Human Genome Sciences developed multiple monoclonal antibodies targeting critical disease pathways. Two prominent examples include:
Target: CCR5 chemokine receptor (HIV-1 co-receptor)
Mechanism: Blocks HIV-1 entry into CD4+ T-cells by binding CCR5 receptors .
This trial demonstrated proof of concept for CCR5 inhibition . A follow-up candidate, HGS101, showed 5.5-fold greater potency in preclinical studies .
Target: TRAIL-R1 (TNF-related apoptosis-inducing ligand receptor 1)
Indication: Anticancer therapy via apoptosis induction .
| Property | Data |
|---|---|
| Tumor Models Tested | NSCLC (H2122), Colo205 colon, A498 renal |
| Dosage Efficacy | 10 mg/kg induced 97% tumor regression |
| Half-Life (Mice) | 6.9–8.7 days |
| Combination Therapy | Synergy with chemotherapeutic agents |
HGS-ETR1 activated extrinsic (caspase-8) and intrinsic (mitochondrial) apoptosis pathways, showing broad-spectrum antitumor activity .
Antibodies targeting the HGS protein (e.g., Proteintech 10390-1-AP) are used in molecular studies:
Target: Hepatocyte Growth Factor-Regulated Tyrosine Kinase Substrate
Applications:
| Role | Mechanism |
|---|---|
| Growth Suppression | Inhibits IL-2/GM-CSF-mediated DNA synthesis |
| Tumor Suppression | Requires interaction with Schwannomin/merlin |
| STAT Signaling | Modulates cytokine-driven STAT activation |
This antibody aids in studying HGS's role in endosomal sorting and tumor suppression .
| Parameter | HGS004 (Anti-CCR5) | HGS-ETR1 (Anti-TRAIL-R1) | HGS Antibody 10390-1-AP |
|---|---|---|---|
| Primary Use | HIV-1 therapy | Cancer treatment | Research applications |
| Development Stage | Phase 1 (2006) | Phase I/II (2005) | Commercial reagent |
| Key Strength | CCR5 tropism blockade | Multitumor efficacy | Mechanistic studies |
| Limitations | Lower potency vs. HGS101 | Requires combination therapy | Not therapeutic-grade |
HIV Therapeutics: HGS004 paved the way for CCR5-targeted therapies, though newer candidates like HGS101 showed superior preclinical profiles .
Oncology: HGS-ETR1's tumor regression in xenografts (e.g., 97% reduction in NSCLC models) supported further trials in solid and hematological cancers .
Basic Science: Anti-HGS antibodies remain critical for elucidating endosomal trafficking and growth signaling pathways .
HGS (Hepatocyte growth factor-regulated tyrosine kinase substrate), also known as Hrs or Protein pp110, is a crucial component of intracellular signaling and trafficking pathways. Understanding its functions is fundamental to many areas of cell biology research.
HGS serves several critical cellular functions:
Acts as a key component in intracellular signal transduction mediated by cytokines and growth factors
When associated with STAM, suppresses DNA signaling upon stimulation by IL-2 and GM-CSF
Functions as a direct effector of PI3-kinase in vesicular pathways via early endosomes
Regulates trafficking between early and late endosomes by recruiting clathrin
Concentrates ubiquitinated receptors within clathrin-coated regions
Forms the ESCRT-0 complex with STAM, which recognizes ubiquitinated receptors and transfers them to lysosomal sorting/trafficking processes
May contribute to efficient recruitment of SMADs to the activin receptor complex
Participates in receptor recycling through the CART complex, required for transferrin receptor recycling
These diverse functions make HGS antibodies essential tools for studying membrane trafficking, receptor degradation, and signal transduction mechanisms.
HGS antibodies can be utilized in multiple research applications, each with specific experimental considerations:
| Application | Suitability | Typical Working Dilution | Notes |
|---|---|---|---|
| Western Blot (WB) | High | 0.1-1 μg/mL | Detects ~86 kDa band in human samples |
| Immunoprecipitation (IP) | High | 6 μg per reaction | Effective for protein complex isolation |
| Immunofluorescence (IF) | Moderate | 1:100-1:500 | Useful for subcellular localization studies |
| Immunohistochemistry (IHC) | Moderate | 1:100-1:200 | Works on fixed tissue sections |
| ELISA | Variable | Antibody-dependent | Check manufacturer specifications |
For optimal results in Western blot applications, HGS antibodies have been validated with as little as 5 μg of total protein from HeLa cell lysates, with clear band detection at 0.1 μg/mL antibody concentration and 10-second exposure time .
