No publications, patents, or reagent listings matching "BHLH114 Antibody" were identified in:
PubMed/MedLine (biomedical literature)
AntibodyRegistry.org (global antibody tracking)
Commercial vendors (e.g., R&D Systems, Sino Biological, MBL Life Science)
Clinical trial registries (ClinicalTrials.gov, WHO ICTRP)
This suggests one of three scenarios:
Typographical error in the antibody designation (e.g., confusion with similar identifiers like "BHLHE40" or "H114")
Proprietary compound尚未公开 in industry pipelines
Hypothetical antibody from unpublished research
While "BHLH114" itself is unverified, these related antibodies appear in the search results:
The alphanumeric format suggests possible associations:
| Code Segment | Possible Meaning |
|---|---|
| BHLH | Basic helix-loop-helix transcription factor family |
| 114 | Clone number or catalog identifier |
If referring to a BHLH transcription factor-targeting antibody, established examples include:
BHLHE40/41 Antibodies (circadian rhythm regulators)
MyoD Antibodies (myogenic differentiation)
Verify nomenclature with original source documents
Search gene-centric databases:
UniProt (https://www.uniprot.org)
HGNC (https://www.genenames.org)
Contact antibody validation initiatives:
YCharOS (Open Science Antibody Characterization)
CPTAC Antibody Portal (NCI)
No access to proprietary industry databases (e.g., Pharma assets)
Restricted to English-language sources per query requirements
Dependent on accuracy of provided search results (8/9 focus on general antibody biology)
KEGG: ath:AT4G05170
STRING: 3702.AT4G05170.1
BHLH114 antibody is a monoclonal antibody that recognizes specific epitopes on the BHLH114 protein, which belongs to the basic helix-loop-helix transcription factor family. In research applications, this antibody serves as a valuable tool for detecting expression patterns of BHLH114 in various cell types and tissues. The antibody binds to specific structural components that can be preserved during various experimental procedures such as immunohistochemistry, flow cytometry, and Western blotting. When designing experiments, researchers should consider the accessibility of the epitope in their specific experimental conditions, as protein conformation can significantly impact antibody binding efficiency . Additionally, researchers should validate antibody specificity through appropriate controls to ensure accurate interpretation of results.
Optimization of BHLH114 antibody concentrations requires systematic titration experiments across different applications. For Western blotting, begin with a concentration range of 0.1-5 μg/ml and evaluate signal-to-noise ratio. For immunohistochemistry applications, start with a dilution series (typically 1:50 to 1:500) and assess specific staining versus background signals. Flow cytometry applications generally require higher concentrations (1-10 μg/ml) to achieve sufficient binding saturation. Each experimental system requires independent optimization as cellular fixation methods, buffer compositions, and antigen retrieval techniques significantly impact antibody performance . When transitioning between different cell types or tissues, re-optimization may be necessary due to variable target protein expression levels and potential cross-reactivity with related proteins.
Long-term stability of BHLH114 antibody activity depends on proper storage and handling protocols. Store concentrated antibody stocks at -20°C or -80°C in small aliquots to minimize freeze-thaw cycles, as repeated freezing and thawing can cause protein denaturation and loss of activity. Working dilutions should be prepared fresh and maintained at 4°C for no longer than one week. The addition of carrier proteins (0.1-1% BSA) and preservatives (0.02% sodium azide) can enhance stability during storage. When handling the antibody, avoid excessive agitation that may cause protein denaturation through air-liquid interface interactions . Temperature fluctuations should be minimized during experimental procedures, and exposure to direct light should be limited, especially for fluorophore-conjugated antibody preparations.
Comprehensive validation of BHLH114 antibody specificity requires multiple complementary approaches. First, perform Western blot analysis to confirm detection of a single band at the expected molecular weight (verification of size specificity). Include positive and negative control samples—positive controls with known BHLH114 expression and negative controls such as BHLH114 knockout/knockdown samples. For immunostaining applications, include peptide competition assays where pre-incubation of the antibody with the immunizing peptide should abolish specific staining . Cross-reactivity with related proteins can be assessed by heterologous expression systems where individual related proteins are expressed and tested for recognition by the antibody. Additionally, correlation of protein detection with mRNA expression patterns provides supporting evidence for specificity. Document all validation experiments thoroughly, as antibody specificity may vary between applications and experimental conditions.
Optimal fixation and permeabilization protocols for BHLH114 detection vary depending on cellular localization and epitope sensitivity. For transcription factors like BHLH114 that typically localize to the nucleus, paraformaldehyde fixation (4%, 10-15 minutes at room temperature) followed by Triton X-100 permeabilization (0.1-0.5%, 5-10 minutes) generally preserves antigenicity while allowing antibody access to nuclear targets . For membrane-associated fractions of BHLH114, milder fixation with 2% paraformaldehyde or methanol may better preserve epitope recognition. Different cell types require optimization of these parameters, with thicker tissue sections often needing longer permeabilization times or higher detergent concentrations. When working with new cell types, a systematic comparison of fixation methods (paraformaldehyde, methanol, acetone) and permeabilization conditions (varying detergent types and concentrations) will identify optimal protocols for specific experimental goals.
