BHLH76 Antibody

Shipped with Ice Packs
In Stock

Description

Possible Explanations for the Absence of "BHLH76 Antibody" in Literature

  • Nomenclature Variations:

    • BHLH76 may refer to a protein or gene not widely studied in the context of antibody development (e.g., BHLH76 is a basic helix-loop-helix transcription factor, but no antibodies targeting it are documented in the provided sources).

    • The term may represent a proprietary or experimental compound not yet published in peer-reviewed literature.

  • Target Specificity:

    • Antibodies targeting transcription factors like BHLH proteins are less common due to challenges in accessing intracellular targets. Most therapeutic antibodies target extracellular proteins (e.g., cell surface receptors, cytokines).

  • Data Gaps:

    • The provided sources focus on well-characterized antibodies (e.g., anti-TNFα, anti-VEGF, anti-IL-17) and broadly neutralizing antibodies (e.g., HIV, COVID-19). No studies mention BHLH76 as a target.

Recommendations for Further Research

To address gaps in information, consider the following steps:

ActionDetails
Verify NomenclatureConfirm the correct spelling and context of "BHLH76." Cross-reference with gene/protein databases (e.g., UniProt, NCBI Gene).
Explore Preclinical StudiesSearch platforms like PubMed or ClinicalTrials.gov using keywords such as "BHLH76 monoclonal antibody" or "BHLH76-targeted therapy."
Consult Proprietary DatabasesCheck antibody catalogs from companies like Sigma-Aldrich (source ) or Cell Signaling Technology (source ) for unlisted products.
Review Transcription Factor AntibodiesInvestigate antibodies targeting similar BHLH family members (e.g., BHLH2, BHLHE41) to infer potential mechanisms.

Contextual Insights from Related Antibodies

While no data on BHLH76 exists, insights can be drawn from antibodies targeting transcription factors or intracellular proteins:

Challenges in Targeting Intracellular Proteins

  • Mechanistic Limitations: Antibodies typically bind to extracellular targets. Intracellular proteins like transcription factors require alternative delivery strategies (e.g., antibody-drug conjugates, cell-penetrating peptides).

  • Examples:

    • B7-H6 Antibody: Targets a tumor-associated antigen on cell surfaces (source ).

    • IL-17A/F Antibody (Bimekizumab): Neutralizes extracellular cytokines (source ).

Emerging Technologies for Intracellular Targeting

  • Bispecific Antibodies: Redirect immune cells (e.g., T cells) to kill target cells (e.g., B7-H6/CD3 ITE in source ).

  • CRISPR-Edited B Cells: Generate humanized antibodies with enhanced affinity (source ).

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
BHLH76 antibody; CIB5 antibody; EN83 antibody; At1g26260 antibody; F28B23.8Transcription factor bHLH76 antibody; Basic helix-loop-helix protein 76 antibody; AtbHLH76 antibody; bHLH 76 antibody; Protein CRYPTOCHROME INTERACTING BASIC-HELIX-LOOP-HELIX 5 antibody; Transcription factor EN 83 antibody; bHLH transcription factor bHLH076 antibody
Target Names
BHLH76
Uniprot No.

Target Background

Function
BHLH76 Antibody is a transcriptional activator that plays a crucial role in cell elongation. It regulates the expression of a specific set of genes involved in cell expansion by binding to the G-box motif. This antibody interacts with the chromatin DNA of the FT gene, promoting its expression and ultimately triggering flowering in response to blue light.
Database Links

KEGG: ath:AT1G26260

STRING: 3702.AT1G26260.1

UniGene: At.41276

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

Q&A

What validation methods should be used to confirm BHLH76 antibody specificity?

Validation of antibody specificity is crucial for reliable experimental outcomes. For BHLH76 antibody validation, researchers should implement multiple complementary approaches. Begin with immunoblotting to confirm antibody recognition of the target protein at the expected molecular weight. Flow cytometry can verify binding to native protein in intact cells, while immunoprecipitation confirms interaction with the target protein in solution . For definitive validation, include controls such as BHLH76 knockout or knockdown samples. Results from multiple techniques should be consistent to establish high confidence in antibody specificity .

How should optimal BHLH76 antibody concentration be determined for different applications?

Determining optimal antibody concentration requires systematic titration for each application. For immunoblotting, perform a dilution series (typically 1:500 to 1:5000) to identify the concentration that provides the strongest specific signal with minimal background. For flow cytometry, prepare cell samples with antibody dilutions ranging from 1:50 to 1:500 and analyze signal-to-noise ratios . For immunoprecipitation, test concentrations between 1-10 μg per sample. Document all optimization results in a standardized format to ensure reproducibility across experiments. Remember that optimal concentration may vary between different experimental conditions and sample types .

What storage conditions maximize BHLH76 antibody stability and performance?