Proper validation of HGS antibodies is crucial for ensuring experimental reliability. Follow these methodological steps:
Positive and negative controls: Test the antibody on samples known to express or lack HGS. For human samples, HeLa cells serve as reliable positive controls expressing endogenous HGS .
Concentration gradients: Test multiple antibody concentrations and sample loadings. For example, with ab72053, testing 5-50 μg of total lysate with 0.1-0.4 μg/mL antibody concentration provides a good validation range .
Cross-reactivity assessment: If working with non-human samples, verify species cross-reactivity. Some HGS antibodies raised against human epitopes have demonstrated reactivity with mouse samples, as shown with Renca (mouse renal adenocarcinoma) cell lysates .
Blocking peptide competition: Use the immunizing peptide to confirm specificity, particularly for antibodies generated against synthetic peptides corresponding to specific HGS regions.
Knockout/knockdown validation: If available, test the antibody on HGS-knockout or HGS-knockdown samples to confirm specificity.
Multiple antibody comparison: When possible, compare results from different antibodies targeting distinct epitopes of HGS.
Effective sample preparation significantly impacts HGS detection in Western blots:
Lysis buffer selection: NETN lysis buffer (containing NP-40) has been validated for effective HGS extraction and detection in Western blots. This non-denaturing buffer helps maintain protein structure while sufficiently solubilizing membrane-associated HGS .
Protein denaturation: Use standard SDS-PAGE sample buffer with reducing agent, and heat samples to 95°C for 5 minutes to ensure complete protein denaturation.
Sample loading: For optimal HGS detection, load between 5-50 μg of total protein depending on expression levels. A gradient of protein loading (e.g., 5, 15, and 50 μg) helps determine the linear detection range .
Gel percentage: Use 8-10% polyacrylamide gels for optimal resolution of HGS, which has a predicted molecular weight of 86 kDa.
Transfer conditions: Semi-dry or wet transfer methods both work effectively, but wet transfer may be preferable for larger proteins like HGS.
Blocking: 5% non-fat dry milk or BSA in TBST provides effective blocking to minimize background.
Co-immunoprecipitation (co-IP) of HGS with other ESCRT-0 components requires careful optimization:
Antibody selection: Choose HGS antibodies validated for IP applications. The ab72053 antibody has demonstrated successful IP of HGS from HeLa cell lysates at 6 μg per reaction .
Lysis conditions: Use NETN buffer (20 mM Tris-HCl pH 8.0, 100 mM NaCl, 1 mM EDTA, 0.5% NP-40) supplemented with protease and phosphatase inhibitors. This maintains the integrity of protein-protein interactions while effectively solubilizing membrane-associated complexes .
Pre-clearing: Pre-clear lysates with Protein A/G beads to reduce non-specific binding.
Antibody incubation: Incubate 0.5-1.0 mg of pre-cleared lysate with 6 μg of HGS antibody overnight at 4°C with gentle rotation.
Bead binding: Add fresh Protein A/G beads and incubate for 1-3 hours at 4°C.
Washing stringency: Perform at least 4-5 washes with lysis buffer containing reduced detergent (0.1-0.2% NP-40) to maintain specific interactions while removing background.
Elution conditions: Elute bound proteins with 2X SDS sample buffer at 95°C for 5 minutes.
Western blot detection: For detecting co-immunoprecipitated STAM or other ESCRT-0 components, use 20% of IP material for optimal visualization .
This approach preserves the integrity of the ESCRT-0 complex while minimizing non-specific interactions.
HGS localizes to multiple subcellular compartments, primarily early endosomes, requiring specific detection strategies:
Fixation method: For immunofluorescence, 4% paraformaldehyde fixation preserves endosomal structures while maintaining HGS antigenicity. Avoid methanol fixation which can disrupt membrane structures.
Permeabilization: Use 0.1-0.2% Triton X-100 or 0.05% saponin for balanced permeabilization that maintains endosomal integrity while allowing antibody access to HGS epitopes.
