GRF5 Antibody

Shipped with Ice Packs
In Stock

Description

GRF5 and Its Biological Role

GRF5 belongs to the Growth-Regulating Factor (GRF) family, which promotes cell proliferation and organ growth in plants. Key features include:

  • Function: Directs leaf and root development by modulating cell division and expansion .

  • Regulation: Repressed by transcription factors like ARF2 (Arabidopsis) and GmFDLs (soybean) .

  • Expression: Localized in meristematic tissues, roots, shoots, and flowers .

Applications of GRF5 Antibody

GRF5 antibodies are utilized in diverse experimental workflows:

ApplicationMethodKey Use Cases
Protein DetectionWestern Blot (WB)Quantify GRF5 expression levels in mutants or overexpression lines .
LocalizationImmunohistochemistry (IHC)Map GRF5 distribution in plant tissues (e.g., leaf primordia, root tips) .
Binding StudiesChIP-qPCR, EMSAIdentify GRF5 target genes (e.g., GmGTG1, GmTAA1.1 in soybean) .
Functional AnalysisLuciferase AssaysAssess GRF5’s transcriptional activation/repression of promoters .

GRF5 Repression Mechanisms

  • ARF2 Interaction: In Arabidopsis, ARF2 binds directly to the GRF5 promoter, repressing its expression. Mutations in ARF2 increase GRF5 transcript levels >3-fold, leading to enhanced leaf growth .

  • miRNA Synergy: GRF5 works synergistically with miR396-regulated GRFs (e.g., GRF1–4). Combined loss of GRF5 and miR396 overexpression causes severe growth defects .

Soybean GRF5-1 Regulation

  • Direct Targets: GmGRF5-1 activates genes like GmGTG1 (chloroplast development) and GmTAA1.1 (auxin biosynthesis) .

  • Feedback Loop: GmGRF5-1 represses GmFTL3 (florigen), delaying flowering in overexpression lines .

Challenges and Future Directions

  • Validation: Antibody specificity remains critical, as GRFs share conserved domains .

  • Therapeutic Potential: While GRF5 is plant-specific, insights into antibody design (e.g., CDR optimization ) could improve reagent reliability for agricultural biotechnology.

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (made-to-order)
Synonyms
GRF5 antibody; At5g16050 antibody; F1N13_19014-3-3-like protein GF14 upsilon antibody; General regulatory factor 5 antibody
Target Names
GRF5
Uniprot No.

Target Background

Function
This antibody targets a protein associated with a DNA-binding complex that interacts with the G-box, a well-established cis-acting DNA regulatory element in plant genes. It may play a role in cell cycle regulation through its interaction with soluble EDE1, potentially sequestering it and rendering it inactive during early mitosis.
Gene References Into Functions
PMID: 25604530, GRF5 stimulates chloroplast division, leading to increased chloroplast number per cell and consequently higher chlorophyll levels., .
PMID: 21558460, 14-3-3 upsilon exhibits the strongest association with EDE1 in its free form, but also demonstrates a weaker interaction when EDE1 is microtubule-bound., .
Database Links

KEGG: ath:AT5G16050

STRING: 3702.AT5G16050.1

UniGene: At.25563

Protein Families
14-3-3 family
Subcellular Location
Cytoplasm. Nucleus.

Q&A

What is LGR5/GPR49 and why is it significant for stem cell research?

Leucine-rich repeat G-protein-coupled receptor 5 (LGR5), also called GPR49, is a seven-transmembrane glycoprotein receptor in the LGR family with significant implications for stem cell research. LGR5 serves as a critical marker found on embryonic and adult epithelial stem cells, making it valuable for identifying and isolating stem cell populations . The significance of LGR5 in research stems from several key factors:

  • LGR5 functions as a G-protein-independent mediator of R-Spondins' potentiating effect on Wnt signaling .

  • LGR5-positive stem cells possess the capacity to produce all epithelial cell types of the intestinal crypts .

  • LGR5 expression is frequently upregulated in stem cells that give rise to various cancers, including intestinal, hepatocellular, pancreatic, and ovarian carcinomas .

  • The ability to detect and track LGR5-expressing cells enables researchers to study epithelial stem cell dynamics in development, homeostasis, and disease conditions.

Methodologically, LGR5 antibodies provide a robust tool for identifying these stem cell populations through various detection techniques including immunohistochemistry, flow cytometry, and fluorescent immunocytochemistry, offering researchers reliable identification of these important cellular populations.

What detection methods can be used with LGR5/GPR49 antibodies?

LGR5/GPR49 antibodies can be utilized in multiple detection methods to visualize and quantify LGR5-expressing cells in various experimental contexts. Each method has specific advantages depending on research questions:

  • Flow Cytometry: Particularly effective for quantitative analysis of LGR5 expression in cell populations. Studies demonstrate successful detection in transfected cell lines where LGR5 antibodies can distinguish positive cells with high specificity compared to isotype controls . This method allows for simultaneous analysis of multiple parameters and potential sorting of LGR5-positive populations.

