-specificity antibodies may improve genetic screening for GJB2 mutations in prenatal diagnostics .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the order method and location. Please consult your local distributor for specific delivery time estimates.
Synonyms
connexin 26 antibody; Connexin-26 antibody; Cx26 antibody; CXB2_HUMAN antibody; DFNA3 antibody; DFNB1 antibody; Gap junction beta-2 protein antibody; GJB2 antibody; HID antibody; KID antibody; NSRD1 antibody; PPK antibody
Target Names
Uniprot No.

Target Background

Function
Connexin 26 (Cx26), encoded by the GJB2 gene, is a structural component of gap junctions. Gap junctions are specialized intercellular channels formed by the docking of two hexameric hemichannels, one from each cell membrane. These channels allow the diffusion of small molecules and ions between the cytoplasm of adjacent cells, facilitating cell-to-cell communication and coordination.
Gene References Into Functions
  1. In triple-negative breast cancer, Cx26 is elevated in self-renewing cancer stem cells (CSCs) and is essential for their maintenance. Cx26 promotes CSC self-renewal by forming a signaling complex with the pluripotency transcription factor NANOG and focal adhesion kinase (FAK), leading to NANOG stabilization and FAK activation. PMID: 29422613
  2. Children with GJB2 gene mutations exhibit phenotypic variability in terms of their hearing loss severity and symmetry. Individuals with T/NT mutations of the GJB2 gene are more likely to pass newborn hearing screening (UNHS) and have milder hearing loss compared to those with T/T mutations. PMID: 30146550
  3. Almost half of children with sensorineural hearing loss carry a common deafness-related mutation, and nearly one-third carry a pathogenic mutation. At least one mutated allele was detected in 48 patients, and 30 patients carried pathogenic mutations. The most prevalent mutations were GJB2 c.235delC and SLC26A4 c.919-2A>G. PMID: 30036422
  4. Studies have shown that thiol levels increase and disulfide levels decrease in patients with autosomal recessive non-syndromic hearing loss and GJB2 gene mutations. PMID: 30055731
  5. The mutation frequencies of GJB2, SLC26A4, GJB3, and mitochondrial genes were 3.04%, 3.51%, 0.16%, and 0.88%, respectively, among the Hakka population of Southern China. PMID: 30235673
  6. A novel missense mutation in GJB2 (DFNA3), affecting the second extracellular domain of the protein, was identified in a family with autosomal dominant non-syndromic hearing loss. PMID: 28102197
  7. Current findings indicate an association between GJB2 polymorphisms (rs2274084) and nasopharyngeal carcinoma (NPC) susceptibility. The TT genotype of GJB2 may be a risk factor for NPC. PMID: 29103018
  8. Mutations in the GJB2 gene are associated with deafness. PMID: 29634755
  9. The p.Lys22Asn GJB2 mutation causes a dominant form of hearing loss associated with variable expression of palmoplantar keratoderma, representing a model of full penetrance, with an age-dependent effect on the phenotype. PMID: 28872160
  10. Biallelic variations in the GJB2 gene cause up to 50% of cases of newborn hearing loss. PMID: 28821934
  11. GJB2 mutations are associated with hearing loss. PMID: 28405014
  12. Family studies implicate mutations in GJB2 and USH2A in Usher's syndrome with congenital hearing loss. PMID: 29151245
  13. Compared to previous studies, the c.109G>A mutation allele of GJB2 was found to be relatively lower in the profound Chinese nonsyndromic sensorineural hearing loss population compared to the moderate-to-profound ones. Conversely, the c.1174A>T mutation allele of SLC26A4 was relatively higher. PMID: 28786104
  14. The identification of a previously identified c.100C>T mutation and a novel homozygous mutation, c.1283C>A in TMC1, in this study supports TMC1 gene as one of the second-tier hearing loss genes after GJB2 in India. Testing for TMC1 may be considered in all GJB2-negative nonsyndromic hearing loss cases. PMID: 28862181
  15. These findings strongly suggest a pathogenic role for GJB3 p.V37I in various hearing loss phenotypes and provide a quantitative assessment of the risk associated with carrying this variant and developing hearing loss. PMID: 28489599
