The BHLH115 antibody targets the BHLH115 protein, a transcription factor that plays a key role in iron homeostasis in plants . BHLH115 belongs to the basic helix-loop-helix (bHLH) family of transcription factors . These proteins are involved in various developmental processes and stress responses in both plants and animals . In Arabidopsis thaliana, BHLH115, along with other bHLH proteins like bHLH105, regulates the expression of genes involved in iron uptake and distribution .
BHLH115 plays a crucial role in maintaining iron homeostasis in plants . It is regulated by BRUTUS, which ubiquitinates and degrades BHLH115 under iron-sufficient conditions . Under iron-deficient conditions, BHLH115 is stabilized and activates the expression of genes involved in iron uptake and distribution .
The interaction between BHLH115 and BRUTUS is also regulated by IMA peptides, which compete with BHLH115 for binding to BTS, thereby inhibiting the degradation of BHLH115 . BHLH115 also interacts with PYE, a negative regulator of bHLH Ib genes, to modulate iron homeostasis .
Several studies have investigated the role of BHLH115 in iron homeostasis . These studies have used various techniques, including genetic analyses, protein interaction assays, and expression analyses, to elucidate the function of BHLH115.
Interaction with BRUTUS: BHLH115 interacts with BRUTUS via its C-terminal region, which contains a BID . Mutation of the BID domain disrupts the interaction between BHLH115 and BRUTUS, leading to altered iron homeostasis .
Regulation by IMA peptides: IMA peptides compete with BHLH115 for binding to BRUTUS, thereby inhibiting the degradation of BHLH115 . This competition is important for maintaining iron homeostasis under iron-deficient conditions .
Interaction with PYE: BHLH115 interacts with PYE, a negative regulator of bHLH Ib genes . This interaction modulates the expression of genes involved in iron homeostasis .
Transcriptional regulation: BHLH115 activates the expression of genes involved in iron uptake and distribution . It directly binds to the promoters of genes like BTS and IMA3 under iron-deficient conditions .
BHLH115 antibodies are used in various research applications, including:
Protein detection: BHLH115 antibodies can be used to detect BHLH115 protein in plant tissues and cells .
Immunohistochemistry: BHLH115 antibodies can be used to localize BHLH115 protein in plant tissues using immunohistochemistry .
Protein interaction studies: BHLH115 antibodies can be used to identify proteins that interact with BHLH115 .
Functional studies: BHLH115 antibodies can be used to inhibit the function of BHLH115 and study its role in iron homeostasis .
KEGG: ath:AT1G51070
UniGene: At.17182
bHLH115 is a basic helix-loop-helix transcription factor that functions as a positive regulator of the iron-deficiency response in plants, particularly Arabidopsis thaliana. Its importance stems from its role in maintaining iron homeostasis, which is crucial for normal plant growth and development. Loss-of-function of bHLH115 causes strong iron-deficiency symptoms and reduces expression of genes responsive to iron deficiency . This transcription factor is part of a regulatory network involving other bHLH transcription factors (bHLH34, bHLH104, and bHLH105) that collectively maintain iron homeostasis . Understanding bHLH115 provides insights into plant adaptation mechanisms to nutrient-deficient environments, which has implications for crop improvement research.
bHLH115 antibodies serve multiple critical functions in plant molecular biology research. They are instrumental in protein detection via Western blotting, enabling quantification of bHLH115 expression levels under various experimental conditions. For studying protein-protein interactions, these antibodies are utilized in co-immunoprecipitation (Co-IP) assays to identify binding partners of bHLH115, particularly its interactions with other bHLH transcription factors like bHLH34, bHLH104, and bHLH105 . In chromatin immunoprecipitation (ChIP) assays, these antibodies help identify DNA binding sites, confirming that bHLH115 binds to the promoters of iron-deficiency-responsive genes such as bHLH38/39/100/101 and POPEYE (PYE) . Additionally, immunohistochemistry applications using these antibodies enable visualization of bHLH115 localization within plant tissues, providing spatial context to expression patterns.
bHLH115 functions as a key positive regulator within a complex transcriptional network controlling iron homeostasis. This protein directly binds to the promoters of iron-deficiency-responsive genes, including bHLH38/39/100/101 and POPEYE (PYE), activating their expression . bHLH115 can interact with itself (homodimerize) as well as form heterodimers with other regulatory bHLH transcription factors (bHLH34, bHLH104, and bHLH105), suggesting coordinated regulation of downstream targets .