  • Immunocytochemistry/Immunofluorescence: Enables visualization of subcellular localization of LGR5. Published studies show LGR5 antibodies can detect the protein predominantly at cell surfaces and in cytoplasm of transfected cells . The method typically employs fluorophore-conjugated secondary antibodies (such as NorthernLights™ 557-conjugated Anti-Rat IgG) with counterstaining using nuclear markers like DAPI.

  • Immunohistochemistry: Allows detection of LGR5 in tissue contexts, preserving spatial relationships. This technique has been applied to various tissues including rectal epithelium in radiation proctitis studies and in dermal adipocyte lineage cells during skin development .

  • Functional Activity Assays: Beyond simple detection, some LGR5 antibodies can induce biological activity, such as TOPflash activity in cells expressing LGR5, providing functional readouts in addition to expression data .

For optimal results, researchers should perform antibody titration experiments to determine ideal concentrations for each application, as manufacturer recommendations suggest that "optimal dilutions should be determined by each laboratory for each application" .

How do I choose the right LGR5/GPR49 antibody for my specific research application?

Selecting the appropriate LGR5/GPR49 antibody requires careful consideration of multiple factors to ensure experimental success. The selection process should follow this methodological approach:

This systematic approach ensures selection of an antibody that will provide reliable, reproducible results for your specific research application.

How can computational modeling improve LGR5/GPR49 antibody design and specificity?

Computational modeling offers powerful approaches to enhance LGR5/GPR49 antibody design and specificity through several sophisticated methodologies:

  • Antibody Structure Prediction: Advanced computational techniques can model antibody structures with increasing accuracy. For non-H3 loops, canonical structure-based approaches achieve backbone RMSD values of approximately 0.7 Å when appropriate templates are available . For more variable regions like CDR H3, combined approaches using homology modeling with knowledge-based and energy-based methods generate more reliable predictions . These structural models serve as the foundation for rational antibody engineering.

  • Affinity Maturation In Silico: When antibody-antigen complex structures are available, computational methods can identify mutations likely to enhance binding affinity. This approach typically follows a multi-step process:

    • Initial evaluation using rigid protein backbones with discrete side-chain rotamer searches

    • Subsequent refinement using more accurate but computationally expensive models like Poisson-Boltzmann or Generalized Born continuum electrostatics

    • Unbound-state side-chain conformation searches and minimization

  • Epitope-Paratope Mapping: Tools like SnugDock (based on RosettaDock algorithm) can predict antibody-antigen complexes through alternating rounds of low-resolution rigid body perturbations and high-resolution side-chain and backbone minimization . This is particularly valuable for LGR5 research where understanding the precise binding interface can guide optimization of specificity.

  • Aggregation Prediction and Prevention: Computational methods can identify aggregate-prone regions (APRs) based on sequence composition and structural properties like hydrophobicity, charge, and secondary structure propensity . For LGR5 antibodies intended for therapeutic applications, this allows preemptive design of variants with improved stability and reduced immunogenicity risk.

  • Biophysics-Informed Modeling: Recent advances combine experimental selection data with computational modeling to disentangle multiple binding modes associated with specific ligands. This approach enables the prediction and generation of antibody variants with customized specificity profiles, even for discriminating very similar epitopes .

These computational approaches significantly expand the capabilities of traditional experimental antibody design, allowing researchers to explore a much broader sequence space and predict properties before experimental validation.

What strategies can resolve cross-reactivity issues with LGR5/GPR49 antibodies?

Cross-reactivity challenges with LGR5/GPR49 antibodies can significantly impact experimental outcomes, particularly when attempting to distinguish between closely related proteins or across species. Several methodological approaches can mitigate these issues:

  • Biophysics-Informed Computational Modeling: Recent advances employ models trained on experimentally selected antibodies to identify distinct binding modes associated with different potential ligands. This approach enables:

    • Prediction of cross-reactivity patterns based on binding mode analysis

    • Generation of novel antibody variants with customized specificity profiles

    • Design of antibodies that either specifically recognize a particular target or deliberately cross-react with multiple defined targets

  • Phage Display Selection with Negative Selection Strategies: Implementing sophisticated phage display protocols that include both positive selection against the target antigen (LGR5) and negative selection against potential cross-reactive antigens. This approach can be enhanced by:

    • Sequential rounds of alternating positive and negative selection

    • Inclusion of closely related family members (like LGR4 or LGR6) in negative selection steps

    • Utilizing computational analysis to identify sequences enriched in positive but not negative selection rounds

  • Epitope-Focused Engineering: When structural data is available, targeted modifications to antibody complementarity-determining regions (CDRs) can enhance specificity:

    • Analysis of antibody-antigen complex structures to identify key interaction residues

    • Strategic mutation of CDR residues that interact with regions of LGR5 that differ from homologous proteins

    • Application of affinity maturation techniques specifically focused on specificity-determining residues

  • Validation across Multiple Detection Systems: Comprehensive validation strategy using orthogonal methods:

    • Side-by-side comparison with different antibody clones recognizing distinct LGR5 epitopes

    • Testing in cell systems with controlled expression of LGR5 versus related proteins

    • Application of knockout/knockdown controls to confirm signal specificity

  • Species-Specific Considerations: When working across species, consider that mouse LGR5 shares 90% amino acid sequence identity with human LGR5 and 95% with rat LGR5 . This high homology may lead to cross-species reactivity, which can be either advantageous or problematic depending on experimental goals.