  16. Mutations in the GJB2 gene, particularly c.35delG, are significant causes of autosomal recessive non-syndromic hearing loss in the center and west of Iran. PMID: 29501291
  17. For the first time, a p.R75Q mutation exhibits intra-familial phenotypic variability. Profoundly deaf twins and their deaf maternal grandmother exhibit the p.R75Q mutation with palmoplantar keratoderma, while their deaf mother shows absence of skin disorders. The twins also had a recessive 35delG, which leads to a truncated premature protein inhibiting any action of the dominant p.R75Q mutation. PMID: 27316387
  18. The DFNB1 locus does not appear to be a major contributor to nonsyndromic sensorineural hearing loss (NSSHL) in Sao Tome and Principe. However, the presence of both pathogenic and likely pathogenic mutations in GJB2 suggests that GJB2-related NSSHL might still occur in this population. PMID: 27501294
  19. Given that a previous paper suggested TMPRSS3 and GJB2 genes as responsible for a digenic form of hearing loss, these data support and reinforce this hypothesis. PMID: 28263784
  20. A series of molecular dynamics simulations has been performed to investigate the effect of applied static and alternating electric fields on the stability and conformation of human connexin 26 hemichannel. PMID: 28259639
  21. The homozygous mutation c.35delG was identified as the cause of hearing loss in six participants (12%). The mutation c.506G>A was identified in three affected individuals (6%). The allelic frequency (14%) and low percentage of individuals that were homozygous (12%) and heterozygous (2%) for the c.35delG mutation suggest that other genes are also responsible for nonsyndromic deafness in the UAE population. PMID: 29016196
  22. Simultaneous hearing screening and genetic screening targeting four common deafness mutations (p.V37I and c.235delC of GJB2, c.919-2A>G of SLC26A4, and the mitochondrial m.1555A>G) were performed in 5173 newborns at a tertiary hospital between 2009 and 2015. This study delineated the longitudinal auditory features of the highly prevalent GJB2 p.V37I mutation on a general population basis. PMID: 27308839
  23. The proportion of carriers for GJB2 gene mutations in patients with hearing loss from southern Zhejiang has reached 21.5%. PMID: 28777850
  24. Genotype may affect deafness severity, but environmental and other genetic factors may also modulate the severity and evolution of GJB2-GJB6 deafness. PMID: 29106882
  25. Results suggest that GJB2 and CIB2 are common causes of hearing loss in different Pakistani ethnicities. PMID: 29086887
  26. GJB2 and ERO1LB are implicated in pancreatic cancer progression and can be used to predict patient survival. PMID: 28177904
  27. A significant proportion of children with unilateral sensorineural hearing loss may have positive genetic testing, with the vast majority of these children presenting with heterozygous mutations of connexin 26 (GJB2). PMID: 27466889
  28. WFS1 and GJB2 mutations were identified in eight out of 74 cases of Low-Frequency Sensorineural Hearing Loss. Four cases had heterozygous WFS1 mutations; one had a heterozygous WFS1 mutation and a heterozygous GJB2 mutation; and three cases had biallelic GJB2 mutations. Three cases with WFS1 mutations were sporadic; two of them were confirmed to be caused by a de novo mutation based on the genetic analysis of their parents. PMID: 28271504
  29. Results demonstrate that 19.2% of patients with nonsyndromic deafness were caused by mutations in three common deafness genes (GJB2, SLC26A4, and 12S rRNA) in a northern China patient group. PMID: 28583500
  30. GJB2-related deafness leads to significantly better cochlear implantation outcomes compared to acquired deafness caused by environmental etiologies. However, GJB2 mutation is not associated with a significantly better prognosis when compared to those whose deafness results from either nonsyndromic hearing loss of unknown origin or other types of genetic mutations in the absence of other neurologic deficits. PMID: 28322114
  31. Expression of Cx26 (also known as GJB2) in HeLa cells specifically enhances cell motility in scrape wounding and sparse culture models. PMID: 27777264
