The LPAR2 antibody is a polyclonal or monoclonal immunoglobulin raised against specific epitopes of the LPAR2 protein, which is a 39 kDa transmembrane receptor expressed in macrophages, fibroblasts, and cardiac myocytes . Biotin conjugation involves chemically attaching biotin molecules to the antibody’s amino groups, enabling high-affinity binding to streptavidin or avidin-based detection systems . This conjugation enhances sensitivity in immunoassays by amplifying signal detection .
The LPAR2 Antibody, Biotin conjugated is utilized in multiple research and diagnostic workflows:
Western Blotting: Detects LPAR2 expression in lysates (e.g., human peripheral blood lymphocytes) . Combined with HRP-conjugated streptavidin, it enables chemiluminescent detection .
Immunohistochemistry (IHC): Localizes LPAR2 in tissue sections, as demonstrated in studies of gastric cancer and intestinal barrier integrity .
Flow Cytometry: Analyzes LPAR2 expression on immune cells, such as T cells in lymph nodes .
ELISA: Quantifies LPAR2 levels in biological fluids using biotin-streptavidin complexes .
Gastric Cancer: Overexpression of LPAR2 correlates with tumor progression and metastasis. LPA signaling via LPAR2 promotes β-catenin activation, enhancing cell proliferation .
Radiation Injury: LPAR2 activation protects colonic epithelial tight junctions during γ-irradiation, mitigating barrier dysfunction and endotoxemia .
Amyotrophic Lateral Sclerosis (ALS): LPAR2 signaling has a dual role, exacerbating early microglial activation but providing neuroprotection in later stages .
Signaling Pathways: LPAR2 activates Rho-ROCK-LIM kinase pathways, which regulate cytoskeletal dynamics and cell motility .
Therapeutic Targets: Small-molecule agonists (e.g., RP1) and antagonists (e.g., Ki16425) modulate LPAR2 activity in preclinical models .
Cross-Reactivity: Polyclonal antibodies may exhibit higher non-specific binding compared to monoclonal counterparts .
Optimization: Dilution factors (1:500–1:2000) must be validated for specific assays .
Storage: Requires -20°C storage and avoidance of freeze-thaw cycles .
LPAR2 (also known as EDG4 or LPA-2) is a G protein-coupled receptor for lysophosphatidic acid (LPA), a mediator of diverse cellular activities. This receptor is primarily located in the cell membrane and plays key roles in multiple signaling pathways. LPAR2 appears to couple to multiple G protein families including G(i)/G(o), G(12)/G(13), and G(q), allowing it to participate in various downstream cellular processes .
It plays a critical role in the phospholipase C-beta (PLC-beta) signaling pathway and stimulates phospholipase C activity in a manner that is independent of RALA activation . In the context of immunity, LPAR2 has been shown to promote T cell motility, contrasting with LPAR6 which primarily suppresses T cell migration .
Commercially available LPAR2 polyclonal antibodies with biotin conjugation typically have the following specifications:
Parameter | Specification |
---|---|
Host | Rabbit |
Clonality | Polyclonal |
Conjugation | Biotin |
Source | KLH conjugated synthetic peptide derived from human LPAR2 |
Immunogen Range | 1-100/351 |
Isotype | IgG |
Concentration | 1ug/ul |
Purification Method | Protein A |
Storage Buffer | Aqueous buffered solution containing 0.01M TBS (pH 7.4) with 1% BSA, 0.03% Proclin300 and 50% Glycerol |
Storage Condition | -20°C for up to 12 months |
Applications | ELISA (1:500-1000), IHC-P (1:200-400), IHC-F (1:100-500) |
Reactivity | Mouse, Rat |
Predicted Reactivity | Human, Dog, Cow, Sheep, Pig, Horse, Rabbit |
These specifications ensure consistent performance in various experimental applications and appropriate cross-reactivity with relevant species .
LPAR2 exhibits distinct functional characteristics compared to other LPA receptors:
While LPAR1 is primarily associated with tumor cell migration and metastasis, LPAR2 has been specifically linked to T cell motility .
Unlike LPAR6, which preferentially couples to the Gα12/13-RhoA pathway that drives cytoskeletal contraction and suppression of cell motility, LPAR2 couples to Gi-mediated Rac GTPase activation and promotes the random motility of T cells .
The signaling pathway distinction is critical: LPAR2 activation generally enhances cell movement, while LPAR6 restricts it through different G-protein coupling mechanisms .
