LGR5 is a G protein-coupled receptor that functions as a receptor for R-spondins, potentiating the canonical Wnt signaling pathway. The canonical LGR5 protein is 907 amino acids in length with a molecular weight of approximately 100 kDa, featuring Golgi and membrane subcellular localization . LGR5 serves as a biomarker for adult stem cells in specific tissues and has gained significant attention due to its overexpression in multiple cancer types, particularly colorectal cancer (CRC), hepatocellular carcinoma (HCC), and pre-B-ALL tumors .
LGR5 functions in G protein-coupled receptor activity and G protein-coupled peptide receptor activity, playing a crucial role in signal transduction . Unlike classical G-protein coupled receptors, LGR5 does not activate heterotrimeric G-proteins to transduce signals but instead associates with phosphorylated LRP6 and frizzled receptors upon binding to R-spondins . This mechanism is central to its role in stem cell maintenance and cancer development.
Researchers can access a diverse range of LGR5 antibodies:
Monoclonal antibodies: Highly specific antibodies like clone OTI2A2 that target defined epitopes
Polyclonal antibodies: Generated against specific regions of the LGR5 protein
Domain-specific antibodies: Some targeting the extracellular domain (N-terminal 101 amino acids), which is particularly valuable for live cell applications
Species-specific antibodies: Many are reactive with human LGR5, while some cross-react with other species including mouse, rat, and cynomolgus macaque
Therapeutic antibody formats: Including antibody-drug conjugates (ADCs), bispecific T-cell engagers (BiTEs), and chimeric antigen receptor (CAR) constructs
Over 700 anti-LGR5 antibodies from more than 30 different suppliers are available for various applications such as Western blot, ELISA, Flow Cytometry, IHC, and Immunofluorescence .
Selecting the appropriate LGR5 antibody requires consideration of several factors:
Application compatibility: Ensure the antibody has been validated for your specific application (Western blot, IHC, flow cytometry, etc.)
Epitope location: For detecting cell surface LGR5, choose antibodies targeting the extracellular domain
Species reactivity: Verify cross-reactivity with your experimental model organism (human, mouse, rat, etc.)
Clonality: Monoclonal antibodies offer higher specificity while polyclonals may provide stronger signals in certain applications
Validation data: Review published literature and supplier validation data showing specificity testing against related proteins like LGR4 and LGR6
For example, when studying LGR5 in live cells, antibodies targeting the N-terminal extracellular domain (like those described in search result ) are preferable as they can bind the native conformation of the protein without requiring cell fixation and permeabilization.
For effective LGR5 detection in tissue samples by immunohistochemistry, follow this optimized protocol:
Sample preparation:
Fix tissues in 10% neutral-buffered formalin
Process and embed in paraffin
Section at 4-5 μm thickness
Staining protocol:
Deparaffinize and rehydrate sections
Perform antigen retrieval with 10 mM citrate buffer (pH 6.0)
Block endogenous peroxidase with 3% H₂O₂ in PBS for 4 minutes
Block endogenous biotin using an Avidin/Biotin Blocking Kit
Block non-specific binding with 10% donkey serum in 3% BSA/PBS
Incubate with primary anti-LGR5 antibody (4 μg/ml) for 60 minutes at room temperature
Apply biotinylated secondary antibody (e.g., biotinylated donkey anti-rabbit IgG) for 30 minutes
Treat with Vectastain ABC Elite HRP for 30 minutes
Visualize with metal-enhanced DAB for 5 minutes
Critical considerations:
Include positive controls (e.g., intestinal crypts) and negative controls (isotype antibody)
Evaluate slides in a double-blinded fashion using established scoring criteria
For multi-label studies, carefully select compatible detection systems
This protocol has been validated for detecting LGR5 in various tissue types including colorectal cancer samples .
Thorough validation of LGR5 antibodies is critical for reliable results:
Western blot validation:
Test against cells overexpressing LGR5 (e.g., transfected HEK293T cells)
Compare with non-transfected controls and cells expressing related proteins (LGR4, LGR6)
Verify the expected molecular weight (~100 kDa)
Perform peptide competition assays with specific epitope fragments
Flow cytometry validation:
Compare staining patterns between LGR5-expressing and non-expressing cells
Use isotype controls at matching concentrations
Confirm specificity through blocking peptides (e.g., Frag1A as described in search result )
Analyze co-expression with established markers in relevant cell populations
Immunofluorescence validation:
Co-localize with tagged LGR5 constructs (e.g., LGR5-GFP)
Demonstrate absence of staining in knockout or knockdown models
Compare subcellular localization patterns with established literature
For example, search result describes validating antibody specificity by testing against human LGR5, cynomolgus LGR5, and related human LGR4, LGR6, and murine Lgr4/Lgr5 expressed in HEK293T cells, demonstrating specific detection of only human and cynomolgus LGR5.