  32. Results found that the contribution of the GJB2 gene pathogenic variants to hearing impairment in the population of the Sakha Republic was the highest among all regions of Asia studied previously. This suggests that extensive accumulation of the c.-23+1G>A pathogenic variant in the indigenous Yakut population may indicate a possible selective advantage for c.-23+1G>A carriers living in the subarctic climate. PMID: 27224056
  33. These findings suggest that Cx26 mutants that promote cell death or exert transdominant effects on other connexins in keratinocytes will lead to skin diseases and hearing loss. PMID: 28428247
  34. Cx26-Asp50Asn with the second-site mutations identified in the patient displayed no formation of gap junction channel plaques. It is argued that the second-site mutations independently inhibit Cx26-Asp50Asn expression in gap junction channels, reverting the dominant negative effect of the p.Asp50Asn mutation. PMID: 28158657
  35. Cx26 and Cx30 proteins appear not to be co-expressed but to form closely associated assemblies of gap junction plaques. PMID: 26941236
  36. A study attempted to identify the genetic epidemiology of hereditary hearing loss among the Chinese Han population using next-generation sequencing. The entire length of the genes GJB2, SLC26A4, and GJB3 were sequenced from 116 individuals suffering from hearing loss. In this study, SLC26A4 and GJB2 were the most frequently affected genes among the Chinese Han population with hearing loss. PMID: 27610647
  37. Deafness associated with G109V could result from decreased gap junction channel (GJCs) activity, whereas deafness associated with L10P may have a more complex mechanism involving changes in hair cell permeability. PMID: 26769242
  38. Somatic mutations in the GJB2 gene cause nevoid spiny hyperkeratosis. PMID: 27087580
  39. There was a high prevalence of the IVS1+1G>A mutation in this sample of deaf families in Syria. PMID: 28012540
  40. The homozygous GJB2 c.109G[A mutation may be a cause of sudden death involving both ears. PMID: 26119842
  41. This study suggests that the GJB2 235delC polymorphism, but not the 30-35delG variant, contributes to congenital deafness susceptibility in the Chinese population examined. PMID: 28198501
  42. This report describes a non-consanguineous assortatively mating hearing impaired family with one of the hearing impaired partners, their hearing impaired sibling, and hearing impaired offspring showing compound heterozygosity in the GJB2 gene, involving a dominant mutation p.R184Q and two recessive mutations p.Q124X and c.IVS 1+1G>A in a unique triallelic combination. PMID: 27481527
  43. More than 39 deafness genes have been reported to cause non-syndromic hereditary hearing loss (HHL) in Iran, of which the most prevalent causative genes include GJB2, SLC26A4, MYO15A, and MYO7A. This review highlights some of the more common genetic causes of syndromic HHL in Iran. PMID: 27743438
  44. Two GJB2 mutations, c.del35G with an allele frequency of 4.7% and R32C (3.7%), were detected in Mauritanian children with non-syndromic hearing loss. PMID: 27067584
  45. Results showed that the GJB2 gene is a major contributor to non-syndromic hearing loss in Morocco. PMID: 27169813
  46. Compound heterozygous variants c.94C > T (p.R32C) and c.235delC (p.L79Cfs*3) in the GJB2 gene were identified in the two patients of an autosomal recessive non-syndromic hearing loss family, and the heterozygous GJB2 c.94C > T and c.235delC variants were identified in his unaffected father and mother, respectively. PMID: 27045574
  47. Mutations were detected in 35 of 156 deaf patients. PMID: 27066914
  48. GJB2 gene mutations are highly prevalent in pre-lingual hearing loss patients from China. 83.64% of the 330 patients carried variations in the GJB2 gene. Seventeen different genotypes were identified. A total of 31.2% of the patients carried two confirmed pathogenic mutations. The frequency of c.235delC was higher than that reported previously in the Jiangsu province. PMID: 27534436
  49. GJB2 gene mutations are the most common mutation for congenital hearing loss in Chinese newborns. PMID: 25649612
  50. 48.67% of the patients were identified with hereditary hearing loss caused by mutations in GJB2, SLC26A4, and mtDNA12SrRNA. PMID: 27247933