Implementation of these strategies requires integration of computational prediction, careful experimental design, and rigorous validation to ensure antibody specificity for the intended LGR5/GPR49 target.

How can I optimize immunohistochemistry protocols for detecting LGR5/GPR49 in different tissue types?

Optimizing immunohistochemistry (IHC) protocols for LGR5/GPR49 detection across diverse tissue types requires systematic adjustment of multiple parameters based on tissue-specific characteristics. Below is a methodological approach to optimizing your LGR5 IHC protocol:

  • Tissue-Specific Fixation Considerations:

    • Epithelial tissues (intestine, skin): Standard 10% neutral buffered formalin fixation for 24-48 hours typically preserves LGR5 epitopes

    • Neural tissues: Shorter fixation times (12-24 hours) may better preserve antigenicity

    • Fatty tissues (e.g., dermal adipocyte lineage cells): Consider fixatives with better penetration like Bouin's solution

    • Document successful applications in specific tissues, such as rectal epithelium in radiation proctitis studies

  • Antigen Retrieval Optimization:

    • Heat-Induced Epitope Retrieval (HIER): Test multiple pH conditions (pH 6.0 citrate buffer vs. pH 9.0 EDTA buffer)

    • Enzymatic Retrieval: For highly fixed tissues, test proteinase K or trypsin digestion at varying concentrations and incubation times

    • Combination Approaches: Sequential application of HIER followed by mild enzymatic treatment

  • Blocking Protocol Refinement:

    • Tissue-Specific Background: Adjust blocking solutions based on tissue characteristics

    • For high-background tissues: Increase blocking time (2-3 hours) and consider dual blocking with both serum and protein-based blockers

    • For tissues with endogenous biotin: Include avidin-biotin blocking steps if using biotin-based detection systems

  • Antibody Concentration and Incubation Conditions:

    • Perform systematic titration for each tissue type (starting with manufacturer recommendations)

    • Test multiple incubation temperatures (4°C, room temperature, 37°C)

    • Evaluate different incubation durations (1 hour to overnight)

    • Follow published successful applications, such as the 10 μg/mL concentration used for fluorescent ICC staining

  • Signal Amplification Selection:

    • For low-abundance LGR5 expression: Consider tyramide signal amplification

    • For co-localization studies: Use fluorescent secondary antibodies as demonstrated in published protocols

    • For chromogenic detection: Test different enzyme-substrate combinations (HRP-DAB, AP-Fast Red)

  • Validation Controls:

    • Positive Control: Include known LGR5-expressing tissues (intestinal crypts, specific cancer samples)

    • Negative Controls: Include isotype controls (like Rat IgG2B Isotype Control mentioned in the search results)

    • Absorption Controls: Pre-incubate antibody with recombinant LGR5 protein

    • Technical Controls: No-primary-antibody controls

  • Counterstaining Optimization:

    • Adjust hematoxylin intensity for chromogenic IHC

    • For fluorescent detection, select counterstains (like DAPI) that don't interfere with your fluorophores

By systematically optimizing each parameter for specific tissue types, researchers can develop robust protocols for detecting LGR5/GPR49 across diverse experimental contexts.

What are common causes of false positive or negative results when using LGR5/GPR49 antibodies?

False positive and negative results represent significant challenges in LGR5/GPR49 antibody-based detection. Understanding their causes and implementing mitigation strategies is essential for reliable experimental outcomes.

Causes of False Positive Results:

  • Cross-Reactivity with Related Proteins: LGR5 belongs to a family that includes highly similar proteins like LGR4 and LGR6 . The extracellular domains particularly share significant homology that may lead to antibody cross-recognition.
    Mitigation: Employ antibodies raised against unique regions of LGR5. Validate specificity using cells expressing individual LGR family members.

  • Non-Specific Binding in Tissue Sections: Particularly problematic in tissues with high extracellular matrix content or endogenous peroxidase/phosphatase activity.
    Mitigation: Optimize blocking protocols (use species-specific serum plus BSA or casein). Include appropriate quenching steps for endogenous enzymes.

  • Detection System Artifacts: Secondary antibody binding to endogenous immunoglobulins or Fc receptors.
    Mitigation: Include isotype controls (as shown in flow cytometry validation data where Rat IgG2B Isotype Control was used alongside anti-LGR5 antibody) .

  • Post-Translational Modifications: Altered glycosylation patterns may create epitopes recognized by certain antibodies.
    Mitigation: Compare results from antibodies recognizing different epitopes of LGR5.

Causes of False Negative Results:

  • Epitope Masking: Fixation procedures may alter protein conformation or cross-link epitopes.
    Mitigation: Test multiple antigen retrieval methods. The success of antibodies in both fixed cell immunocytochemistry and flow cytometry suggests optimized protocols can overcome this issue .

  • Low Expression Levels: LGR5 may be expressed at levels below detection thresholds in some tissues.
    Mitigation: Employ signal amplification systems. Consider more sensitive detection methods like RNAscope for validation.