The activity of bHLH115 is negatively regulated by BRUTUS (BTS), an E3 ubiquitin ligase that physically interacts with bHLH115, leading to its ubiquitination and subsequent degradation . This post-translational regulation creates an important control point in the iron response pathway. Additionally, the IMA3 protein can inhibit BTS activity, which buffers the degradation of bHLH115, ultimately promoting iron deficiency responses . This multilayered regulatory system ensures appropriate responses to changing iron availability, with bHLH115 playing a central role in this homeostatic mechanism.
When designing experiments with bHLH115 antibodies, several controls are essential for result validation:
Positive controls: Include wild-type Arabidopsis samples with known bHLH115 expression. This confirms the antibody is functioning as expected and provides a reference signal intensity.
Negative controls: Utilize bhlh115 knockout mutants to confirm antibody specificity. These samples should show no signal if the antibody is specific .
Loading controls: Standard proteins like Actin-11 (detected with appropriate antibodies as described in source ) should be used to normalize protein loading across samples.
Competition assays: Pre-incubate the antibody with purified bHLH115 protein before application to verify binding specificity.
Secondary antibody controls: Omit the primary bHLH115 antibody while retaining the secondary antibody to identify any non-specific binding of the secondary antibody.
Cross-reactivity assessment: Test antibody against similar bHLH family members (bHLH34, bHLH104, bHLH105) to ensure specificity, particularly important given their structural similarities and overlapping functions .
These controls collectively ensure that experimental results reflect genuine bHLH115 detection rather than artifacts or non-specific reactions.
bHLH115 antibodies provide powerful tools for investigating complex transcription factor assemblies through several sophisticated approaches:
Sequential ChIP (ChIP-reChIP): This technique involves performing a primary ChIP with bHLH115 antibodies followed by a secondary ChIP with antibodies against potential partner proteins (bHLH34, bHLH104, or bHLH105). This approach reveals genomic regions where multiple transcription factors co-bind, offering insights into combinatorial gene regulation .
Proximity ligation assays (PLA): By combining bHLH115 antibodies with antibodies against other transcription factors, PLA can visualize and quantify protein-protein interactions within native cellular contexts, including spatial information about where these complexes form.
Size-exclusion chromatography coupled with antibody detection: This method separates protein complexes by size before immunoblotting with bHLH115 antibodies, revealing the composition and stoichiometry of various bHLH115-containing complexes.
Cross-linking coupled with immunoprecipitation: Cross-linking proteins prior to immunoprecipitation with bHLH115 antibodies preserves weaker or transient interactions that might be lost in standard Co-IP procedures, providing a more comprehensive interactome.
Glycerol gradient ultracentrifugation with immunodetection: This technique separates protein complexes by size and shape, allowing identification of different bHLH115-containing complexes when followed by immunoblotting with bHLH115 antibodies.
These methodologies collectively enable researchers to dissect the protein interaction network surrounding bHLH115, which is crucial for understanding its role in transcriptional regulation during iron deficiency responses.
When investigating post-translational modifications (PTMs) of bHLH115, researchers should consider several critical factors:
Antibody selection specificity: Use modification-specific antibodies alongside total bHLH115 antibodies. For phosphorylation studies, phospho-specific antibodies that recognize specific phosphorylated residues on bHLH115 are essential for accurate detection.
Sample preparation preservation: PTMs can be labile during extraction. Include phosphatase inhibitors (sodium fluoride, sodium orthovanadate) when studying phosphorylation, and proteasome inhibitors (MG132) when investigating ubiquitination related to BTS-mediated degradation of bHLH115 .
Time-course experiments: Since BTS-mediated ubiquitination of bHLH115 is likely dynamic and iron-dependent, design time-course experiments that capture the temporal dynamics of these modifications in response to changing iron conditions.