For optimal LGR5 detection by flow cytometry:
Sample preparation:
Harvest cells using gentle dissociation methods (e.g., non-enzymatic cell dissociation solutions)
For tissues, prepare single-cell suspensions using appropriate tissue-specific dissociation protocols
Maintain cold conditions throughout to preserve cell surface antigens
Staining protocol:
Block Fc receptors with appropriate blocking solution
Stain with fluorophore-conjugated anti-LGR5 or primary/secondary antibody combinations
Include viability dye to exclude dead cells
Add appropriate compensation controls
Critical parameters:
Antibody concentration: Typically 1-10 μg/ml, titrate for optimal signal-to-noise ratio
Staining time: 30-60 minutes on ice
Washing steps: Gentle but thorough to reduce background
Essential controls:
Isotype control matched to primary antibody
FMO (Fluorescence Minus One) controls
For example, search result shows flow cytometry detection of LGR5 in NS0 mouse cell line transfected with human LGR5 and eGFP, comparing specific staining with isotype control and demonstrating clear separation of positive and negative populations.
Successfully detecting LGR5 by Western blot requires attention to several critical parameters:
Sample preparation:
Use appropriate lysis buffers containing detergents (e.g., RIPA buffer with 1% NP-40 or Triton X-100)
Include protease inhibitors to prevent degradation
For membrane proteins like LGR5, avoid excessive heating (65°C for 5 minutes instead of boiling)
Consider membrane fraction enrichment for enhanced detection
Electrophoresis and transfer conditions:
Use gradient gels (4-12%) for optimal resolution of the 100 kDa LGR5 protein
Transfer to PVDF membranes (preferred over nitrocellulose for high molecular weight proteins)
Use low SDS transfer buffer and longer transfer times (90-120 minutes) or semi-dry transfer systems
Consider wet transfer at 4°C overnight for large proteins
Antibody incubation:
Recommended dilutions: 1:500-1:1000 for most anti-LGR5 antibodies
Block with 5% non-fat milk or BSA (depending on antibody recommendations)
Incubate primary antibody overnight at 4°C for optimal binding
Use validated secondary antibody at appropriate dilution (typically 1:2000-1:5000)
Signal detection:
Use enhanced chemiluminescence (ECL) detection with sufficient sensitivity
Adjust exposure times to prevent oversaturation while capturing authentic signals
Consider using signal enhancers for low-abundance samples
Troubleshooting weak signals:
Increase protein loading (up to 50-75 μg per lane)
Reduce washing stringency
Increase antibody concentration or incubation time
Use signal amplification systems
This approach has been validated for detecting the canonical 100 kDa LGR5 protein in various tissue samples and cell lines .
Isolating and characterizing LGR5-positive cells requires a systematic approach:
Cell isolation strategies:
Flow cytometry-based sorting:
Use antibodies against the extracellular domain of LGR5
Implement gentle sorting parameters to maintain viability
Include viability dyes to exclude dead cells
Consider double-sorting for higher purity
Magnetic-activated cell sorting (MACS):
Generally gentler than FACS, potentially preserving stem cell properties
May yield higher cell numbers but with lower purity
Reporter systems (when applicable):
LGR5-GFP knock-in systems provide alternative isolation methods
Useful for animal models but less applicable to primary human samples
Functional characterization:
In vitro stem cell assays:
Sphere formation efficiency in low-attachment conditions
Organoid formation capacity
Colony formation efficiency
Differentiation potential assessments
Molecular characterization:
Gene expression profiling (qPCR for stem cell markers)
Assessment of Wnt pathway activity
Analysis of self-renewal and differentiation markers
In vivo assays:
Xenotransplantation at limiting dilutions to assess tumor-initiating capacity
Lineage tracing studies (in appropriate models)
Validation of LGR5-positivity:
Confirm LGR5 expression in sorted populations by alternative methods (qPCR, Western blot)
Compare with established LGR5+ model systems
Correlate LGR5 expression with functional stem cell properties
These approaches have been successfully applied to isolate and characterize LGR5+ cancer stem cells from colorectal cancer, hepatocellular carcinoma, and pre-B-ALL samples .