  • Antibody Clone Limitations: Some clones may recognize only specific conformations or isoforms of LGR5.
    Mitigation: Test multiple antibody clones recognizing different epitopes. Published studies using clone 803420 document successful detection in multiple applications .

  • Sample Processing Issues: Prolonged fixation or improper storage of samples.
    Mitigation: Standardize sample handling protocols. Include positive control samples with known LGR5 expression.

  • Technical Factors: Suboptimal antibody concentration or incubation conditions.
    Mitigation: Perform systematic titration experiments. The recommendation that "optimal dilutions should be determined by each laboratory for each application" highlights the importance of this approach .

By understanding these potential issues and implementing appropriate controls and optimization strategies, researchers can minimize false results and improve the reliability of LGR5/GPR49 detection in their specific experimental systems.

How do I quantitatively analyze LGR5/GPR49 expression data from different experimental techniques?

Quantitative analysis of LGR5/GPR49 expression requires technique-specific approaches to ensure accurate and reproducible data interpretation. Below is a methodological framework for analysis across different experimental platforms:

Flow Cytometry Quantification:

  • Population Gating Strategy:

    • Implement hierarchical gating: first exclude debris/dead cells, then identify single cells, followed by LGR5+ population gating

    • Use isotype controls to set positive/negative thresholds as demonstrated in the HEK293 transfection experiments

    • For co-expression studies, employ fluorescence minus one (FMO) controls

  • Expression Metrics:

    • Report both percentage of positive cells and median fluorescence intensity (MFI)

    • For comparison between experiments, calculate relative MFI (rMFI = sample MFI/isotype control MFI)

    • When comparing across different experimental conditions, normalize to appropriate controls

Immunohistochemistry/Immunofluorescence Quantification:

  • Image Acquisition Parameters:

    • Standardize exposure times and gain settings across all samples

    • Capture multiple representative fields (minimum 5-10 per sample)

    • Include internal control regions within each tissue section

  • Analysis Approaches:

    • Categorical scoring: Establish clear scoring criteria (0=negative, 1=weak, 2=moderate, 3=strong)

    • Digital image analysis: Use software to quantify:

      • Positive cell counting (number or percentage of positive cells)

      • Signal intensity measurement (integrated density values)

      • Subcellular localization patterns (membrane vs. cytoplasmic as observed in transfected cells)

  • Normalization Strategies:

    • Normalize to tissue area or cell number

    • For comparative studies, express as fold change relative to control samples

    • For developmental studies, track changes against defined time points

Western Blot Quantification:

  • Loading Controls:

    • Normalize LGR5 band intensity to appropriate loading controls (β-actin, GAPDH)

    • Consider using total protein normalization methods (Ponceau S, REVERT™ staining)

  • Densitometry Approach:

    • Use linear range exposure times

    • Perform background subtraction consistently

    • Present data as relative expression compared to control samples

Functional Assay Quantification:

  • Activity-Based Metrics:

    • For TOPflash assays measuring LGR5 activity, normalize luciferase readings to control reporter

    • Present data as fold-induction over baseline

    • Include appropriate positive controls (Wnt-3a treatment) and negative controls

  • Statistical Analysis:

    • Apply appropriate statistical tests based on data distribution

    • For multiple comparisons, implement correction methods (Bonferroni, FDR)

    • Report variability measures (standard deviation or standard error)

    • Present individual data points alongside means for small sample sizes

  • Integrated Multi-technique Analysis:

    • Correlate protein expression levels with functional readouts

    • Compare expression patterns across different detection methods

    • Integrate with gene expression data when available

This systematic approach enables robust quantification of LGR5/GPR49 expression, facilitating meaningful comparisons across experimental conditions and detection methods.

How can I differentiate between membrane-bound and internalized LGR5/GPR49 in my experimental data?

Distinguishing between membrane-bound and internalized LGR5/GPR49 is crucial for understanding receptor dynamics, trafficking, and signaling mechanisms. Several methodological approaches can be employed to differentiate these populations:

High-Resolution Microscopy Techniques:

  • Confocal Microscopy with Z-stack Analysis:

    • Acquire optical sections at submicron intervals

    • Perform 3D reconstruction to precisely localize LGR5 signals

    • Co-stain with membrane markers (e.g., Na+/K+-ATPase, WGA) and endosomal/lysosomal markers (e.g., EEA1, LAMP1)

    • Published images showing LGR5 localization "to cell surfaces and cytoplasm" suggest this approach can effectively distinguish populations

  • Super-Resolution Microscopy:

    • Techniques like STORM, PALM, or STED provide resolution below diffraction limit (~250 nm)

    • Enables precise discrimination between membrane-localized and proximal cytoplasmic vesicles

    • Requires specialized fluorophores and equipment

Biochemical Fractionation Approaches:

  • Cell Surface Biotinylation:

    • Biotinylate cell surface proteins in intact cells at 4°C

    • Lyse cells and isolate biotinylated proteins with streptavidin

    • Compare LGR5 levels in biotinylated (membrane) vs. non-biotinylated (internal) fractions by immunoblotting