Validation through multiple techniques: Combine immunoprecipitation followed by mass spectrometry with traditional Western blotting to identify and confirm PTM sites on bHLH115.
Comparative analysis: Study PTMs across different iron conditions and genetic backgrounds (wild-type vs. bts mutants) to correlate modifications with functional outcomes .
PTM site mutation studies: Generate transgenic lines expressing bHLH115 with mutations at putative modification sites to validate the functional significance of specific PTMs identified using antibody-based approaches.
These considerations ensure accurate detection and functional characterization of bHLH115 PTMs, which likely play crucial roles in regulating its stability and activity during iron stress responses.
The temporal and spatial expression dynamics of bHLH115 significantly impact experimental design decisions:
Tissue-specific antibody selection: Different antibodies may have varying efficacies in different plant tissues. The distinct tissue-specific expression patterns of bHLH115 noted in research necessitate validating antibody performance across multiple tissue types (roots, shoots, leaves).
Developmental timing considerations: bHLH115 expression likely varies across developmental stages. Experiments should be timed to capture relevant developmental windows, with sampling protocols adjusted accordingly.
Subcellular localization targeting: For localization studies, select antibodies validated for immunohistochemistry that maintain specificity under fixation conditions needed for preserving subcellular structures.
Signal amplification needs: In tissues with lower bHLH115 expression, more sensitive detection methods may be required, such as tyramide signal amplification coupled with immunodetection.
Iron status normalization: Since bHLH115 expression is regulated by iron availability , standardize iron conditions across experiments or explicitly test multiple iron regimes when comparing antibody performance.
Cross-tissue comparison controls: When comparing bHLH115 expression across different tissues, include recombinant protein standards at known concentrations to normalize detection efficiency across tissue types.
This multilayered approach accounts for the biological complexity of bHLH115 expression patterns, ensuring that antibody-based detection methods accurately capture its in vivo dynamics across tissues, developmental stages, and environmental conditions.
Investigating protein-protein interactions involving bHLH115 presents several technical challenges:
Transient interactions: The interactions between bHLH115 and other regulators may be transient or condition-dependent. Cross-linking reagents like formaldehyde or DSP (dithiobis[succinimidyl propionate]) should be employed before immunoprecipitation to capture these fleeting interactions.
Competitive binding patterns: Given that bHLH115 interacts with multiple partners (bHLH34, bHLH104, bHLH105, and BTS) , competitive binding may occur. Sequential immunoprecipitation with different antibodies can help resolve complex interaction networks.
Iron-dependent interactions: Since these interactions likely respond to iron status, experiments must be performed under carefully controlled iron conditions, including both sufficient and deficient states to capture condition-specific interactions.
Steric hindrance with epitope accessibility: Antibody binding may disrupt protein interactions if the epitope overlaps with interaction interfaces. Multiple antibodies recognizing different epitopes of bHLH115 should be tested.
Post-translational modification effects: PTMs like ubiquitination by BTS may alter epitope recognition. Antibodies should be validated under conditions where these modifications are present or absent.
Native complex preservation: Harsh extraction conditions may disrupt native complexes. Gentle extraction buffers that maintain physiological pH and salt concentrations help preserve genuine interactions.
Addressing these challenges requires combining multiple approaches (BiFC, FRET, Co-IP, yeast two-hybrid) to build a comprehensive and accurate picture of bHLH115's interaction network in iron homeostasis regulation.