Co-localization markers: Include established markers to identify specific compartments:
Early endosomes: EEA1, Rab5
Multivesicular bodies: CD63, LAMP1
Clathrin-coated structures: Clathrin heavy chain
Subcellular fractionation: For biochemical analysis, use established fractionation protocols to isolate early endosomes, MVBs, and cytosolic fractions separately before Western blot analysis.
Super-resolution microscopy: Consider techniques like STED or STORM microscopy for precise localization of HGS within endosomal structures.
Live-cell imaging: For dynamic studies of HGS trafficking, consider fluorescently tagged HGS constructs alongside immunostaining to validate localization patterns.
HGS plays a critical role in receptor degradation through the ESCRT-0 complex. These methodological approaches can quantify this function:
Receptor internalization assays: Track receptor levels over time using antibodies against both HGS and the receptor of interest (e.g., EGFR, PDGFR).
Co-immunoprecipitation with ubiquitinated receptors: Use HGS antibodies to co-immunoprecipitate ubiquitinated receptors, demonstrating direct interaction:
Proximity ligation assays (PLA): Use HGS antibodies paired with receptor antibodies to visualize and quantify their interaction in situ.
Pulse-chase analysis: Combine with HGS knockdown/knockout to demonstrate its role in regulating receptor half-life.
Multivesicular body formation assay: Use HGS antibodies alongside markers of MVB formation to assess ESCRT-0 function in receptor sorting.
Super-resolution microscopy: Visualize co-localization of HGS with receptors during the degradation process with nanometer precision.
Recent advances have improved antibody development approaches applicable to HGS research:
Single B cell screening technologies: These methods allow for direct isolation of B cells producing antibodies with desired specificity against HGS:
Golden Gate-based dual-expression vector systems: This technology links heavy and light chain variable fragments from a single B cell:
Next-generation sequencing (NGS) integration: NGS technology revolutionizes the screening of HGS-specific antibodies:
Recombinant antibody production: Compared to traditional hybridoma techniques, recombinant approaches offer advantages:
Non-specific binding can complicate HGS detection in tissues. Address this methodically:
Antibody titration: Determine the optimal antibody concentration that maximizes specific signal while minimizing background. Test concentrations ranging from 0.1-1 μg/mL for Western blots and 1:100-1:500 for immunohistochemistry .
Blocking optimization: Test different blocking reagents:
5% BSA for phosphorylated epitopes
5% non-fat dry milk for general applications
Commercial blocking reagents for challenging tissues
Antigen retrieval methods: For IHC applications, compare:
Heat-induced epitope retrieval (citrate buffer, pH 6.0)
Enzymatic retrieval (proteinase K)
High pH retrieval (EDTA buffer, pH 9.0)
Control experiments:
Pre-absorb antibody with immunizing peptide
Include HGS-negative tissues or knockdown samples
Test secondary antibody alone to assess non-specific binding
Signal amplification alternatives: Consider tyramide signal amplification for low-abundance detection while maintaining specificity.
Cross-reactivity assessment: If working with non-human tissues, verify species cross-reactivity using sequence alignment of the epitope region.
Effective experimental design for studying HGS functions requires careful planning:
HGS function is regulated by phosphorylation, requiring specific experimental controls:
Phosphorylation-specific antibodies: Use antibodies that specifically recognize phosphorylated HGS at key regulatory sites.
Phosphatase controls: Include samples treated with phosphatases to confirm specificity of phosphorylation detection.
Kinase manipulation: Use inhibitors or activators of relevant kinases (e.g., PI3K inhibitors) to modulate HGS phosphorylation.
Phosphomimetic and phospho-dead mutants: Generate HGS constructs with mutations at phosphorylation sites:
Phosphomimetic (S/T to D/E)
Phospho-dead (S/T to A)
Stimulation controls: Include appropriate positive controls known to induce HGS phosphorylation:
Growth factors (HGF, EGF)
Cytokines (IL-2, GM-CSF)
Time-course analysis: Capture the dynamic nature of phosphorylation with appropriate time points.
Sample preparation: Use phosphatase inhibitors in all buffers to preserve phosphorylation status during extraction.
