BHLH90 Antibody

Shipped with Ice Packs
In Stock

Description

Absence in Established Antibody Repositories

None of the major antibody characterization initiatives referenced in the materials mention BHLH90:

  • NeuroMab (UC Davis) catalog: 800+ neural targets

  • Developmental Studies Hybridoma Bank (DSHB): 6,000+ hybridoma lines

  • CPTAC Antibody Portal: 946 cancer-related antibodies

  • Therapeutic antibody registries (WHO INN, FDA databases)

Potential Explanations for the Discrepancy

ScenarioProbabilityRecommended Action
Typographical errorHighVerify spelling against known antibodies (e.g., BL-CAM, HLH-1)
Undisclosed research compoundMediumConsult clinicaltrials.gov or preclinical trial registries
Proprietary therapeutic candidateLowReview recent patent filings (USPTO, WIPO)
Non-standard abbreviationHighRequest full chemical name/sequence

Alternative Research Pathways

If investigating basic helix-loop-helix (bHLH) transcription factors (a possible misinterpretation of "BHLH"):

  1. Related Antibodies: MYOD1 (bHLH myogenic factor), TWIST1 (bHLH transcription regulator)

  2. Commercial Alternatives:

    • Anti-HES1 (IgG2aκ, Invitrogen MA5-32616)

    • Anti-NEUROD1 (IgG1κ, Abcam ab60704)

Verification Protocol

To confirm the existence of BHLH90 Antibody:

  1. Sequence Analysis: Request full amino acid sequence (heavy/light chains)

  2. Epitope Mapping: Validate target antigen via SPR/BLI

  3. Functional Testing: Reproduce results in ≥2 independent labs

  4. Database Crosscheck:

    • NCBI Protein (XP_XXXXXXX)

    • UniProt (QXXXXX)

    • EMBL-EBI ABCam

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
BHLH90 antibody; EN50 antibody; At1g10610 antibody; F20B24.4 antibody; F20B24.5 antibody; T10O24.26Transcription factor bHLH90 antibody; Basic helix-loop-helix protein 90 antibody; AtbHLH90 antibody; bHLH 90 antibody; Transcription factor EN 50 antibody; bHLH transcription factor bHLH090 antibody
Target Names
BHLH90
Uniprot No.

Target Background

Database Links

KEGG: ath:AT1G10610

STRING: 3702.AT1G10610.1

UniGene: At.42172

Subcellular Location
Nucleus.
Tissue Specificity
Expressed constitutively in roots, leaves, stems, and flowers.

Q&A

What is BHLHB9 and why is it significant for research?

BHLHB9 (Basic Helix-Loop-Helix Domain Containing, Class B, 9) is a protein of interest in multiple research areas. The significance lies in its helix-loop-helix domain structure, which is characteristic of a family of transcription factors involved in various developmental and regulatory processes. When designing experiments utilizing BHLHB9 antibodies, researchers should consider the specific isoforms and structural domains they wish to target. For instance, antibodies targeting amino acids 451-547 of the BHLHB9 protein provide specificity for the C-terminal region, which may be particularly relevant depending on your experimental goals . A methodological approach would involve preliminary Western blot validation to confirm the antibody recognizes your target of interest before proceeding to more complex applications.

How does antibody conjugation affect experimental design with BHLHB9 antibodies?

The conjugation of BHLHB9 antibodies to fluorescent markers like FITC directly impacts experimental design and data interpretation. With FITC-conjugated antibodies, researchers can perform direct detection without secondary antibodies, which simplifies protocols and reduces background in multi-labeling experiments . When planning experiments, consider that FITC has an excitation maximum at approximately 495nm and emission at 519nm, which may influence your choice of other fluorophores to avoid spectral overlap. Additionally, FITC is sensitive to photobleaching and pH conditions, necessitating appropriate storage (protected from light at 2-8°C) and experimental controls. For quantitative analyses, include calibration standards and consider that fluorescence intensity may not maintain linearity at very high or low signal ranges.

What are the optimal conditions for Western blotting with BHLHB9 antibodies?

For Western blotting with BHLHB9 antibodies, optimization begins with sample preparation. Tissue homogenization should be performed in RIPA buffer supplemented with protease inhibitors, followed by centrifugation at 14,000×g for 20 minutes at 4°C to remove cellular debris. For protein separation, 10-12% polyacrylamide gels typically provide optimal resolution for BHLHB9 detection. When transferring to membranes, PVDF is often preferred over nitrocellulose due to its higher protein binding capacity and mechanical strength. For the rabbit polyclonal anti-BHLHB9 antibody, blocking with 5% non-fat dry milk in TBST (pH 7.4) for 1 hour at room temperature helps minimize non-specific binding . Primary antibody dilutions typically range from 1:500 to 1:2000, with overnight incubation at 4°C yielding optimal results. Include positive controls (tissues known to express BHLHB9) and negative controls (tissues with low or no expression) to validate specificity.

How should immunofluorescence protocols be optimized for BHLHB9 antibody applications?