For optimal bHLH115 antibody performance, consider these extraction and immunoprecipitation protocol elements:
Buffer composition optimization:
Base buffer: 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1% Triton X-100
Protease inhibitors: Complete protease inhibitor cocktail
PTM preservation: 10 mM NaF, 1 mM Na₃VO₄ (phosphatase inhibitors)
For ubiquitination studies: 50 μM MG132 (proteasome inhibitor)
Nuclease: DNase I (25 U/mL) to reduce chromatin interference in nuclear extracts
Tissue preparation considerations:
Flash-freeze tissue in liquid nitrogen before grinding to powder
Maintain cold chain throughout extraction (4°C)
For crosslinking studies: Add 1% formaldehyde for 10 minutes before quenching with glycine
Immunoprecipitation parameters:
Antibody binding: Overnight incubation at 4°C with gentle rotation
Protein A/G beads: Pre-block with 1% BSA to reduce non-specific binding
Wash stringency: Graduated washes with increasing salt concentration (150 mM to 300 mM NaCl)
Elution options: Either gentle (native conditions) with competition peptide or stringent (denaturing) with SDS
Specific considerations for transcription factor immunoprecipitation:
Nuclear isolation: Consider nuclear extraction protocols specifically designed for plant transcription factors
Chromatin shearing: If studying DNA-bound bHLH115, include controlled sonication steps
Salt concentration balance: High enough to reduce non-specific binding but not so high as to disrupt legitimate interactions with other bHLH proteins
These optimized protocols maximize both the yield and specificity of bHLH115 immunoprecipitation, providing cleaner results for downstream applications like mass spectrometry or immunoblotting.
For optimal Western blot detection of bHLH115 protein, consider these technical parameters:
Protein separation optimization:
Transfer parameters:
Blocking optimization:
Antibody incubation parameters:
Signal detection considerations:
Troubleshooting approaches:
These optimized conditions should provide clear, specific detection of bHLH115 protein in plant extracts while minimizing background and non-specific signals.
For effective ChIP assays with bHLH115 antibodies, follow these protocol recommendations:
This protocol has been optimized specifically for plant transcription factors like bHLH115 and accounts for the challenges of working with plant tissues that contain cell walls and abundant secondary metabolites.
When developing or selecting bHLH115 antibodies, researchers should consider these critical factors:
Epitope selection strategy:
Avoid the bHLH domain (amino acids ~150-200) to prevent cross-reactivity with related bHLH transcription factors
Target unique regions in the C-terminal domain, which appears less conserved based on interaction studies
Consider multiple epitopes to generate a panel of antibodies recognizing different regions
Application-specific considerations:
For Western blotting: Linear epitopes that remain accessible after denaturation
For IP/Co-IP: Epitopes outside protein interaction interfaces, particularly avoiding regions that interact with bHLH34, bHLH104, bHLH105, or BTS
For ChIP: Epitopes accessible when bHLH115 is bound to DNA
For IHC/IF: Epitopes preserved after fixation procedures
Validation requirements for different applications:
Production platform selection:
Monoclonal advantages: Consistent reproducibility, higher specificity
Polyclonal advantages: Recognition of multiple epitopes, potentially higher sensitivity
Recombinant antibody options: Consider nanobodies or single-chain antibodies for specialized applications
PTM-specific considerations:
Quality control metrics:
Minimum titer values for polyclonal preparations
Affinity measurements (KD values) for monoclonal antibodies
Lot-to-lot consistency testing protocols
These considerations ensure that selected antibodies will perform optimally for the intended experimental applications while minimizing cross-reactivity with related bHLH family proteins.
When facing specificity challenges with bHLH115 antibodies, implement these troubleshooting strategies:
Cross-reactivity with other bHLH proteins:
Validation approach: Test antibody against recombinant bHLH34, bHLH104, and bHLH105 proteins
Mitigation strategy: Pre-absorb antibody with recombinant related proteins to deplete cross-reactive antibodies
Genetic approach: Use multiple mutant lines (bhlh115, bhlh104, bhlh105) to confirm band identity
Epitope mapping: Identify which regions cause cross-reactivity for better antibody selection
Non-specific background signals:
Blocking optimization: Test different blocking agents (milk, BSA, commercial blockers)
Antibody dilution series: Titrate to find optimal concentration that maximizes signal-to-noise ratio
Washing modifications: Increase washing stringency with higher detergent concentrations or longer washing times
Secondary antibody controls: Include secondary-only controls to identify non-specific binding
Inconsistent results between experiments:
Standardization approach: Create a standard operating procedure with consistent protein amounts
Reference standards: Include purified recombinant bHLH115 as positive control in each experiment
Batch tracking: Document antibody lot numbers and prepare larger aliquots to reduce variability
Environmental standardization: Control temperature, incubation times, and buffer compositions precisely
Antibody performance changes over time:
Storage optimization: Store antibodies in small aliquots at -80°C with stabilizing proteins
Freeze-thaw monitoring: Track number of freeze-thaw cycles and performance changes
Fresh preparation considerations: For critical experiments, consider using newly purchased or prepared antibodies
Stability testing protocol: Periodically test antibody performance against standard samples
Epitope masking in protein complexes:
Denaturation strategy: Use stronger denaturing conditions to expose hidden epitopes
Alternative antibody approach: Use multiple antibodies targeting different epitopes
Sequential immunoprecipitation: Use antibodies against known partners first, then detect bHLH115
These troubleshooting approaches address the most common specificity challenges when working with antibodies against transcription factors like bHLH115 that are part of protein families with high sequence similarity.