    • Include controls for biotinylation efficiency and cellular integrity

  • Differential Detergent Extraction:

    • Use mild detergents (e.g., digitonin) to selectively permeabilize plasma membrane

    • Extract and analyze membrane fraction separately from internal pools

    • Validate fractionation with known membrane and intracellular markers

Flow Cytometry-Based Methods:

  • Non-permeabilized vs. Permeabilized Analysis:

    • Stain intact cells to detect only surface-exposed LGR5

    • Permeabilize parallel samples to detect total LGR5

    • Calculate internalized fraction by subtraction

    • This approach can be adapted from protocols used for transfected HEK293 cells

  • Antibody Feeding Assay:

    • Label surface LGR5 with antibodies at 4°C

    • Allow internalization at 37°C for various timepoints

    • Detect remaining surface antibody with one fluorophore

    • Permeabilize and detect internalized antibody with different fluorophore

    • Quantify ratio changes over time

pH-Sensitive Fluorophore Approaches:

  • pH-sensitive Dye Conjugation:

    • Conjugate LGR5 antibody to pH-sensitive fluorophores (e.g., pHrodo)

    • These probes increase fluorescence intensity in acidic environments (endosomes/lysosomes)

    • Monitor fluorescence changes to track internalization kinetics

    • Combine with live-cell imaging for temporal analysis

Quantification and Analysis Strategies:

  • Co-localization Analysis:

    • Calculate Pearson's or Mander's coefficients for LGR5 with membrane vs. endocytic markers

    • Present data as percent co-localization or coefficients

    • Include positive controls (known membrane proteins) and negative controls

  • Internalization Kinetics:

    • Measure ratio of membrane:internal LGR5 over time following stimulation

    • Calculate internalization rates and recycling kinetics

    • Compare between experimental conditions or genetic manipulations

These methodologies provide complementary approaches to distinguish and quantify membrane-bound versus internalized LGR5/GPR49, enabling detailed investigation of receptor trafficking and its relationship to Wnt signaling dynamics.

How can LGR5/GPR49 antibodies be used to study cancer stem cell dynamics?

LGR5/GPR49 antibodies serve as powerful tools for investigating cancer stem cell (CSC) dynamics due to LGR5's established role as a marker for both normal and cancer stem cells. Several methodological approaches leverage these antibodies for CSC research:

  • Lineage Tracing in Cancer Models:

    • LGR5 antibodies can identify potential CSCs in primary tumors and patient-derived xenografts

    • Sequential sampling and staining enables tracking of LGR5+ cell fate during tumor progression

    • This approach builds on established protocols for detecting LGR5 in tissue sections

    • Quantitative analysis can determine whether LGR5+ cells expand, contract, or maintain stable populations during tumor evolution

  • Prospective Isolation and Functional Characterization:

    • Flow cytometry-based protocols using LGR5 antibodies allow isolation of putative CSCs

    • Sorted LGR5+ and LGR5- populations can be compared for:

      • Tumorigenic potential in limiting dilution assays

      • Sphere-forming efficiency in vitro

      • Drug resistance profiles

      • Differentiation capacity

    • The validated flow cytometry protocols with LGR5 antibodies provide a foundation for these applications

  • Dual Marker Strategies:

    • Co-staining with LGR5 antibodies and other CSC markers (CD133, CD44, ALDH)

    • This approach identifies distinct CSC subpopulations with potentially different functional properties

    • Enabled by compatible fluorescent secondary antibodies as demonstrated in immunofluorescence protocols

    • Quantitative co-expression analysis determines marker overlap and heterogeneity

  • Monitoring Therapy Response:

    • LGR5 antibody staining before and after treatment to assess CSC dynamics

    • This approach can reveal whether treatments:

      • Selectively eliminate or spare LGR5+ cells

      • Induce phenotypic changes in LGR5 expression

      • Affect the spatial distribution of LGR5+ cells within tumors

    • Time-course analysis quantifies kinetics of CSC elimination or enrichment

  • Spatial Context Analysis:

    • Multiplex immunohistochemistry combining LGR5 antibodies with:

      • Other cell type markers (immune cells, cancer-associated fibroblasts)

      • Extracellular matrix components

      • Hypoxia markers

    • This reveals microenvironmental niches supporting CSC maintenance

    • Digital image analysis quantifies spatial relationships between LGR5+ cells and other cellular/molecular features

  • Therapeutic Targeting Strategies:

    • LGR5 antibodies can be developed into therapeutic agents targeting CSCs

    • Functional antibodies may induce:

      • Direct cytotoxicity

      • Immune effector recruitment

      • Internalization of conjugated toxins

    • The documented activity of LGR5 antibodies in functional assays suggests therapeutic potential

These methodological approaches collectively provide a comprehensive toolkit for investigating cancer stem cell dynamics, potentially leading to improved therapeutic strategies targeting this critical cell population in various malignancies.

What are the latest developments in using biophysics-informed modeling for designing highly specific LGR5/GPR49 antibodies?