Show More

Hide All

Database Links

HGNC: 4284

OMIM: 121011

KEGG: hsa:2706

STRING: 9606.ENSP00000372295

UniGene: Hs.524894

Involvement In Disease
Deafness, autosomal recessive, 1A (DFNB1A); Deafness, autosomal dominant, 3A (DFNA3A); Vohwinkel syndrome (VOWNKL); Keratoderma, palmoplantar, with deafness (PPKDFN); Keratitis-ichthyosis-deafness syndrome (KID syndrome); Bart-Pumphrey syndrome (BPS); Ichthyosis hystrix-like with deafness syndrome (HID syndrome)
Protein Families
Connexin family, Beta-type (group I) subfamily
Subcellular Location
Cell membrane; Multi-pass membrane protein. Cell junction, gap junction.

Q&A

What is GJB2 protein and what is its primary function in cellular physiology?

GJB2 is a structural component of gap junctions, which are dodecameric channels that connect the cytoplasm of adjoining cells . These junctions are formed by the docking of two hexameric hemichannels, one from each cell membrane . GJB2 plays a crucial role in intercellular communication by facilitating the diffusion of small molecules and ions from one cell to neighboring cells via the central pore . In the auditory system specifically, GJB2 facilitates the recycling of potassium ions necessary for converting sound waves into electrical signals . Mutations in GJB2 are associated with hereditary hearing loss, emphasizing its physiological importance.

How should researchers select the appropriate GJB2 antibody for their specific experimental needs?

Selection should be based on:

ApplicationRecommended Antibody TypeKey Considerations
Western BlotPolyclonal (ab65969)Validated for 26 kDa band detection in rat liver lysate
IHC-P/IHC-FrRecombinant Monoclonal (ab303498)Superior specificity in tissue sections, validated for mouse/rat samples
Flow CytometryMonoclonal (CSB-RA009452MA1HU)Specifically validated for FC applications
Multi-species studiesConsider reactivity profileab65969 (human, rat), ab303498 (mouse, rat), CSB-RA009452MA1HU (human)

Consider the experimental model organism, application technique, and specific research question when selecting between available antibodies. Preliminary validation is recommended when applying antibodies to new research contexts.

What are the optimal sample preparation methods for detecting GJB2 using recombinant monoclonal antibodies in immunohistochemistry?

For frozen tissue sections:

  • Fix tissue in 4% PFA (paraformaldehyde)

  • Permeabilize with 0.2% Triton X-100 to enhance antibody accessibility

  • Use a dilution of 1:500 (0.974 μg/ml) for optimal results with antibodies like ab303498

  • Counterstain with DAPI for nuclear visualization

  • For fluorescence detection, secondary antibodies such as Goat Anti-Rabbit IgG H&L (Alexa Fluor® 488) at 1:1000 dilution (2 μg/mL) provide good results

For FFPE (formalin-fixed paraffin-embedded) tissues, antigen retrieval is essential due to potential masking of epitopes during fixation. Both heat-induced epitope retrieval (using citrate buffer pH 6.0) and enzymatic retrieval methods should be evaluated to determine optimal conditions for specific sample types.

How can researchers optimize Western blot protocols for GJB2 detection?

For optimal Western blot detection of GJB2:

  • Sample preparation:

    • Use RIPA buffer supplemented with protease inhibitors

    • Rat liver lysate serves as an excellent positive control

  • Electrophoresis and transfer:

    • Use 12-15% gels for optimal separation

    • Transfer to PVDF membranes (preferred over nitrocellulose for membrane proteins)

  • Antibody incubation:

    • Use anti-GJB2 antibody at 1 μg/mL concentration

    • Incubate overnight at 4°C for best results

  • Detection parameters:

    • Expected band size: 26 kDa (both predicted and observed)

    • Include molecular weight markers flanking the expected GJB2 band

  • Controls:

    • Include both positive tissue controls and negative controls (tissues lacking GJB2 expression)

    • Primary antibody omission controls to assess secondary antibody specificity

What methodological considerations are important for studying GJB2 in the context of auditory function research?