When bHLH115 protein levels are below standard detection thresholds, employ these sensitivity-enhancing strategies:
Sample enrichment approaches:
Nuclear fraction isolation: Concentrate nuclear proteins where transcription factors reside
Immunoprecipitation concentration: Perform IP before Western blotting to concentrate bHLH115
TCA precipitation: Concentrate total protein before gel loading
Organelle-specific extraction: Target extraction to tissues with higher bHLH115 expression based on tissue-specific patterns
Signal amplification methods:
Enhanced chemiluminescence: Use high-sensitivity ECL reagents like Clarity Max
Fluorescent secondary antibodies: Consider near-infrared fluorescent detection systems
Tyramide signal amplification: For immunohistochemistry applications
Biotin-streptavidin systems: Use biotinylated secondary antibodies with streptavidin-HRP for amplification
Detection system optimization:
Extended exposure times: Capture weak signals with longer exposures while monitoring background
Cooled CCD cameras: Use high-sensitivity digital imaging systems
Photomultiplier gain adjustment: Optimize scanner settings for fluorescent detection
Digital signal integration: Combine multiple exposures computationally to improve signal-to-noise ratio
Protocol modifications for low abundance proteins:
Reduce transfer time: Shorter transfer times prevent small proteins from passing through membrane
Membrane selection: 0.2 μm PVDF can retain smaller proteins better than 0.45 μm
Loading buffer optimization: Add carrier proteins to prevent loss during handling
Blocking time reduction: Minimize epitope masking by reducing blocking time while maintaining specificity
Environmental induction approaches:
Iron deficiency treatment: Since bHLH115 responds to iron deficiency, grow plants under iron-limited conditions to upregulate expression
Proteasome inhibition: Treat samples with MG132 to prevent BTS-mediated degradation of bHLH115
Stress response induction: Apply appropriate stresses known to affect iron homeostasis pathways
These strategies collectively enhance detection sensitivity while maintaining specificity, enabling the study of bHLH115 even when expressed at physiologically low levels or in specific cell types.
When facing discrepancies between different antibody-based detection methods for bHLH115, implement this systematic resolution approach:
Methodological comparison analysis:
Create a comparison table documenting exact protocols used across techniques
Identify key differences in sample preparation, antibody concentrations, and detection methods
Standardize critical parameters where possible to eliminate methodological variables
Test both techniques on identical sample preparations to isolate technique-specific issues
Epitope accessibility assessment:
Different techniques expose epitopes differently (e.g., Western blot - denatured; IP - native)
Test antibodies recognizing different epitopes within bHLH115
For techniques requiring native protein (IP, ChIP), ensure epitope is surface-exposed in tertiary structure
For techniques using fixed tissues, validate epitope preservation under fixation conditions
Validation through complementary approaches:
Confirm Western blot results with mass spectrometry identification
Validate ChIP results with EMSA or reporter gene assays
Complement co-IP findings with yeast two-hybrid or split-GFP assays
Compare antibody-based protein detection with transcript levels (accounting for post-transcriptional regulation)
Control analysis matrix:
Implement a comprehensive control series including:
Document control results systematically to identify pattern-specific discrepancies
Environmental and physiological variable assessment:
Iron status standardization: Given bHLH115's role in iron homeostasis, ensure consistent iron conditions
Developmental timing: Confirm samples are at equivalent developmental stages
Tissue specificity: Account for differential expression patterns across tissues
Stress conditions: Control for additional variables that might affect bHLH115 levels
By systematically addressing these factors, researchers can pinpoint the source of conflicting results and develop a consistent experimental approach that yields reproducible data across multiple antibody-based techniques.
