HES2 antibodies target the HES2 protein, a member of the basic helix-loop-helix (bHLH) transcription factor family. HES2 regulates gene expression by repressing transcriptional activators, particularly in the Notch signaling pathway, which governs cell differentiation and tissue patterning . These antibodies are widely used in techniques such as Western blotting (WB), immunohistochemistry (IHC), and immunofluorescence (IF) to investigate HES2's role in development and disease .
HES2 maintains the balance between cell proliferation and differentiation by forming complexes with TLE proteins via its WRPW motif, enabling transcriptional repression . Studies using HES2 antibodies highlight its overexpression in cancers, such as ovarian carcinomas, where it correlates with tumor aggressiveness . For example:
Hesca-2 Antibody: Targets glycan epitopes (e.g., Lewis C) on human embryonic stem cells (hESCs) and ovarian cancer cells, showing cytotoxic effects at low concentrations .
SSEA-5 and Glycan Shifts: HES2-associated glycans transition from type 1 (e.g., Lewis(a)) in hESCs to type 2 (e.g., CD15) during differentiation, suggesting lineage-specific biomarkers .
Western Blot Validation: Recombinant HES2 protein (20.9 kDa, His-tagged) is used as a positive control, with antibodies like Novus 4H6 detecting bands at ~34 kDa in human cell lines .
Immunohistochemistry: Polyclonal antibodies (e.g., Novus NBP2-27110) stain nuclear HES2 in primate tissues, aiding developmental biology studies .
Storage: Most antibodies require storage at -20°C in stabilizing buffers (e.g., PBS + 0.05% BSA) to prevent aggregation .
Validation: Antibodies are validated using knockout controls or recombinant proteins (e.g., Novus NBP2-23099) .
Species Reactivity: While some antibodies cross-react with primates, others are species-specific (e.g., Elabscience’s rat-reactive antibody) .
Recent studies emphasize HES2's potential as a therapeutic target. For instance:
HES2 (Hairy and Enhancer of Split 2) is a transcriptional repressor belonging to the basic helix-loop-helix (bHLH) transcription factor family. It plays a crucial role in gene expression regulation during embryonic development, particularly within the Notch signaling pathway, which is essential for cell differentiation and tissue patterning . The protein forms complexes with TLE proteins involved in transcriptional repression, with this interaction mediated by the carboxy-terminal WRPW motif, enabling effective inhibition of transcriptional activator activity . This regulatory mechanism maintains balance between cell proliferation and differentiation, particularly important in neural and muscle development.
HES2 contains a specialized basic domain with a helix-interrupting proline that preferentially binds to N-box (CACNAG) sequences rather than canonical E-box (CANNTG) motifs typically recognized by other bHLH proteins . The protein's C-terminal WRPW motif functions as a transcriptional repression domain necessary for interaction with corepressors of the Groucho/TLE family . HES2 is expressed in various embryonic and adult tissues, with notable expression in placenta and several cancer types including pancreatic cancer, colon cancer with RER, cervical cancer, and head and neck tumors .
Multiple types of HES2 antibodies are available for research applications, each with specific characteristics suitable for different experimental approaches:
Monoclonal Antibodies:
Polyclonal Antibodies:
These antibodies are validated for multiple applications including western blotting (WB), immunoprecipitation (IP), immunofluorescence (IF), enzyme-linked immunosorbent assay (ELISA), and immunohistochemistry (IHC) . Both unconjugated forms and various conjugated versions (including agarose, HRP, PE, FITC, and Alexa Fluor® conjugates) are available for specialized applications .
Proper storage is critical for maintaining antibody activity and preventing degradation. Based on manufacturer recommendations, the following storage protocols should be implemented:
Long-term storage: Store at -20°C, with antibodies aliquoted to minimize freeze-thaw cycles
Shipping conditions: Most HES2 antibodies are shipped with ice packs and should be stored immediately upon receipt at the recommended temperature
Formulation considerations: Many HES2 antibodies are supplied in buffers containing stabilizers such as PBS with 50% glycerol and 0.02% sodium azide or other protein protectants
It is crucial to avoid repeated freeze-thaw cycles as these significantly reduce antibody activity . For optimal results, smaller working aliquots should be prepared upon receipt to minimize the need for repeated freezing and thawing of the entire stock.