To maintain antibody integrity, divide stock solutions into small aliquots (20-50 μl) to minimize freeze-thaw cycles, which can cause protein denaturation and diminished activity . Store aliquots at -20°C for short-term or -80°C for long-term preservation. Working dilutions can be stored at 4°C with preservatives like 0.1% sodium azide but should be used within 1-2 weeks. Monitor antibody performance over time using consistent positive controls to detect any degradation. For applications requiring maximum sensitivity, use fresh aliquots of antibody rather than repeatedly thawed samples .

How can cross-reactivity issues with BHLH76 antibody be identified and mitigated?

Cross-reactivity assessment is essential for accurate data interpretation. Perform preliminary screening using tissue or cell panels with varying BHLH76 expression levels, including negative controls where the target is absent. Western blotting can reveal unexpected bands indicating cross-reactivity with other proteins. For advanced characterization, consider epitope mapping to understand potential cross-reactive regions . If cross-reactivity occurs, mitigation strategies include using more stringent washing conditions, optimizing blocking reagents, pre-absorbing the antibody with cross-reactive proteins, or implementing computational analysis to distinguish specific from non-specific binding patterns in complex datasets .

How can bioinformatic approaches enhance BHLH76 antibody epitope characterization?

Bioinformatic analysis can provide critical insights into antibody-epitope interactions. Begin with in silico prediction of BHLH76 protein structure using tools like AlphaFold to identify surface-exposed regions likely to serve as antibody binding sites . Compare epitope conservation across species using multiple sequence alignments to predict cross-reactivity. Molecular dynamics simulations can reveal conformational epitopes that may not be evident in static models. Integration of experimental epitope mapping data with computational predictions creates a comprehensive epitope profile, guiding applications where epitope accessibility may vary (e.g., detecting denatured versus native protein) .

What strategies can resolve contradictory results between different detection methods using BHLH76 antibody?

When facing contradictory results, implement a systematic troubleshooting approach. First, verify antibody integrity and experimental conditions by repeating experiments with fresh reagents and standardized protocols. Consider epitope accessibility differences between methods—certain fixation procedures may mask epitopes recognized by your antibody . Explore whether post-translational modifications affect antibody recognition in different sample preparations. Implement alternative antibodies targeting different BHLH76 epitopes to confirm findings. Combine antibody-based detection with antibody-independent methods such as mass spectrometry or nucleic acid-based detection to resolve discrepancies .

How can BHLH76 antibody be effectively used in antibody-dependent cellular cytotoxicity (ADCC) assays?

For ADCC applications, antibody format selection is critical. Humanized formats with intact Fc regions, similar to FF-01 described in lymphoma studies, optimize interaction with immune effector cells . Begin assay development by confirming target binding using flow cytometry with cell lines expressing variable levels of BHLH76. For the ADCC assay, prepare target cells expressing BHLH76 at physiologically relevant levels and incubate with the antibody at concentrations from 0.1-10 μg/ml before adding effector cells (typically PBMCs or NK cells) . Measure cytotoxicity using methods like LDH release or flow cytometry-based viability assays. Include controls with isotype-matched antibodies and target-negative cell lines to establish specificity of the observed ADCC effect .

What parameters should be optimized when developing internalization assays for BHLH76 antibody?

Antibody internalization assays require careful optimization of multiple parameters. Begin by confirming cell surface binding using flow cytometry at 4°C (which prevents internalization). For internalization studies, switch to 37°C incubation and assess time-dependent uptake at intervals from 15 minutes to 24 hours . pH-sensitive fluorophores can distinguish membrane-bound from internalized antibodies. Confocal microscopy with co-localization markers for endosomes and lysosomes can track the intracellular fate of internalized antibodies. Quantitative analysis should measure both the rate and efficiency of internalization, comparing high and low BHLH76-expressing cell lines to establish expression-dependent internalization kinetics .

How can RNA-seq data be integrated with BHLH76 antibody-based protein detection to enhance experimental rigor?

Integrating transcriptomics with protein-level detection creates a powerful multi-omics approach. Begin by establishing baseline correlation between BHLH76 mRNA expression from RNA-seq data and protein levels detected by your antibody across a panel of relevant tissues or cell lines . When discrepancies occur, investigate potential post-transcriptional regulation mechanisms. For complex samples like tissue biopsies, single-cell RNA-seq can identify specific cell populations expressing BHLH76 mRNA, which can then be targeted for antibody-based protein detection using techniques like imaging mass cytometry or multiplexed immunofluorescence . This integration allows for validation of antibody specificity while providing contextual information about BHLH76 expression patterns across different cell types .

How should BHLH76 antibody performance be assessed in different fixation and embedding conditions?