Several cutting-edge technologies present exciting opportunities for advancing bHLH115 research:
Single-cell protein analysis technologies:
Single-cell Western blotting: Enables detection of bHLH115 in individual cells, revealing cell-to-cell variation
Mass cytometry (CyTOF): Allows multiplexed protein detection at single-cell resolution using metal-tagged antibodies
Microfluidic antibody capture: Permits analysis of protein expression in rare cell populations
Advanced imaging technologies:
Super-resolution microscopy: Techniques like STORM or PALM can visualize bHLH115 localization with nanometer precision
Proximity ligation imaging: Enables visualization of bHLH115 interactions with partners (bHLH34, bHLH104, bHLH105, BTS) in situ
Lattice light-sheet microscopy: Allows real-time imaging of bHLH115 dynamics with minimal phototoxicity
High-throughput antibody approaches:
Microarray-based antibody screening: Tests thousands of antibody variants simultaneously for improved specificity
Synthetic antibody libraries: Generates highly specific recombinant antibodies against multiple epitopes
AI-designed antibodies: Leverages emerging AI platforms to design antibodies with optimal specificity and affinity
Integrative multi-omics approaches:
Spatial transcriptomics with protein detection: Correlates bHLH115 protein levels with gene expression patterns in tissue context
Proteogenomics platforms: Connects genetic variation to bHLH115 protein abundance and modification states
Systems biology frameworks: Integrates antibody-derived data with metabolomic and phenotypic datasets
Novel antibody formats and modifications:
Nanobodies: Single-domain antibodies with superior tissue penetration for in vivo imaging
Split-epitope complementation: Enables detection of specific protein conformations or complexes
Photoswitchable antibodies: Allows temporal control of antibody binding for dynamic studies
These emerging technologies promise to advance our understanding of bHLH115 function by providing higher resolution, greater specificity, and more comprehensive data integration capacities than current approaches.
Computational methods can significantly enhance bHLH115 antibody data analysis through these advanced approaches:
Machine learning for image analysis:
Automated detection: Train neural networks to identify bHLH115 localization patterns in immunofluorescence images
Feature extraction: Identify subtle phenotypic changes in bHLH115 mutants or overexpression lines
Classification algorithms: Categorize cellular responses to iron status based on bHLH115 distribution patterns
Quantitative image analysis: Measure co-localization coefficients between bHLH115 and interaction partners
Network inference from protein interaction data:
Bayesian network modeling: Infer probabilistic relationships in the iron homeostasis network
Temporal network dynamics: Model how bHLH115 interactions change over time in response to iron availability
Perturbation analysis: Predict network responses to mutations or environmental changes
Multi-omics data integration: Combine antibody-based interaction data with transcriptomics and metabolomics
Protein structure and epitope prediction:
Molecular dynamics simulations: Model how bHLH115 structure changes during interactions
Epitope mapping algorithms: Predict optimal epitopes for new antibody development
Binding interface prediction: Identify critical residues in bHLH115 interactions with BTS and other partners
Post-translational modification site prediction: Anticipate how PTMs might affect antibody binding
Advanced statistical approaches for ChIP-seq analysis:
Peak calling optimization: Develop specialized algorithms for transcription factor binding patterns
Motif discovery: Identify novel DNA binding motifs for bHLH115
Differential binding analysis: Compare binding patterns across genetic backgrounds and conditions
Integrative genomics: Connect binding sites to gene expression changes and chromatin states
Data visualization and integration platforms:
Interactive visualization tools: Create platforms for exploring complex bHLH115 datasets
Comparative display frameworks: Visualize differences between experimental conditions
Multi-scale integration: Connect molecular-level antibody data to whole-plant phenotypes
Knowledge base development: Build searchable repositories of bHLH115-related experimental data
These computational approaches transform raw antibody-derived data into biological insights by revealing patterns, predicting mechanisms, and generating testable hypotheses about bHLH115 function in iron homeostasis.