Optimal dilution ratios vary based on both the specific antibody and the application. The following dilution ranges are recommended based on manufacturer guidelines:
For optimal results, researchers should perform a titration experiment using different dilutions of antibody on control samples with known expression levels of HES2. Start with the manufacturer's recommended range and adjust based on signal-to-noise ratio. When switching to a new batch of the same antibody, validation of the optimal dilution should be repeated to account for potential lot-to-lot variations in antibody concentration and affinity.
When studying HES2 in contexts with low protein expression, several methodological refinements can enhance detection sensitivity:
Signal Amplification Systems:
Consider using tyramide signal amplification (TSA) for immunohistochemistry applications
For Western blotting, enhanced chemiluminescence (ECL) substrates with higher sensitivity can improve detection limits
Utilize biotin-streptavidin amplification systems with HRP-conjugated secondary antibodies
Sample Preparation Optimization:
Enrich for nuclear fractions, as HES2 is primarily localized in the nucleus
Use proteasome inhibitors (e.g., MG132) during sample preparation to prevent rapid degradation of HES2
Optimize protein extraction buffers with appropriate detergents (0.1-1% NP-40 or Triton X-100) to effectively solubilize nuclear proteins
Detection Systems:
For immunofluorescence, consider using conjugated primary antibodies (such as HES2 Antibody with Alexa Fluor® conjugates)
Implement fluorescent secondary antibodies with longer wavelengths to reduce background autofluorescence
Use high-sensitivity imaging systems with cooled CCD cameras for fluorescence applications
Controls:
The observed molecular weight for HES2 in Western blot applications is approximately 19 kDa , although the theoretical molecular weight is 33.77 kDa , which should be noted when interpreting results.
Optimizing IHC protocols for FFPE tissues requires attention to several critical factors:
Antigen Retrieval:
Heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) at 95-98°C for 20-30 minutes
Enzymatic retrieval using proteinase K may be considered for certain tissues, but this should be empirically determined
Pressure cooker or microwave-based retrieval systems often provide more consistent results than water bath methods
Blocking Parameters:
Implement dual blocking approach: first block endogenous peroxidase activity (3% H₂O₂ for 10 minutes), then block non-specific binding (5% normal serum from secondary antibody species)
Consider using commercial blocking reagents containing both protein blockers and detergents
For tissues with high endogenous biotin, use avidin-biotin blocking kits prior to antibody incubation
Antibody Incubation:
Detection Systems:
Polymer-based detection systems often provide superior results compared to traditional ABC methods
Consider using amplification systems for tissues with low HES2 expression
Optimize DAB development time through careful monitoring to achieve optimal signal-to-noise ratio
Validated Controls:
Verifying antibody specificity is crucial for ensuring reliable research outcomes. Multiple complementary approaches should be implemented:
Genetic Knockdown/Knockout Validation:
Use siRNA or CRISPR-Cas9 to reduce or eliminate HES2 expression, then confirm corresponding reduction in antibody signal
Employ inducible expression systems to demonstrate signal correlation with controlled HES2 expression levels
Peptide Competition Assays:
Pre-incubate the HES2 antibody with excess immunizing peptide before application
For polyclonal antibodies raised against specific peptides (such as the 8-25 amino acid region or 40-120 amino acid range ), use the corresponding synthetic peptides
A significant reduction in signal indicates specific binding to the target epitope
Multiple Antibody Validation:
Heterologous Expression Systems:
Express tagged versions of HES2 in cell lines with low endogenous expression
Demonstrate co-localization of tag-specific antibody signals with HES2 antibody signals
Show absence of signal in empty vector transfected controls
Mass Spectrometry Validation:
Perform immunoprecipitation using the HES2 antibody followed by mass spectrometry analysis
Confirm that peptides identified correspond to HES2 protein
Analyze non-specific binding partners to assess potential cross-reactivity
HES2 functions through interactions with various proteins, particularly TLE family corepressors. The following IP strategy can effectively capture these interactions:
Nuclear Extract Preparation:
Since HES2 is predominantly nuclear-localized , optimize nuclear extraction protocols
Use gentle lysis buffers (e.g., 20 mM HEPES pH 7.9, 150 mM NaCl, 0.5% NP-40, 10% glycerol) supplemented with protease and phosphatase inhibitors
Include 1-2 mM DTT to preserve protein-protein interactions involving cysteine residues
Immunoprecipitation Protocol:
Pre-clear nuclear extracts with protein A/G beads to reduce non-specific binding
Use 2-5 μg of HES2 antibody per 500 μg of nuclear extract
The mouse monoclonal HES2 antibody (H-8) has been validated for immunoprecipitation applications
Consider using agarose-conjugated HES2 antibody (HES2 Antibody H-8 AC) for direct IP without secondary antibody beads
Co-Immunoprecipitation for Binding Partners:
Focus on known interaction partners such as TLE proteins that bind via the C-terminal WRPW motif
Include appropriate detergent conditions (0.1-0.5% NP-40) to maintain interactions while reducing background
After IP, perform Western blotting for both HES2 (to confirm successful precipitation) and potential binding partners
Controls and Validation:
Include IgG control immunoprecipitations to establish background binding levels
For TLE interaction studies, consider using wild-type HES2 versus WRPW motif mutants to demonstrate specificity
Validate interactions through reciprocal co-IPs (i.e., IP with anti-TLE antibody and blot for HES2)
Analysis of Post-Translational Modifications:
After IP, probe for potential modifications (phosphorylation, acetylation, ubiquitination) that may regulate HES2 function
Consider using modification-specific antibodies in Western blotting after HES2 immunoprecipitation
HES2 binds to specific DNA sequences as part of its transcriptional regulatory function. The following methods can characterize its DNA binding properties and genomic targets:
Chromatin Immunoprecipitation (ChIP):
Use cross-linking agents (1% formaldehyde for 10 minutes) to stabilize DNA-protein interactions
Sonicate chromatin to 200-500 bp fragments for optimal resolution
Immunoprecipitate using 3-5 μg of HES2 antibody per ChIP reaction
Include appropriate controls (IgG ChIP, input samples)
Analyze enriched regions by qPCR (for known targets) or sequencing (for genome-wide analysis)
DNA Binding Specificity Assays:
Electrophoretic Mobility Shift Assays (EMSA) using recombinant HES2 or nuclear extracts
Focus on N-box (CACNAG) sequences, which are preferentially bound by HES2's specialized basic domain
Compare binding to canonical E-box (CANNTG) sequences to demonstrate binding specificity
Use competition assays with unlabeled probes to confirm sequence-specific binding
Reporter Gene Assays:
Construct luciferase reporters containing predicted HES2 binding sites
Test HES2-mediated repression through co-transfection experiments
Use site-directed mutagenesis of N-box sequences to confirm specificity
Test the functional importance of the WRPW motif by comparing wild-type and mutant HES2 constructs
Genome-wide Approaches:
ChIP-seq to identify all genomic binding sites
RNA-seq following HES2 knockdown/overexpression to identify genes with expression changes
Integration of ChIP-seq and RNA-seq data to identify direct transcriptional targets
Motif enrichment analysis to confirm binding site preferences in vivo
Non-specific background in Western blotting can significantly complicate data interpretation. Several strategies can minimize these issues:
Antibody-Related Factors:
Optimize primary antibody dilution: for Western blotting, use 1:500-1:2000 for polyclonal antibodies or 1-5 μg/mL for monoclonal antibodies
Extend washing steps (4-5 times, 5-10 minutes each) with TBST (TBS + 0.1% Tween-20)
Consider using more specific monoclonal antibodies (such as clone H-8 or clone 1D5 ) rather than polyclonal antibodies if background persists