Fixation methods significantly impact epitope accessibility and antibody performance. Implement a systematic evaluation across multiple conditions: fresh-frozen tissues, paraformaldehyde fixation (typically 2-4%), formalin fixation with paraffin embedding (FFPE), and alternative fixatives like methanol or acetone . For each condition, perform standardized immunostaining with appropriate antigen retrieval methods (heat-induced or enzymatic). Document staining intensity, background levels, and subcellular localization patterns. Consider chromatin immunoprecipitation (ChIP) requirements separately, as they may demand specialized crosslinking procedures. Create a comprehensive performance matrix to guide future application-specific protocol selection .

What strategies can enhance BHLH76 antibody performance in challenging samples with high background?

High background represents a common challenge requiring multi-faceted optimization. Begin by testing different blocking agents (BSA, serum, commercial blocking buffers) at various concentrations and incubation times . Implement more stringent washing procedures, potentially incorporating detergents like Tween-20 or Triton X-100 at concentrations from 0.05-0.3%. For tissue samples with endogenous biotin or peroxidase activity, include specific blocking steps. Consider signal amplification methods like tyramide signal amplification for detecting low-abundance targets while maintaining favorable signal-to-noise ratios . For fluorescence applications, include autofluorescence quenching steps and select fluorophores that avoid spectral overlap with sample autofluorescence. Document optimization steps systematically to establish reproducible protocols for challenging sample types .

How can BHLH76 antibody be incorporated into multiplex detection systems?

Multiplex systems require careful antibody panel design. First, verify BHLH76 antibody compatibility with multiplexing fixation and staining protocols using single-stain controls . For immunofluorescence multiplexing, select antibodies from different host species or isotypes to enable specific secondary detection. Consider sequential staining with intermittent antibody stripping or quenching for same-species antibodies. For mass cytometry approaches, conjugate BHLH76 antibody with rare earth metals following validated conjugation protocols and verify retention of binding properties post-conjugation . For all multiplex systems, conduct comprehensive controls including fluorescence minus one (FMO) or metal minus one (MMO) controls to accurately identify positive populations and assess spillover between channels .

What approach should be taken to quantify absolute BHLH76 protein levels using antibody-based techniques?

Absolute quantification requires rigorous standardization. Develop a calibration curve using recombinant BHLH76 protein at known concentrations subjected to identical processing as experimental samples . For ELISA-based quantification, implement a sandwich format using capture and detection antibodies recognizing different BHLH76 epitopes to maximize specificity. For immunoblotting quantification, include internal loading controls and analyze band intensities using calibrated densitometry software. Consider mass spectrometry with isotope-labeled peptide standards as an orthogonal validation method . Account for extraction efficiency by spiking known quantities of recombinant protein into negative samples prior to processing. Report results with appropriate uncertainty metrics, including both technical and biological variation .

How should batch-to-batch variability in BHLH76 antibody performance be systematically assessed and managed?

Implement a comprehensive quality control program to maintain experimental consistency. Upon receiving new antibody batches, perform side-by-side comparison with previous lots using standardized positive controls . Document key performance metrics including specific signal intensity, background levels, and detection thresholds across all relevant applications. Consider creating a batch calibration factor if complete consistency cannot be achieved. Maintain reference samples (e.g., lysates from cells expressing BHLH76 at known levels) that can be used for normalization across experiments using different antibody batches . For critical long-term studies, consider purchasing sufficient antibody from a single lot or developing standardized validation protocols that can qualify new batches for specific applications .

What steps should be taken when BHLH76 antibody shows unexpected subcellular localization patterns?

Unexpected localization requires careful validation to distinguish genuine biological findings from technical artifacts. First, confirm the observation using multiple microscopy techniques and fixation methods . Test alternative BHLH76 antibodies targeting different epitopes to determine if the localization is epitope-specific. Perform subcellular fractionation followed by immunoblotting to biochemically validate microscopy observations. Use fluorescently-tagged BHLH76 expression constructs as independent confirmation of localization patterns in live cells . Consider whether experimental conditions might trigger non-canonical localization (e.g., stress responses, cell cycle stages). Comprehensive controls and orthogonal validation are essential before reporting novel localization findings .

What strategies can resolve non-specific binding issues with BHLH76 antibody in flow cytometry applications?

Non-specific binding in flow cytometry requires systematic troubleshooting. Begin by optimizing the blocking protocol, testing various blocking agents (human serum, FcR blocking reagents, BSA) at different concentrations and incubation times . Implement more stringent washing steps using buffers with optimized detergent concentrations. Consider pre-absorbing the antibody with cells lacking BHLH76 expression to remove cross-reactive antibodies. Test different antibody concentrations to identify the optimal signal-to-noise ratio. For multicolor panels, address potential fluorophore interactions through careful panel design and compensation . Compare results from multiple antibody clones or formats targeting BHLH76 to identify those with optimal performance characteristics for flow cytometry applications .