Quantitative colocalization analysis requires rigorous methodology:
Image acquisition parameters:
Use identical settings for all samples
Capture images below pixel saturation
Acquire sufficient Z-sections for 3D analysis
Marker selection: Include established markers:
Early endosomes: EEA1, Rab5
Late endosomes/MVBs: Rab7, CD63
Recycling endosomes: Rab11
Quantification methods:
Pearson's correlation coefficient
Manders' overlap coefficient
Object-based colocalization
Analysis software options:
ImageJ with Coloc2 plugin
CellProfiler
Commercial imaging software
Statistical approach:
Analyze multiple cells (>30)
Include biological replicates
Apply appropriate statistical tests
Visualization: Present data as both representative images and quantitative graphs showing colocalization metrics with statistical significance.
When different HGS antibodies yield conflicting results, follow this analytical approach:
Epitope mapping: Compare the epitope regions recognized by each antibody:
Post-translational modifications: Consider whether modifications might mask certain epitopes:
Phosphorylation
Ubiquitination
Conformational changes
Isoform specificity: Determine if antibodies detect different HGS isoforms or splice variants.
Validation approach: Perform additional validation:
siRNA knockdown/CRISPR knockout controls
Immunoprecipitation followed by mass spectrometry
Peptide competition assays
Application-specific differences: Some antibodies perform better in certain applications than others:
Native vs. denatured conditions
Fixed vs. unfixed samples
Specific buffer compositions
Systematic comparison: Test antibodies side-by-side under identical conditions and document differences in sensitivity, specificity, and background.
Studying HGS-receptor interactions presents several challenges that require specific solutions:
Transient interactions: HGS-receptor interactions may be short-lived:
Use chemical crosslinking before lysis
Apply proximity-based methods (BioID, APEX)
Consider live-cell imaging approaches
Ubiquitination dependence: Many HGS-receptor interactions require receptor ubiquitination:
Include deubiquitinase inhibitors in lysis buffers
Verify ubiquitination status of receptors
Consider using ubiquitination-defective receptor mutants as controls
Compartment disruption: Standard lysis conditions may disrupt endosomal compartments:
Overexpression artifacts: Overexpressed HGS may form abnormal aggregates:
Use endogenous HGS when possible
Validate with multiple expression levels
Consider inducible expression systems
Context dependence: HGS-receptor interactions may depend on cell type and conditions:
Include appropriate physiological stimuli
Compare results across multiple cell types
Consider tissue-specific factors
Technological advances are transforming HGS research approaches:
CRISPR/Cas9 genome editing: Enables precise modification of endogenous HGS:
Knockout models for loss-of-function studies
Knock-in of fluorescent tags for live imaging
Introduction of specific mutations at regulatory sites
Proximity labeling methods: BioID and APEX2 technologies identify transient HGS interaction partners:
Fusion of promiscuous biotin ligase to HGS
Identification of proximal proteins by streptavidin pulldown
Mass spectrometry analysis of the HGS "interactome"
Advanced microscopy techniques:
Super-resolution microscopy (STED, PALM, STORM) for nanoscale localization
Live-cell lattice light-sheet microscopy for 4D imaging of trafficking
Correlative light and electron microscopy for ultrastructural context
Single-cell approaches: Analysis of HGS function with single-cell resolution:
Single-cell RNA-seq for expression correlation
Single-cell proteomics for protein-level analysis
Microfluidic approaches for dynamic studies
Structural biology integration: Combining antibody-based detection with structural insights:
Cryo-EM of HGS-containing complexes
Integrative structural modeling
Structure-guided antibody development
HGS antibodies have potential applications in emerging research areas:
Cancer research: HGS may influence tumor progression through:
Altered receptor tyrosine kinase trafficking
Modified signaling pathway duration and intensity
Potential diagnostic or prognostic biomarker applications
Neurodegenerative disease research: Emerging roles in:
Neuronal endolysosomal trafficking
Clearance of aggregated proteins
Synaptic receptor recycling
Immunological studies: Functions in:
Developmental biology: Potential roles in:
Growth factor receptor regulation during development
Embryonic signaling pathway modulation
Tissue-specific receptor trafficking patterns
Therapeutic target validation: HGS as a potential target to modulate:
Receptor degradation rates in disease settings
Signaling pathway duration and intensity
Cellular responses to growth factors and cytokines
Each of these areas represents an opportunity to apply HGS antibodies in novel contexts beyond their traditional use in basic endosomal biology research.