Pre-adsorb polyclonal antibodies with cell/tissue lysates from HES2-negative samples
Blocking Optimization:
Test different blocking agents (5% non-fat dry milk, 5% BSA, or commercial blocking buffers)
Extend blocking time to 1-2 hours at room temperature or overnight at 4°C
Add 0.1-0.2% Tween-20 to blocking buffer to reduce hydrophobic interactions
Sample Preparation Improvements:
Ensure complete protein denaturation by heating samples at 95°C for 5 minutes in loading buffer
Include reducing agents (DTT or β-mercaptoethanol) in sample buffer
Centrifuge samples after heating to remove insoluble material
Consider nuclear extraction protocols to enrich for HES2, which is primarily nuclear-localized
Membrane Handling:
Use PVDF membranes instead of nitrocellulose for potentially better signal-to-noise ratio
After transfer, rinse membranes thoroughly in TBST before blocking
Consider membrane-specific blocking agents recommended by membrane manufacturers
Achieving clear immunofluorescence results requires attention to fixation, permeabilization, and detection parameters:
Fixation and Permeabilization:
For nuclear proteins like HES2, 4% paraformaldehyde fixation (15-20 minutes) followed by 0.1-0.5% Triton X-100 permeabilization (10 minutes) is typically effective
Test different fixatives (methanol, acetone, or PFA) if initial results are suboptimal
For certain applications, methanol fixation (-20°C, 10 minutes) may provide better nuclear antigen accessibility
Blocking and Antibody Incubation:
Block with 5-10% normal serum (from secondary antibody species) with 0.1-0.3% Triton X-100
Consider adding 1-2% BSA to reduce non-specific binding
For primary antibody incubation, dilute appropriately and incubate overnight at 4°C in a humidified chamber
For HES2 antibody (H-8), which has been validated for immunofluorescence applications , follow manufacturer's recommended dilutions
Signal Detection:
Use directly conjugated primary antibodies (such as HES2 antibody with fluorophore conjugates) to reduce background
When using secondary antibodies, highly cross-adsorbed versions minimize species cross-reactivity
Include DAPI or other nuclear counterstains to visualize nuclear localization of HES2
Mount slides using anti-fade mounting media to prevent photobleaching
Controls and Validation:
Include negative controls (primary antibody omission, isotype controls)
Use positive control cells/tissues with known HES2 expression
Consider siRNA knockdown controls to demonstrate specificity
Perform z-stack imaging to accurately determine subcellular localization
Quantitative analysis of HES2 requires careful attention to methodology and normalization:
Western Blot Quantification:
Use gradient gels (4-20%) for optimal separation and resolution of the 19 kDa HES2 band
Load equal amounts of protein (validated by BCA or Bradford assay)
Include loading controls appropriate for nuclear proteins (e.g., Lamin B1, Histone H3)
Use fluorescent secondary antibodies or digital chemiluminescence systems for wider linear dynamic range
Analyze band intensities using software such as ImageJ, normalizing to loading controls
qPCR Analysis of HES2 mRNA Levels:
Design gene-specific primers spanning exon-exon junctions to avoid genomic DNA amplification
Validate primer efficiency using standard curves
Use multiple reference genes (GAPDH, ACTB, 18S rRNA) for normalization
Analyze data using the 2^(-ΔΔCt) method or absolute quantification with standard curves
Immunohistochemistry Quantification:
Use digital image analysis software to quantify staining intensity
Develop scoring systems based on both staining intensity and percentage of positive cells
Implement automated tissue analysis platforms for unbiased assessment
Include calibration standards in each staining batch to account for batch-to-batch variation
Flow Cytometry for Single-Cell Analysis:
HES2 expression has been documented in various cancer types, including pancreatic cancer, colon cancer with RER, cervical cancer, and head and neck tumors . The following methodological approaches can help elucidate its role in cancer:
Tissue Microarray Analysis:
Cell Line Models:
Screen cancer cell line panels for HES2 expression using Western blotting (1:500-1:2000 dilution)
Generate stable HES2 knockdown or overexpression cell lines
Assess effects on proliferation, migration, invasion, and therapy resistance
Investigate synergy with Notch pathway inhibitors (γ-secretase inhibitors, anti-Notch antibodies)
Xenograft Models:
Develop in vivo models with modulated HES2 expression
Use HES2 antibodies for IHC analysis of tumor sections
Correlate HES2 expression with tumor growth, angiogenesis, and metastatic potential
Assess effects of Notch pathway modulation on HES2 expression in vivo
Patient-Derived Models:
Analyze HES2 expression in patient-derived xenografts and organoids
Correlate expression with treatment response
Implement HES2 as a potential biomarker for patient stratification in clinical trials
HES2's role in development involves complex interactions with various transcriptional regulators:
Co-Immunoprecipitation Studies:
Use HES2 antibodies validated for IP applications to pull down protein complexes
Focus on known interactors like TLE proteins that bind via the WRPW motif
Analyze samples from different developmental stages to track temporal dynamics of interactions
Implement tandem affinity purification for higher purity complex isolation
Proximity Ligation Assays (PLA):
Visualize in situ protein-protein interactions at single-molecule resolution
Combine HES2 antibodies with antibodies against potential interaction partners
Analyze spatial and temporal patterns of interactions during development
Quantify interaction frequencies in different cell types or developmental stages
ChIP-reChIP (Sequential ChIP):
Identify genomic loci co-occupied by HES2 and other transcription factors
First immunoprecipitate with HES2 antibody, then re-immunoprecipitate with antibody against potential partner
Compare binding profiles at different developmental timepoints
Integrate with gene expression data to identify cooperatively regulated targets
CRISPR-Based Approaches:
Generate tagged endogenous HES2 for more physiological interaction studies
Implement domain-specific mutations to disrupt specific interactions
Use CUT&RUN or CUT&Tag methods with HES2 antibodies for higher resolution chromatin binding profiles
Combine with single-cell approaches to address cellular heterogeneity
Emerging imaging technologies offer new opportunities for studying HES2 dynamics:
Live-Cell Imaging Approaches:
Use fluorescently tagged HES2 constructs to track protein dynamics
Implement FRAP (Fluorescence Recovery After Photobleaching) to analyze protein mobility
Apply FLIM-FRET (Fluorescence Lifetime Imaging Microscopy - Förster Resonance Energy Transfer) to study protein-protein interactions in real-time
Correlate with fixed-cell immunofluorescence using HES2 antibodies for validation
Super-Resolution Microscopy:
Apply techniques like STORM, PALM, or SIM for nanoscale localization of HES2
Use HES2 antibodies with appropriate fluorophores optimized for super-resolution imaging
Investigate subnuclear localization and potential association with specific nuclear domains
Combine with proximity labeling approaches to identify proteins in the immediate vicinity
Correlative Light and Electron Microscopy (CLEM):
Use gold-conjugated secondary antibodies to visualize HES2 localization at ultrastructural level
Correlate immunofluorescence patterns with electron microscopy data
Investigate association with specific nuclear substructures at nanometer resolution
Expansion Microscopy:
Apply physical expansion of samples to achieve super-resolution with standard microscopes
Optimize HES2 antibody immunostaining protocols for expanded samples
Visualize fine details of nuclear organization and HES2 distribution
CRISPR-based approaches offer powerful tools for HES2 functional studies:
Guide RNA Design:
Validation Strategies:
Functional Readouts:
Advanced CRISPR Applications:
Implement CRISPRi/CRISPRa for reversible modulation of HES2 expression
Use CRISPR base editors for precise mutation introduction without double-strand breaks
Apply CRISPR screening approaches to identify genetic interactors
Generate cell/animal models with fluorescently tagged endogenous HES2 for live imaging