How should discrepancies between antibody-based detection and functional assays for BHLH76 be investigated?

Discrepancies between detection and function often reveal important biological insights. Begin by verifying antibody detection specificity through knockdown or knockout controls . Consider whether post-translational modifications might affect antibody binding without altering protein function, or conversely, whether functional changes might occur without detectable expression differences. Examine temporal dynamics—protein expression and function may not be perfectly synchronized. For transcription factors like BHLH76, assess nuclear localization and DNA binding activity separately from simple expression levels . Implement dose-response studies in functional assays to determine threshold concentrations required for activity. Integration of proteomics approaches like mass spectrometry can provide orthogonal validation and potentially identify protein isoforms or modifications that reconcile observed discrepancies .

How can BHLH76 antibody be effectively utilized in single-cell protein analysis technologies?

Single-cell protein analysis demands specialized optimization. For mass cytometry (CyTOF), conjugate BHLH76 antibody with rare earth metals using validated protocols and verify signal intensity correlates with expected expression patterns across control cell populations . For microfluidic antibody capture techniques, optimize antibody concentration and capture duration to maximize sensitivity while maintaining specificity. When adapting BHLH76 antibody for single-cell Western blotting, determine optimal cell lysis conditions and antibody concentrations for miniaturized formats . For all single-cell applications, implement robust controls including spike-in standards at known concentrations. Validate findings by correlating single-cell protein data with single-cell RNA-seq of the same populations when possible .

What considerations apply when developing BHLH76 antibody-based proximity ligation assays for protein interaction studies?

Proximity ligation assay (PLA) development requires careful attention to antibody compatibility and assay conditions. Select BHLH76 antibody pairs that target non-overlapping epitopes to prevent competitive binding . Verify that oligonucleotide conjugation doesn't impair antigen recognition using parallel conventional immunoassays. Optimize fixation and permeabilization conditions to preserve protein interactions while enabling antibody access. Establish specificity controls including samples lacking one interaction partner and competition with unconjugated antibodies . Quantitative analysis should include appropriate statistical methods for distinguishing true interaction signals from random colocalization. When studying dynamic interactions, implement time-course experiments with synchronized cell populations to capture temporal regulation of BHLH76 protein complexes .

How can BHLH76 antibody be utilized in antibody-drug conjugate (ADC) development for research applications?

For ADC research applications, antibody characteristics must be carefully evaluated. Assess BHLH76 antibody internalization efficiency using pH-sensitive dyes or quenching techniques to distinguish surface-bound from internalized antibody . Determine the intracellular trafficking pathway through colocalization studies with endosomal and lysosomal markers to select appropriate linker chemistry (e.g., acid-labile versus protease-cleavable linkers). Evaluate target expression heterogeneity across relevant cell populations to predict ADC efficacy and potential off-target effects . Optimize drug-antibody ratio (DAR) through controlled conjugation reactions and verify that conjugation doesn't impair antibody binding properties. Test cytotoxicity of the complete ADC using in vitro models with appropriate controls including unconjugated antibody and free toxin at equivalent concentrations .

What strategies enable effective use of BHLH76 antibody in extracellular vesicle (EV) characterization?

Extracellular vesicle characterization presents unique challenges requiring specialized approaches. For immunocapture of BHLH76-containing EVs, optimize antibody coupling to solid supports (magnetic beads, chips) ensuring proper orientation for antigen binding . Implement rigorous controls including isotype-matched antibodies and competitive binding with soluble BHLH76. For flow cytometric analysis of EVs, use fluorescence calibration beads to establish detection thresholds and provide semi-quantitative measurements . Consider implementing super-resolution microscopy for co-localization studies of BHLH76 with EV markers. Multiple orthogonal techniques (electron microscopy, nanoparticle tracking analysis, and Western blotting) should be combined to comprehensively characterize BHLH76-positive EV populations. Account for EV heterogeneity when interpreting results by implementing single-vesicle analysis when possible .

How can computational approaches enhance interpretation of BHLH76 antibody-based imaging data?

Computational analysis can extract maximal information from antibody-based imaging. Implement machine learning algorithms for automated identification of BHLH76-positive cells and quantification of expression levels across heterogeneous populations . Develop standardized image processing pipelines incorporating background correction, illumination normalization, and segmentation algorithms optimized for your specific tissue or cell type. For spatial analysis, employ computational tools to quantify relationships between BHLH76-positive cells and other relevant cellular populations or tissue structures . When analyzing temporal dynamics, utilize particle tracking algorithms to monitor BHLH76 localization changes. Integrate imaging data with other data types (transcriptomics, proteomics) using multimodal data fusion approaches to place BHLH76 expression patterns within broader biological contexts .

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.