Implementing appropriate controls is critical for reliable interpretation of results with LGR5 antibodies:
Positive controls:
Cell lines with confirmed high LGR5 expression (e.g., NALM6, LoVo cells)
Tissues known to express LGR5 (e.g., intestinal crypts)
LGR5-transfected cells (e.g., HEK293T cells overexpressing LGR5)
Negative controls:
Cell lines lacking LGR5 expression (e.g., untransfected HEK293T cells)
Tissues with minimal/no LGR5 expression
Secondary antibody-only controls
Specificity controls:
Pre-incubation with blocking peptide (e.g., Frag1A as described in search result )
Non-binding antibody variant (e.g., α-LGR5v6 mentioned in search result )
siRNA or CRISPR knockout validation where feasible
Application-specific controls:
For Western blot: Molecular weight markers, loading controls
For IHC/ICC: Adjacent sections with isotype control
For flow cytometry: FMO controls, viability dyes
For functional assays: Non-functional antibody variants (e.g., α-LGR5-ADC NC)
For example, search result describes using a non-binding version of an antibody (α-LGR5v6) generated during the humanization process as a negative control for binding studies, and using a non-cleavable version of an antibody-drug conjugate (α-LGR5-ADC NC) as a specificity control for cell killing assays.
Developing LGR5 antibodies as cancer therapeutics involves several strategic approaches:
Antibody-Drug Conjugates (ADCs):
Antibody selection:
Linker-payload optimization:
Efficacy evaluation:
In vitro cytotoxicity against LGR5-high vs. LGR5-low cell lines
In vivo tumor regression studies in xenograft models
Assessment of safety profile and therapeutic window
Research has demonstrated that α-LGR5-ADC effectively kills LGR5+ cancer cells with an EC50 of approximately 10 nM and has shown potent anti-tumor efficacy in a murine model of human NALM6 pre-B-ALL, reducing tumor burden to less than 1% of control treatment .
Bispecific T-cell Engagers (BiTEs):
Design considerations:
Optimal architecture for T-cell recruitment
Balanced affinity for LGR5 vs. CD3
Size and stability optimization
Effectiveness:
Chimeric Antigen Receptor (CAR) T-cells:
Design elements:
LGR5-specific single-chain variable fragments (scFvs)
Selection of appropriate costimulatory domains
Vector design and T-cell manufacturing considerations
Performance metrics:
These therapeutic modalities represent promising approaches to targeting LGR5-expressing cancers, with ADCs currently showing the most robust efficacy in preclinical models .
Distinguishing between different pools of LGR5 protein requires sophisticated approaches:
Subcellular localization analysis:
Confocal microscopy with compartment markers:
Co-staining with membrane markers (Na+/K+ ATPase, WGA)
Golgi markers (GM130, TGN46)
Endosomal/lysosomal markers (EEA1, LAMP1)
Use z-stack imaging to confirm true co-localization
Subcellular fractionation:
Separate membrane, cytosolic, and organelle fractions
Western blot analysis of fractions with LGR5 antibodies
Include fraction-specific markers as controls
Functional pools characterization:
Active vs. inactive forms:
Surface biotinylation to quantify membrane-localized fraction
Co-immunoprecipitation with pathway components (R-spondins, Frizzled, LRP6)
Phosphorylation status assessment of associated signaling molecules
Dynamic trafficking studies:
Pulse-chase experiments with labeled antibodies
Photoactivatable or photoconvertible LGR5 fusion proteins
Live-cell imaging to track protein movement
Post-translational modification analysis:
Glycosylation status (PNGase F treatment)
Phosphorylation patterns (phospho-specific antibodies or mass spectrometry)
Ubiquitination status (affects degradation and trafficking)
Advanced molecular techniques:
Proximity ligation assays to detect specific LGR5 interactions
FRET/BRET approaches to study real-time molecular associations
Super-resolution microscopy for nanoscale localization patterns
These approaches help distinguish between functionally relevant LGR5 pools (active surface receptors) and processing/storage pools (Golgi, endosomes), providing deeper insights into LGR5 biology in normal and cancer cells.
When faced with contradictory results using different LGR5 antibody clones, implement these systematic approaches:
Epitope mapping analysis:
Determine the exact epitopes recognized by each antibody clone
Assess epitope accessibility in different experimental conditions
Evaluate potential epitope masking by protein interactions or modifications
Clone-specific validation:
Test each clone against:
Overexpression systems with tagged LGR5
Knockout or knockdown models
Purified recombinant LGR5 domains
Binding kinetics analysis:
| Antibody Clone | Kd (nM) for Frag1A | Binding to Frag1B |
|---|---|---|
| α-LGR5 clone 1 | 0.76 ± 0.01 | Not detectable |
| α-LGR5 clone 2 | 1.1 ± 0.01 | Not detectable |
| α-LGR5 clone 3 | 1.0 ± 0.01 | Not detectable |
| α-LGR5 clone 4 | 1.4 ± 0.01 | Not detectable |
| α-LGR5v4 | 2.0 ± 0.02 | Not detectable |
| α-LGR5 clone 2-ADC | 2.7 ± 0.03 | Not detectable |
| α-LGR5v4-ADC | 2.0 ± 0.02 | Not detectable |
Orthogonal validation approaches:
mRNA expression analysis (RT-qPCR, in situ hybridization)
Mass spectrometry validation of protein identity
Combined antibody approaches using antibodies targeting different epitopes
Multi-condition testing:
Systematically compare antibody performance across:
Different fixation methods
Various permeabilization protocols
Range of antigen retrieval techniques
Multiple detection systems
Through comprehensive validation and careful consideration of technical variables, researchers can identify the most reliable antibodies for specific applications and experimental conditions.