Recent advances in biophysics-informed modeling have significantly enhanced the design of highly specific LGR5/GPR49 antibodies. These cutting-edge approaches integrate experimental data with computational methods to generate antibodies with precisely engineered specificity profiles:

  • Binding Mode Identification and Disentanglement:

    • Novel computational frameworks now identify distinct binding modes associated with specific ligands

    • This approach enables discrimination between very similar epitopes that cannot be experimentally dissociated

    • The methodology successfully disentangles binding modes even for chemically similar ligands

    • For LGR5 research, this facilitates designing antibodies that can distinguish between closely related family members (LGR4, LGR5, LGR6)

  • Integration of Phage Display with Computational Modeling:

    • High-throughput sequencing data from phage display experiments train biophysics-informed models

    • The models identify sequence-function relationships beyond what is directly observed experimentally

    • This approach has demonstrated success in "the computational design of antibodies with customized specificity profiles"

    • The methodology can generate antibodies with either highly specific binding to LGR5 or designed cross-specificity to multiple predetermined targets

  • Structural Prediction Advancements:

    • Combined approaches using homology modeling with knowledge-based and energy-based methods generate increasingly accurate antibody structural predictions

    • For non-H3 loops, canonical structure-based approaches now achieve backbone RMSD values of approximately 0.7 Å

    • These precise structural models serve as platforms for rational engineering of binding interfaces

  • Enhanced Antibody-Antigen Complex Prediction:

    • Tools like SnugDock implement alternating rounds of low-resolution rigid body perturbations and high-resolution side-chain and backbone minimization

    • These methods overcome the traditional challenges in antibody-antigen docking where "shape complementarity between antibody and antigen is not a good determinant of correct antibody placement"

    • The approaches generate large numbers of potential complexes (~10^4-10^5) that can be evaluated for specificity determinants

  • Stability and Aggregation Prediction Integration:

    • Computational prediction of aggregate-prone regions (APRs) based on sequence composition and structural properties

    • Design strategies incorporate mutations in these regions to enhance stability while maintaining specificity

    • This enables development of LGR5 antibodies with improved biophysical properties suitable for diverse research applications

  • Application to Epitope-Specific Targeting:

    • Recent methodologies enable "designing antibodies with both specific and cross-specific binding properties"

    • This is particularly valuable for targeting specific functional domains of LGR5 involved in distinct signaling pathways

    • The approach facilitates creation of antibodies that selectively modulate certain LGR5 functions while preserving others

These developments represent significant advances beyond traditional antibody generation approaches, offering unprecedented control over specificity profiles and functional properties of LGR5/GPR49 antibodies for research and potential therapeutic applications.

How can LGR5/GPR49 antibodies be modified for therapeutic applications while maintaining specificity?

Modifying LGR5/GPR49 antibodies for therapeutic applications while preserving their specificity requires sophisticated engineering strategies that address multiple aspects of antibody structure and function:

  • Fc Engineering for Optimized Effector Functions:

    • Selective modification of the hinge region, CH2 domain, N-glycans, and N-glycan-attached residues can modulate therapeutic properties

    • Mutations in the upper and middle hinge regions demonstrably affect FcγRIIIa and C1q binding, crucial for antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC)

    • Computational optimization using Protein Design Automation (PDA) technology and Sequence Prediction Algorithm (SPA) enables precise tuning of Fc-mediated functions

    • These modifications can be implemented without altering the antigen-binding region, thereby preserving LGR5 specificity

  • Stability Enhancement for In Vivo Applications:

    • Computational prediction methods identify aggregate-prone regions (APRs) in antibody sequences

    • Strategic mutations in these regions improve resistance to aggregation—critical for high-concentration formulations required in therapeutic settings

    • The sequence composition and structural properties (hydrophobicity, charge, secondary structure propensity) guide targeted modifications

    • These stability enhancements can be introduced while maintaining the complementarity-determining regions (CDRs) responsible for LGR5 specificity

  • Format Diversification for Optimal Tissue Penetration:

    • Full-length antibodies can be engineered into various formats:

      • Fab fragments for improved tissue penetration

      • scFv constructs for applications requiring smaller size

      • Bispecific formats targeting LGR5 and another relevant antigen

    • Molecular modeling techniques can predict how these structural modifications affect stability and binding properties

    • Format selection should consider the specific therapeutic context (solid tumors vs. circulating cells) and desired pharmacokinetics

  • Surface Charge Modification for Pharmacokinetics Control:

    • Altering surface-exposed residues affects isoelectric point and charge distribution

    • These modifications influence:

      • Serum half-life

      • Tissue distribution

      • Non-specific binding

    • Computational approaches can predict how sequence changes affect surface properties while preserving the LGR5-binding interface

  • Conjugation Chemistry for Antibody-Drug Conjugates (ADCs):

    • Site-specific conjugation technologies target defined residues distant from the antigen-binding site

    • Engineering cysteine residues or incorporating non-natural amino acids at strategic positions enables precise conjugation

    • This approach maintains LGR5 binding specificity while allowing attachment of cytotoxic payloads

    • The documented ability to detect and bind LGR5 in various experimental conditions suggests suitability for ADC development

  • Humanization and Deimmunization:

    • For mouse-derived LGR5 antibodies, humanization is essential to reduce immunogenicity

    • Computational design can identify minimal mutations required for humanization while preserving binding characteristics

    • Deimmunization algorithms predict and eliminate T-cell epitopes that might trigger immune responses

    • The high sequence identity between mouse and human LGR5 (90%) suggests epitope conservation, potentially facilitating humanization while maintaining specificity

These methodological approaches provide a comprehensive framework for developing LGR5/GPR49-targeted therapeutics that combine high specificity with optimized pharmacological properties for clinical applications.