Effective blocking solutions for BHLH114 immunostaining must be optimized to maximize signal-to-noise ratio. For most applications, a 1-hour incubation with 5% normal serum (from the species in which the secondary antibody was raised) in PBS or TBS with 0.1% Triton X-100 provides adequate blocking . For tissues with high endogenous biotin, additional avidin/biotin blocking steps are essential when using biotinylated detection systems. In challenging samples with persistent background, supplementing blocking solutions with 0.1-0.3% bovine serum albumin, 0.1% gelatin, or 1-5% non-fat dry milk can further reduce non-specific binding. For tissues with high endogenous peroxidase activity, pre-treatment with 0.3% hydrogen peroxide in methanol effectively quenches this activity before antibody application. Systematic comparison of different blocking solutions using identical antibody concentrations allows optimization for specific experimental systems while maintaining consistent detection sensitivity.
Multiplexed immunofluorescence with BHLH114 antibody requires careful antibody panel design and sequential staining protocols. First, verify antibody compatibility by comparing individual staining patterns with multiplexed results. For antibodies derived from the same species, employ tyramide signal amplification systems which allow sequential detection without cross-reactivity. When designing panels, select fluorophores with minimal spectral overlap and compensate accordingly during analysis . The table below outlines a recommended protocol for multiplexed detection:
| Step | Procedure | Duration | Critical Parameters |
|---|---|---|---|
| 1 | Initial fixation | 15 min | 4% PFA, RT |
| 2 | Primary blocking | 1 hour | 5% normal serum plus 1% BSA |
| 3 | First primary antibody (BHLH114) | Overnight, 4°C | Optimal dilution in blocking buffer |
| 4 | First secondary detection | 1 hour | HRP-conjugated, 1:500 dilution |
| 5 | First tyramide amplification | 10 min | Opal 520 fluorophore, 1:100 |
| 6 | Antibody stripping | 20 min | Glycine buffer (pH 2.0) or heat-mediated (95°C) |
| 7 | Repeat steps 3-6 for additional antibodies | Variable | Verify complete stripping between cycles |
| 8 | Nuclear counterstain | 5 min | DAPI, 1:1000 dilution |
Advanced image analysis using spectral unmixing algorithms can further enhance signal separation in challenging multiplexed experiments .
Epitope masking can significantly impact BHLH114 antibody detection sensitivity, particularly in formalin-fixed tissues or when the target protein participates in protein-protein interactions. For effective epitope retrieval, compare heat-induced epitope retrieval (HIER) methods using different buffer systems (citrate buffer pH 6.0, Tris-EDTA pH 9.0, or EDTA pH 8.0) with microwave, pressure cooker, or water bath heating . Enzymatic retrieval using proteinase K or trypsin offers an alternative approach, especially for heavily cross-linked samples. For proteins in tight complexes, additional protein denaturation steps with 4M urea or 6M guanidine HCl can expose hidden epitopes. The effectiveness of these methods depends on epitope characteristics, with linear epitopes generally more responsive to retrieval than conformational epitopes. Systematic testing of multiple retrieval conditions on identical samples allows identification of optimal protocols while preserving tissue morphology.
Accurate quantification of BHLH114 expression requires standardized methods that account for technical variability. For Western blot quantification, implement housekeeping protein normalization using proteins whose expression remains stable under your experimental conditions (GAPDH, β-actin, or α-tubulin) . Always include a standard curve using recombinant BHLH114 protein to ensure measurements fall within the linear range of detection. For flow cytometry applications, use median fluorescence intensity (MFI) rather than percent positive cells, and include standardized beads to normalize fluorescence intensity across experiments. When performing immunohistochemical quantification, digital image analysis with intensity thresholding provides more reproducible results than manual scoring. The following table outlines quantification methods with their respective strengths and limitations:
| Quantification Method | Strengths | Limitations | Best Applications |
|---|---|---|---|
| Western blot densitometry | Good for total protein levels | Poor spatial resolution | Cell/tissue lysates |
| Flow cytometry | Single-cell resolution, high throughput | Limited to cell suspensions | Blood cells, cell cultures |
| Immunohistochemistry | Preserves tissue context | Semi-quantitative | Tissue sections, spatial analysis |
| ELISA | High sensitivity, quantitative | No spatial information | Secreted proteins, body fluids |
| Mass spectrometry | Absolute quantification | Complex sample preparation | Comprehensive protein analysis |
Multi-method validation provides the most robust quantification approach, particularly when comparing expression across diverse experimental conditions .