When investigating GJB2 in auditory research:

  • Tissue preparation considerations:

    • Cochlear tissue requires specialized fixation and decalcification protocols

    • Maintain consistent processing between experimental groups

  • Vector delivery approaches:

    • AAV vectors can be used for targeted GJB2 delivery in research models

    • Cell-specific promoters like GREs (Genomic Regulatory Elements) provide more precise expression compared to ubiquitous promoters like CBA

    • Round window membrane injection is an effective delivery route for cochlear studies

  • Functional assessments:

    • Combine morphological studies with functional measurements (e.g., auditory brainstem responses, DPOAEs)

    • Design longitudinal studies as GJB2 function may change over developmental timepoints

  • Safety considerations:

    • Ubiquitous expression of GJB2 can be lethal in animal models, suggesting cell-specific targeting is critical

    • Consider using inducible expression systems for temporal control

What are common causes of non-specific binding when using GJB2 recombinant monoclonal antibodies, and how can they be mitigated?

Common causes of non-specific binding include:

IssueMitigation Strategy
Insufficient blockingExtend blocking time (1-2 hours) with 5-10% serum from secondary antibody species
High antibody concentrationPerform titration experiments to determine optimal concentration
Cross-reactivity with related connexinsUse antibodies validated against multiple connexin family members
Endogenous peroxidases (for IHC)Pre-treat with hydrogen peroxide (0.3-3%) before antibody incubation
High background in specific tissuesConsider tissue-specific blocking agents (e.g., mouse-on-mouse blocking for mouse tissues)

When evaluating specificity, include appropriate negative controls such as isotype controls and primary antibody omission controls. Tissue samples known to lack GJB2 expression can serve as biological negative controls.

How should researchers validate new lots of GJB2 recombinant monoclonal antibodies?

Comprehensive validation includes:

  • Side-by-side comparison with previous antibody lot using identical protocols

  • Positive control testing:

    • Western blot: verify correct molecular weight band (26 kDa) in rat liver lysate

    • IHC: confirm expected staining pattern in known GJB2-expressing tissues

  • Specificity controls:

    • Test on both positive (e.g., cochlea, liver) and negative control tissues

    • Include blocking peptide competition assays when possible

  • Application-specific validation:

    • For IHC: test with both frozen and FFPE samples if both will be used

    • For Western blot: verify under both reducing and non-reducing conditions

    • For Flow cytometry: confirm specificity using cells with known GJB2 expression levels

  • Documentation:

    • Record lot number, validation date, and experimental conditions

    • Archive validation results for future reference and troubleshooting

What approaches can researchers use to address weak or inconsistent GJB2 signal?

When facing weak or variable GJB2 signal:

  • Epitope availability:

    • Test multiple antigen retrieval methods and conditions

    • Consider alternative fixation protocols that better preserve GJB2 epitopes

  • Signal amplification strategies:

    • Tyramide signal amplification (TSA) can enhance detection sensitivity

    • Biotin-streptavidin systems provide amplification but may increase background

  • Antibody concentration and incubation:

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

    • Higher antibody concentrations should be tested systematically

  • Expression level considerations:

    • GJB2 expression varies between tissues and developmental stages

    • Confirm expected expression levels in your experimental system

  • Technical optimization:

    • Fresh antibody aliquots to avoid freeze-thaw degradation

    • Optimize blocking and washing conditions to improve signal-to-noise ratio

How can GJB2 recombinant monoclonal antibodies be utilized for studying auditory development and disorders?

Advanced applications include:

  • Developmental studies:

    • Track GJB2 expression patterns during cochlear development

    • Correlate expression with functional hearing assessments

  • Disease modeling:

    • Compare GJB2 localization and expression in normal vs. disease models

    • Evaluate therapeutic interventions targeting GJB2 restoration

  • Gene therapy assessment:

    • Validate viral vector-mediated GJB2 expression in knockout models

    • Quantify restoration of cell-specific GJB2 expression patterns

    • Use GJB2 antibodies to confirm proper localization of therapeutically delivered protein

  • Structure-function relationships:

    • Combine GJB2 immunolabeling with functional assays of gap junction activity

    • Correlate changes in GJB2 distribution with auditory function measurements

  • Co-localization studies:

    • Investigate GJB2 interactions with other connexins and gap junction components

    • Study subcellular trafficking and assembly of gap junction plaques

What considerations are important when designing multiplexed immunolabeling experiments involving GJB2?