How do different animal models affect LGR5/GPR49 antibody performance and interpretation?

The performance and interpretation of LGR5/GPR49 antibody data vary significantly across animal models due to species-specific differences in protein sequence, expression patterns, and tissue architecture. Below is a comparative analysis of key considerations for major model systems:

Animal ModelSequence Homology to Human LGR5Key Considerations for Antibody PerformanceMethodological Adaptations
Mouse90% amino acid identity - Most widely validated model
- Extensive literature on expression patterns
- Commercial antibodies predominantly validated in mouse tissues
- Standard protocols often directly applicable
- Published applications include flow cytometry, immunofluorescence, and IHC
Rat95% identity to mouse LGR5 - High conservation suggests most mouse antibodies will cross-react
- Expression patterns similar to mouse in most organs
- Less extensively characterized than mouse
- Mouse antibodies typically work with minimal optimization
- May require slight adjustment of antibody concentrations
- Validate cross-reactivity experimentally
Zebrafish~70% identity (variable by domain)- More divergent epitopes
- Different spatiotemporal expression patterns
- Advantages for developmental studies
- Likely requires zebrafish-specific antibodies
- Higher antibody concentrations often needed
- Whole-mount protocols may require extensive optimization
Non-human primates>95% identity to human- High translational relevance
- Similar tissue architecture to humans
- Valuable for safety studies
- Human-targeted antibodies typically effective
- Important to validate with appropriate controls
- Useful for preclinical validation of therapeutic approaches
Canine/Feline~85-88% identity- Relevant for veterinary applications
- Spontaneous tumors provide natural disease models
- Limited validation in literature
- Test multiple antibody clones for cross-reactivity
- Optimize antigen retrieval protocols specifically
- Include definitive positive/negative control tissues

Critical Analysis Considerations Across Models:

This comprehensive comparison enables researchers to select appropriate animal models and implement model-specific methodological adaptations to ensure reliable LGR5/GPR49 antibody performance across diverse experimental systems.

How can LGR5/GPR49 antibodies be integrated with other detection methods for comprehensive stem cell analysis?

Integrating LGR5/GPR49 antibody detection with complementary methodologies creates a powerful multi-modal approach for comprehensive stem cell characterization. This integrated strategy provides deeper insights into stem cell properties, localization, and function than any single method alone:

  • Integration with Transcriptomic Analysis:

    • Combine LGR5 antibody-based cell sorting with RNA-sequencing:

      • Isolate LGR5+ populations using validated flow cytometry protocols

      • Perform single-cell or bulk RNA-seq on sorted populations

      • Compare gene expression profiles between LGR5+ and LGR5- cells

    • Spatial transcriptomics integration:

      • Perform LGR5 immunohistochemistry on serial sections

      • Correlate protein expression with spatial gene expression data

      • Identify co-expression networks specific to LGR5+ stem cell niches

  • Combination with Functional Assays:

    • Link LGR5 expression with functional stemness assays:

      • Organoid formation efficiency correlations

      • Lineage tracing in transgenic models

      • Clone-forming capacity in vitro

    • Real-time activity measurement:

      • LGR5 antibody detection combined with TOPflash assays to measure Wnt pathway activity

      • Live-cell calcium imaging to assess signal transduction

      • Proliferation kinetics in LGR5+ versus LGR5- populations

  • Multi-parameter Flow Cytometry:

    • Design panels incorporating:

      • LGR5 antibody detection

      • Additional stem cell markers (e.g., CD133, CD44)

      • Functional markers (e.g., ALDH activity, side population dyes)

      • Cell cycle indicators (e.g., Ki67, DNA content dyes)

    • Computational analysis using tools like SPADE or viSNE to identify distinct stem cell subpopulations

    • This builds upon established flow cytometry protocols for LGR5 detection

  • Advanced Microscopy Integration:

    • Multiplex immunofluorescence combining:

      • LGR5 antibody staining

      • Additional marker antibodies

      • Functional probes (e.g., proliferation, apoptosis)

    • Intravital microscopy in models with fluorescent reporters:

      • Complement with ex vivo LGR5 antibody validation

      • Track stem cell dynamics in real-time

      • Correlate with fixed tissue analysis

    • Super-resolution approaches:

      • Examine LGR5 nanoscale distribution

      • Study co-localization with signaling partners at molecular scale

  • Epigenetic Analysis Correlation:

    • Antibody-based cell isolation followed by:

      • ATAC-seq for chromatin accessibility

      • ChIP-seq for histone modifications

      • DNA methylation profiling

    • Integration with LGR5 expression data to identify epigenetic mechanisms regulating stemness