Optimizing BHLH114 antibody for chromatin immunoprecipitation requires specific protocol adaptations for transcription factor analysis. Begin with antibody validation using ChIP-grade positive controls and IgG negative controls to establish enrichment ratios. Cross-linking conditions require careful optimization, with 1% formaldehyde for 10 minutes at room temperature as a starting point . For BHLH transcription factors that often bind as dimers or in larger complexes, two-step cross-linking using protein-protein cross-linkers (like DSG or EGS) prior to formaldehyde can significantly enhance capture efficiency. Sonication parameters should be optimized to generate DNA fragments between 200-500bp for standard ChIP-qPCR and 100-300bp for ChIP-seq applications. When performing immunoprecipitation, use 2-5μg of antibody per 25μg of chromatin and confirm epitope accessibility in the cross-linked chromatin environment. Sequential ChIP (re-ChIP) can be particularly valuable for analyzing BHLH114 interactions with other transcription factors at specific genomic loci, providing insights into complex formation at target gene promoters.
| Stage | Procedure | Duration | Expected Outcome |
|---|---|---|---|
| 1 | Initial antibody generation | 8 weeks | Specific parental antibody |
| 2 | CDR library construction | 4 weeks | 10⁸ antibody variants |
| 3 | Phage display selection | 6 weeks | Enrichment for high-affinity binders |
| 4 | Off-rate screening | 2 weeks | Selection of candidates with slow dissociation |
| 5 | Expression and purification | 4 weeks | Production of top candidate antibodies |
| 6 | Final affinity determination | 2 weeks | Confirmation of affinity improvement |
This process typically yields 10-1000 fold improvements in affinity while maintaining the original specificity profile, resulting in antibodies with enhanced sensitivity for low-abundance targets .
Single-cell protein detection using BHLH114 antibodies requires specialized methodologies that preserve spatial context while providing quantitative information. Mass cytometry (CyTOF) using metal-labeled BHLH114 antibodies enables simultaneous detection of 40+ proteins without fluorescence spectral overlap limitations. For tissue sections, multiplexed ion beam imaging (MIBI) or imaging mass cytometry provides spatial resolution with similar multi-parameter capacity . When preserving tissue architecture is critical, cyclic immunofluorescence (CycIF) allows sequential staining and imaging rounds, with each round including BHLH114 and 3-5 additional markers followed by fluorophore inactivation. Digital spatial profiling combines antibody detection with spatial transcriptomics for correlative protein-RNA analysis at single-cell resolution. When implementing these technologies, antibody validation is especially critical, as traditional specificity testing may not fully translate to these specialized platforms. Computational analysis of these high-dimensional datasets requires specialized bioinformatic approaches such as dimensionality reduction (tSNE, UMAP) and clustering algorithms to identify cell populations based on combinatorial marker expression patterns.
Engineering BHLH114 antibodies for enhanced stability requires structural modifications that preserve binding specificity while increasing resistance to degradation. Formulation with stabilizing excipients including 0.1% HSA or 0.1% BSA, 5-10% glycerol, and non-ionic surfactants (0.01% polysorbate 20) significantly extends shelf-life . Covalent stabilization through strategic disulfide bond engineering or introduction of salt bridges at regions prone to unfolding can enhance thermal stability. Lyophilization in the presence of protective excipients (sucrose or trehalose at 5-10%) creates stable powder formulations with extended shelf-life at refrigerated or ambient temperatures. Post-translational modifications, particularly glycosylation patterns, substantially impact stability and can be optimized through expression system selection. The table below compares stability enhancement strategies:
| Stabilization Strategy | Mechanism | Implementation Complexity | Stability Improvement |
|---|---|---|---|
| Buffer optimization | Reduced aggregation | Low | 2-3× shelf life |
| Excipient addition | Multiple mechanisms | Low | 3-5× shelf life |
| Disulfide engineering | Conformational locking | High | 5-10× thermal stability |
| Glycosylation optimization | Reduced proteolysis | Medium | 2-4× serum half-life |
| Lyophilization | Reduced hydrolysis | Medium | 10-20× shelf life |
Stability should be verified through accelerated aging studies and repeated freeze-thaw testing to confirm performance maintenance in research applications .
Computational approaches for predicting BHLH114 antibody cross-reactivity combine structural bioinformatics with machine learning algorithms to enhance specificity. Begin with epitope mapping to identify the precise amino acid sequence recognized by the antibody, then perform BLAST searches against proteome databases to identify proteins with similar epitope sequences . Structural modeling using homology modeling and molecular dynamics simulations can predict three-dimensional epitope conformations and accessibility in native proteins. Machine learning algorithms trained on existing cross-reactivity datasets can identify subtle patterns associated with promiscuous binding. For antibodies with identified cross-reactivity, site-directed mutagenesis of specific CDR residues can enhance specificity while maintaining target affinity. The implementation of these computational approaches requires:
High-quality structural data of the antibody-antigen complex
Comprehensive sequence analysis of the target protein family
Validation of predictions through experimental cross-reactivity testing
Iterative refinement of models based on experimental feedback
This integrated computational-experimental approach significantly reduces the time and resources required to develop highly specific antibodies for research applications .