For successful multiplexed studies:

  • Primary antibody selection:

    • Choose primary antibodies raised in different host species

    • Verify each antibody individually before multiplexing

  • Fluorophore selection:

    • Use fluorophores with minimal spectral overlap

    • Consider brightness characteristics relative to target abundance

  • Sequential labeling strategy:

    • For same-species antibodies, consider sequential immunolabeling with blocking steps

    • Tyramide signal amplification allows same-species antibody multiplexing

  • Controls for multiplexed imaging:

    • Single-antibody controls to assess bleed-through

    • Secondary antibody-only controls to evaluate non-specific binding

  • Analysis approaches:

    • Use specialized software for co-localization analysis (e.g., JACoP plugin for ImageJ)

    • Implement quantitative metrics (Pearson's coefficient, Manders' coefficient) for objective assessment

How can researchers approach studying post-translational modifications of GJB2 using antibody-based methods?

For studying GJB2 post-translational modifications:

  • Specialized antibodies:

    • Use modification-specific antibodies (phospho-specific, ubiquitin-specific)

    • Combine with general GJB2 antibodies to determine modified fraction

  • Enrichment strategies:

    • Immunoprecipitate GJB2 using recombinant monoclonal antibodies

    • Probe with modification-specific antibodies on Western blots

  • Experimental manipulations:

    • Pharmacological induction/inhibition of specific modifications

    • Genetic manipulation of modification sites through mutagenesis

  • Sample preparation considerations:

    • Include phosphatase inhibitors when studying phosphorylation

    • Add deubiquitinase inhibitors when studying ubiquitination

    • Consider rapid extraction methods to preserve labile modifications

  • Advanced analysis:

    • Combine immunoprecipitation with mass spectrometry for comprehensive modification mapping

    • Use proximity ligation assays to detect specific modified forms in situ

What approaches should researchers use to quantify GJB2 expression levels across different experimental conditions?

ApplicationQuantification MethodKey Considerations
Western BlotDensitometryNormalize to appropriate loading controls; use linear range of detection
IHC/IFFluorescence intensity measurementControl for acquisition parameters; analyze multiple fields per sample
Flow CytometryMean/median fluorescence intensityUse isotype controls; analyze population shifts
qPCRRelative expression analysisSelect stable reference genes; correlate with protein levels

Statistical analysis should include:

  • Appropriate sample sizes (minimum n=3 biological replicates)

  • Tests for normality to determine parametric vs. non-parametric analysis

  • Correction for multiple comparisons when analyzing multiple conditions

  • Consideration of inter-individual variation in tissue samples

How can researchers interpret discrepancies in GJB2 localization patterns between different antibodies?

When facing localization discrepancies:

  • Epitope considerations:

    • Different antibodies target distinct epitopes that may be differentially accessible

    • Some epitopes may be masked by protein interactions or conformational states

  • Methodological approach:

    • Compare fixation and permeabilization effects on epitope accessibility

    • Evaluate antibody specificity using knockout controls when possible

  • Biological explanations:

    • GJB2 undergoes trafficking between different subcellular compartments

    • Post-translational modifications may affect epitope recognition

  • Resolution and detection:

    • Consider limitations of optical resolution vs. electron microscopy

    • Super-resolution techniques provide more accurate localization information

  • Validation strategies:

    • Use complementary approaches (fractionation plus Western blot)

    • Correlate antibody localization with tagged GJB2 constructs

How should researchers integrate GJB2 antibody-based findings with genetic and functional data?

For comprehensive interpretation:

  • Correlation analysis:

    • Compare protein expression (antibody-based) with mRNA levels (transcriptomics)

    • Assess how genetic variants impact protein expression and localization

  • Structure-function relationships:

    • Correlate GJB2 expression patterns with functional measures

    • In auditory research, link GJB2 distribution to hearing thresholds

  • Intervention studies:

    • Use antibodies to confirm successful rescue in gene therapy approaches

    • Quantify how interventions restore normal GJB2 patterns

  • Development of an integrated model:

    • Combine protein localization, expression level, and functional data

    • Account for cell-type specific effects and developmental timing

  • Contextual interpretation:

    • Consider physiological context and regulatory networks

    • Recognize limitations of each methodological approach

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.