  • Multi-modal In Vivo Imaging:

    • Development of LGR5-targeting probes for:

      • PET imaging using radiolabeled antibody fragments

      • Optical imaging with fluorescently-labeled antibodies

      • MRI with antibody-conjugated nanoparticles

    • Validation against ex vivo antibody staining

    • Longitudinal tracking of stem cell populations during development or disease progression

  • Computational Integration Framework:

    • Machine learning approaches to integrate:

      • Antibody-based protein expression data

      • Transcriptomic profiles

      • Functional readouts

      • Spatial information

    • Predictive modeling of stem cell behavior based on multi-modal data

    • Network analysis to identify key regulatory nodes in stem cell maintenance

This comprehensive integration strategy leverages the specificity of LGR5/GPR49 antibodies while compensating for their limitations through complementary technologies, resulting in robust stem cell characterization across multiple dimensions.

What are the considerations for developing and validating LGR5/GPR49 antibodies for emerging research areas beyond cancer and stem cell biology?

Developing and validating LGR5/GPR49 antibodies for emerging research areas requires specialized considerations that address unique challenges in diverse biological contexts. These considerations span from antibody design to application-specific validation:

  • Developmental Biology Applications:

    • Temporal Expression Dynamics:

      • Validate antibodies across multiple developmental stages

      • Ensure detection of early expression when protein levels may be low

      • Correlate with established developmental markers

    • Cross-Species Validation:

      • Test conservation of epitopes in evolutionary developmental models

      • Consider embryo-specific optimization for clearing and penetration

      • Develop specialized whole-mount protocols with extended incubation times

    • Integration with Lineage Tracing:

      • Validate antibody compatibility with genetic lineage tracing tools

      • Establish protocols for sequential sacrifices to track developmental trajectories

      • Document successful applications in developmental contexts similar to studies on dermal adipocyte lineage cells

  • Regenerative Medicine Applications:

    • Compatibility with Scaffold Materials:

      • Test for non-specific binding to common biomaterials

      • Develop specialized extraction protocols for protein isolation from scaffolds

      • Optimize antigen retrieval for fixed engineered tissues

    • Detection in Mixed Human-Animal Chimeras:

      • Validate species-specific antibodies for distinguishing host vs. graft cells

      • Establish protocols for simultaneous detection of human and animal LGR5

      • Implement controls to confirm specificity in chimeric contexts

    • Functional Validation in Therapeutic Cells:

      • Correlate LGR5 expression with regenerative capacity

      • Establish quality control standards for therapeutic applications

      • Develop quantitative thresholds for clinical applications

  • Neuroscience Applications:

    • Blood-Brain Barrier Considerations:

      • Optimize antibody format (e.g., sdAb, Fab) for CNS penetration

      • Validate detection in neural stem cell niches

      • Develop specialized delivery methods for in vivo imaging

    • Compatibility with Neural Tissue Processing:

      • Test with brain-specific fixation protocols (e.g., PFA/glutaraldehyde mixtures)

      • Optimize clearing protocols compatible with antibody epitopes

      • Establish antigen retrieval methods suitable for heavily myelinated regions

    • Multiplex with Neural Markers:

      • Validate co-staining with key neural stem cell and progenitor markers

      • Develop protocols distinguishing LGR5+ neural cells from adjacent populations

      • Optimize signal detection in high-autofluorescence neural tissues

  • Aging Research Applications:

    • Detection in Aged Tissues:

      • Validate antibodies in aged tissues with altered post-translational modifications

      • Develop protocols addressing increased autofluorescence in aged tissues

      • Establish age-specific positive controls and quantification standards

    • Senescence Marker Integration:

      • Validate compatibility with senescence detection methods (SA-β-gal, p16)

      • Optimize multiplex protocols for distinguishing senescent LGR5+ cells

      • Develop quantitative approaches for age-related changes in LGR5 expression

  • Infectious Disease Research:

    • Compatibility with Pathogen Detection:

      • Validate antibody performance in infected tissues

      • Test for cross-reactivity with pathogen proteins

      • Develop specialized fixation protocols preserving both LGR5 and pathogen antigens

    • Application in Organoid Infection Models:

      • Establish protocols for organoids derived from LGR5+ cells

      • Validate detection in 3D cultures following infection

      • Optimize extraction from Matrigel/ECM without epitope damage

  • Validation Framework for Novel Applications:

    • Biological Controls:

      • Include tissue-specific positive controls with known LGR5 expression

      • Implement genetic knockdown/knockout validation where possible

      • Use competitive binding with recombinant protein to confirm specificity

    • Technical Controls:

      • Include isotype controls matched to the specific application

      • Perform peptide blocking studies

      • Compare multiple antibody clones recognizing different epitopes

    • Method-Specific Validation:

      • Establish application-specific protocols (as demonstrated for flow cytometry, ICC, and IHC)

      • Document performance characteristics for each application

      • Develop quantitative standards appropriate for the research context

This comprehensive framework enables researchers to confidently extend LGR5/GPR49 antibody applications beyond established fields, ensuring reliable results in diverse biological contexts while addressing the unique challenges of emerging research areas.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.