Monitoring LGR5 antibody internalization is crucial for ADC development:
Quantitative internalization assays:
Flow cytometry-based methods:
Surface quenching assays (pH-sensitive fluorophores)
Acid wash techniques to remove surface-bound antibodies
Time-course analysis at multiple timepoints (15, 30, 60, 120 minutes)
Confocal microscopy approaches:
Live-cell imaging with fluorescently-labeled antibodies
Co-localization with endosomal/lysosomal markers
3D reconstruction to confirm complete internalization
Biochemical methods:
Surface biotinylation followed by streptavidin pulldown
Protease protection assays
ELISA-based internalization assays
Critical parameters to evaluate:
Internalization rate:
Half-time to internalization
Percentage internalized at steady state
Comparison with benchmark internalizing antibodies
Intracellular trafficking:
Endosomal escape efficiency
Lysosomal delivery kinetics (critical for cleavable linkers)
Recycling vs. degradation fate
Factors affecting internalization:
Correlation with ADC efficacy:
Compare internalization metrics with cytotoxicity data
Evaluate linker stability in different cellular compartments
Assess payload release kinetics in relation to trafficking patterns
Search result demonstrates that α-LGR5 antibodies are rapidly internalized by LGR5-overexpressing cell lines, making them suitable candidates for ADC development. The superior efficacy of cleavable-linker ADCs (α-LGR5-ADC) compared to non-cleavable versions (α-LGR5-ADC NC) highlights the importance of proper internalization and intracellular processing for effective ADC function.
Researchers frequently encounter these challenges when detecting LGR5:
Solution: Use signal amplification methods (tyramide signal amplification for IHC, high-sensitivity ECL for Western blot)
Approach: Employ cell enrichment strategies before analysis
Technique: Extend antibody incubation times (overnight at 4°C)
Solution: Test multiple antigen retrieval methods (heat-induced vs. enzymatic)
Approach: Use antibodies targeting different epitopes
Technique: Optimize fixation protocols (avoid overfixation)
Solution: Implement more stringent blocking (5% BSA, 10% serum)
Approach: Include absorption controls with specific blocking peptides (e.g., Frag1A)
Technique: Use highly specific monoclonal antibodies with validated specificity
Solution: Use confocal microscopy with membrane markers
Approach: Implement subcellular fractionation for Western blot analysis
Technique: Compare surface staining (non-permeabilized) with total staining (permeabilized)
Solution: Validate antibody specificity against LGR4 and LGR6 expression
Approach: Confirm findings with genetic approaches (siRNA, CRISPR)
Solution: Review antibody epitope location relative to known splice variants
Approach: Use multiple antibodies targeting different regions
Technique: Correlate protein detection with mRNA expression of specific variants
Addressing these challenges through rigorous controls and optimized protocols ensures more reliable detection and characterization of LGR5 in research applications.
Validating LGR5 antibody specificity against related family members requires a multi-faceted approach:
Comparative expression analysis:
Overexpression systems:
Test antibodies against cells overexpressing LGR5, LGR4, and LGR6
Use tagged constructs for parallel detection
Compare staining/blotting patterns between family members
Western blot analysis:
Run lysates from cells expressing different LGR family members
Compare band patterns and molecular weights
Perform peptide competition assays
Genetic validation:
Knockdown/knockout approaches:
siRNA or CRISPR-based depletion of LGR5
Confirm loss of signal with antibody
Test against LGR4/LGR6 knockdowns as controls
Heterologous expression:
Express LGR5 in cells lacking endogenous expression
Compare signal between transfected and non-transfected cells
Express sequence variants to map epitope requirements
Cross-reactivity testing:
Species cross-reactivity:
Domain-specific detection:
Immunoprecipitation analysis:
Pull down with anti-LGR5 antibody
Analyze precipitated proteins by mass spectrometry
Check for presence of related family members
For example, search result demonstrates that α-LGR5 antibody clones 1-4 detect human and cynomolgus LGR5 but show no immunoreactivity to murine Lgr4 and Lgr5 or the closely related human LGR4 and LGR6 in Western blot analysis of lysates from transfected HEK293T cells.