Immunofluorescence with BHLHB9 antibodies requires careful optimization of fixation, permeabilization, and detection parameters. For cultured cells, 4% paraformaldehyde fixation for 15 minutes at room temperature preserves both antigenicity and cellular architecture. Permeabilization with 0.2% Triton X-100 for 10 minutes enables antibody access to intracellular targets while minimizing structural disruption . For paraffin-embedded sections, antigen retrieval is critical—10mM citrate buffer (pH 6.0) with 20-minute heat-induced epitope retrieval generally yields optimal results. When using FITC-conjugated BHLHB9 antibodies, dilutions between 1:50 and 1:200 in antibody diluent containing 1% BSA typically provide optimal signal-to-noise ratios. Counterstaining nuclei with DAPI (1μg/ml) aids in cellular localization, but avoid mounting media containing anti-fade agents that might quench FITC fluorescence. Include absorption controls (pre-incubation of antibody with immunizing peptide) to verify specificity of staining patterns.

What cross-reactivity considerations are important when working with BHLHB9 antibodies?

Cross-reactivity analysis is essential for accurate data interpretation when working with BHLHB9 antibodies. The rabbit polyclonal antibody targeting amino acids 451-547 demonstrates confirmed reactivity with mouse BHLHB9, while exhibiting predicted cross-reactivity with human, rat, dog, cow, sheep, pig, and horse BHLHB9 orthologs . When designing experiments across species, preliminary validation through Western blotting is recommended to confirm cross-reactivity. If unexpected bands appear, consider performing peptide competition assays or utilizing mass spectrometry to identify potentially cross-reactive proteins. The sequence homology between target regions should be assessed through bioinformatic analysis (BLAST alignment) when extending applications to predicted species. For critical experiments, especially in species without confirmed reactivity, parallel validation with multiple antibodies targeting different BHLHB9 epitopes provides stronger evidence of specificity.

How can bispecific antibody technology be applied to BHLHB9 research?

Bispecific antibody technology presents advanced opportunities for BHLHB9 research by enabling simultaneous targeting of BHLHB9 and another protein of interest. When designing bispecific antibodies incorporating BHLHB9 binding domains, researchers must consider molecular geometry, fusion site selection, and optimal linker design . For symmetric bispecific formats, fusion of exogenous antigen-binding domains onto IgG scaffolds offers a modular approach, with glycine-serine linkers of 10-25 amino acids providing favorable flexibility and stability . For optimal performance, the choice between symmetric formats (HC₂LC₂) and asymmetric designs depends on experimental goals—symmetric formats reduce misassembly risks but limit valency flexibility, while asymmetric formats allow more versatile antigen targeting but require careful optimization of chain pairing . When proceeding with bispecific BHLHB9 antibody development, incorporate early developability screening (thermal stability, aggregation propensity, expression yield) to identify candidates most likely to succeed in complex research applications.

What strategies can address antibody mispairing challenges in complex BHLHB9 antibody engineering?

Engineering complex BHLHB9 antibodies, particularly bispecific formats, requires addressing heavy chain-light chain (HC:LC) mispairing challenges. Advanced researchers can implement several strategic approaches: (1) Utilize inherent preferential cognate HC:LC pairing by selecting Fab domains with demonstrated pairing specificity largely determined by complementarity-determining regions (CDRs) ; (2) Replace one or both Fabs with single-chain fragments (scFv) or single-domain antibodies (sdAbs) to reduce the number of separate chains requiring assembly ; (3) Employ post-expression assembly methods where each antibody half is expressed and purified separately before controlled combination through reduction and oxidation of hinge disulfides ; (4) Implement advanced analytics (hydrophobic interaction chromatography, mass spectrometry) to identify and quantify mispaired species. For BHLHB9 applications requiring maximum specificity, consider leveraging chemical processing strategies such as leucine zipper-controlled association or click chemistry for precise assembly control .

How can non-specific binding be minimized when using BHLHB9 antibodies?

Non-specific binding represents a common challenge when working with BHLHB9 antibodies. A systematic approach to minimization includes: (1) Optimize blocking conditions—for Western blotting, test various blocking agents (5% BSA, 5% non-fat dry milk, commercial blockers) to identify optimal formulations; (2) Titrate antibody concentrations—perform dilution series to determine the minimum concentration yielding specific signal ; (3) Modify incubation parameters—shorter incubation times at room temperature may reduce non-specific interactions compared to overnight incubations; (4) Incorporate detergents—adding 0.1-0.3% Tween-20 to wash and antibody dilution buffers disrupts weak, non-specific interactions; (5) Pre-adsorb antibodies—incubate diluted antibody with tissues/cells lacking BHLHB9 expression to remove cross-reactive antibodies. For FITC-conjugated antibodies specifically, include an additional control using isotype-matched FITC-conjugated non-specific IgG to establish baseline fluorescence levels. Document optimization steps methodically to ensure reproducibility across experiments.

What quality control measures should be implemented when working with BHLHB9 antibodies?

Rigorous quality control is essential for reliable BHLHB9 antibody research. Implement a multi-faceted approach including: (1) Lot-to-lot validation—test each new antibody lot against a reference standard to ensure consistent performance; (2) Specificity testing—employ peptide competition assays where pre-incubation with the immunizing peptide should abolish specific binding ; (3) Positive and negative controls—include tissues/cells with known BHLHB9 expression profiles in each experiment; (4) Orthogonal validation—confirm key findings using alternative detection methods (e.g., validate IF results with Western blotting); (5) Antibody storage validation—assess performance before and after storage to determine stability. For FITC-conjugated antibodies, additional quality control includes measuring fluorophore-to-protein ratio (ideally 3-7 fluorophores per antibody) and monitoring photobleaching rates. Maintain detailed records of antibody performance parameters, including optimal dilutions, signal intensity, and background levels across applications to establish internal reference standards.

How should researchers validate BHLHB9 antibody specificity across different species?

Cross-species validation of BHLHB9 antibodies requires a structured approach to ensure reliable results. Begin with bioinformatic analysis comparing the immunogen sequence (human BHLHB9/p60TRP amino acids 451-547) with orthologous sequences across target species to predict conservation . For experimental validation in species with predicted reactivity (human, rat, dog, cow, sheep, pig, horse), implement a staged process: (1) Perform Western blotting on tissue lysates from each species, comparing band patterns and molecular weights; (2) Confirm specificity through immunoprecipitation followed by mass spectrometry to identify pulled-down proteins; (3) Conduct immunohistochemistry with appropriate positive and negative tissue controls for each species; (4) Where possible, utilize genetic approaches (siRNA knockdown, CRISPR knockout) to confirm antibody specificity. When discrepancies arise between predicted and observed cross-reactivity, consider post-translational modifications or splice variants that might affect epitope availability. Document species-specific optimization parameters (antibody dilution, incubation conditions) as these may vary significantly even with confirmed cross-reactivity.

How can advanced imaging techniques enhance BHLHB9 antibody applications?

Advanced imaging methodologies significantly expand the analytical capabilities of BHLHB9 antibody applications. Super-resolution microscopy techniques—including Structured Illumination Microscopy (SIM), Stimulated Emission Depletion (STED), and Stochastic Optical Reconstruction Microscopy (STORM)—overcome the diffraction limit of conventional microscopy, enabling visualization of BHLHB9 subcellular localization with up to 20nm resolution . When implementing these techniques, FITC-conjugated BHLHB9 antibodies require specific considerations: for STED microscopy, photobleaching resistance becomes critical, potentially necessitating alternative fluorophores like Alexa Fluor dyes; for STORM imaging, buffer composition must be optimized for FITC blinking behavior. Multiplexed imaging approaches, such as Imaging Mass Cytometry or CO-Detection by indEXing (CODEX), allow simultaneous detection of BHLHB9 alongside dozens of other proteins without spectral overlap limitations. When designing advanced imaging experiments, incorporate computational analysis workflows (machine learning-based segmentation, colocalization analysis) to extract quantitative data from complex image datasets.

What developments in antibody engineering might impact future BHLHB9 research?

Emerging antibody engineering technologies present transformative opportunities for BHLHB9 research. CRISPR-based display platforms facilitate rapid selection of high-affinity antibodies against conformational epitopes, potentially yielding BHLHB9 antibodies with enhanced specificity for distinct protein states . Computational antibody design is increasingly enabling in silico optimization of binding domains before experimental validation, reducing development timelines and enhancing performance characteristics . For complex research questions, designed bispecific antibodies simultaneously targeting BHLHB9 and interaction partners could elucidate protein-protein interactions in intact cellular systems. The integration of non-natural amino acids into antibody structures offers opportunities for site-specific conjugation, enabling precise control over fluorophore positioning to optimize FRET-based applications or proximity assays . Nanobody and single-domain antibody technologies provide compact alternatives with superior tissue penetration and stability, particularly valuable for intravital imaging of BHLHB9. Researchers should evaluate these emerging technologies based on specific experimental requirements, recognizing that established techniques may remain optimal for many applications despite the allure of novel approaches.

How might recent discoveries in antibody binding mechanisms inform BHLHB9 antibody development?

Recent structural and mechanistic insights into antibody-antigen interactions are reshaping BHLHB9 antibody development strategies. Cryo-electron microscopy has revealed previously uncharacterized binding modes, including asymmetric engagement and induced conformational changes, which can be leveraged to design BHLHB9 antibodies with enhanced specificity or function-modulating capabilities . High-throughput screening approaches combining yeast display with next-generation sequencing enable comprehensive epitope mapping, facilitating the development of antibody panels targeting distinct BHLHB9 functional domains . Understanding the spatial segregation of complementarity-determining regions (CDRs) has led to innovative formats like DutaFab, where CDRs are strategically distributed between VH and VL domains to create bispecific recognition within a single Fab domain—a promising approach for targeting BHLHB9 alongside interaction partners . For researchers developing new BHLHB9 antibodies, integrated computational-experimental pipelines incorporating these mechanistic insights can accelerate development while enhancing performance characteristics. When evaluating candidate antibodies, consider not only binding affinity but also binding kinetics, epitope accessibility in native protein conformations, and capacity to distinguish between closely related protein family members.

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.