In cancer biology, LPAR2 expression correlates with different prognoses across cancer types - associated with worse outcomes in some cancers (ACC, KIRC, MESO) but improved prognosis in others (HNSC, OV, STAD) .
This functional diversity highlights the importance of receptor-specific research approaches when investigating LPA signaling pathways.
For optimal immunohistochemistry results with biotin-conjugated LPAR2 antibodies:
For Paraffin-Embedded Sections (IHC-P):
Antigen retrieval: Heat-induced epitope retrieval using citrate buffer (pH 6.0)
Blocking: 5-10% normal serum from the same species as the secondary antibody
Incubation time: Overnight at 4°C for primary antibody
Detection: Streptavidin-HRP system followed by DAB or similar chromogen
Counterstain: Hematoxylin for nuclear visualization
For Frozen Sections (IHC-F):
Fixation: Acetone or 4% paraformaldehyde
Blocking: 5-10% normal serum with 0.1-0.3% Triton X-100
Incubation time: 1-2 hours at room temperature
Detection: Same as IHC-P
Controls: Include negative controls (omitting primary antibody) and positive controls (tissues known to express LPAR2)
Optimization may be necessary for each specific tissue type, and validation with alternative detection methods is advised for conclusive results.
Based on recent research findings, experiments investigating LPAR2's role in immune cell function should consider:
Cell Migration Assays:
Transwell migration assays comparing LPAR2-expressing vs. LPAR2-knockout or silenced immune cells
Live cell imaging to track T cell motility patterns following LPA stimulation
Chemokinesis vs. chemotaxis assessment using gradient-free and gradient conditions
Signaling Pathway Analysis:
Immune Microenvironment Context:
Specificity Controls:
This multi-faceted approach enables researchers to delineate the specific contributions of LPAR2 to immune function while controlling for potential confounding effects of other LPA receptors.
Validating antibody specificity is crucial for reliable research outcomes. For LPAR2 antibodies, the following validation methods are recommended:
Genetic Approaches:
CRISPR/Cas9 knockout of LPAR2 in relevant cell lines
siRNA or shRNA knockdown of LPAR2 followed by immunostaining
Overexpression of tagged LPAR2 (e.g., FLAG-tag) and co-localization analysis
Peptide Competition Assays:
Multiple Antibody Validation:
Use of alternative antibodies targeting different epitopes of LPAR2
Comparison of staining patterns across various detection methods
Cross-Reactivity Assessment:
Western Blot Analysis:
Verification of band size corresponding to LPAR2 (approximately 39 kDa)
Analysis of cells with differential LPAR2 expression
Orthogonal Validation:
Correlation of protein detection with mRNA expression (RT-PCR or RNA-seq)
Mass spectrometry confirmation of the immunoprecipitated protein
These methods collectively provide robust validation of antibody specificity, which is essential before employing these reagents in critical research applications.
LPAR2 signaling appears to have complex, context-dependent effects on the tumor microenvironment and immune infiltration:
Correlation with Immune Cell Infiltration:
Recent studies show that LPAR2 expression significantly correlates with the presence of various immune markers in immune cell subsets of head and neck squamous cell carcinoma (HNSC) and kidney renal clear cell carcinoma (KIRC) . This suggests LPAR2 may influence the composition of tumor-infiltrating immune cells.
Dual Role in Cancer Prognosis:
High LPAR2 expression correlates with improved survival in certain cancers (BLCA, CESC, HNSC, TYHM, UCEC) but worse prognosis in others (ACC, KIRC, MESO) . This suggests tissue-specific functions of LPAR2 in modulating anti-tumor immunity.
Interaction with ATX/LPA Axis:
While LPAR2 promotes T cell motility, the ATX-LPA axis can have chemorepulsive effects on tumor-infiltrating lymphocytes. This creates a complex signaling environment where the balance between different LPA receptors determines immune cell infiltration patterns .
Clinical Significance:
Survival analyses reveal that LPAR2 expression significantly impacts patient outcomes:
These findings highlight the potential of LPAR2 as both a prognostic biomarker and therapeutic target, particularly in cancers where it influences immune infiltration.
LPAR2 demonstrates specific G-protein coupling preferences that influence cell invasion mechanisms:
G-Protein Coupling Specificity:
LPAR2 couples to multiple G protein families, including G(i)/G(o), G(12)/G(13), and G(q), allowing it to participate in various downstream cellular processes . This multi-pathway activation enables diverse cellular responses.
Role in Entotic Invasion:
LPAR2 has been identified as a signal transducer specifically required for actively invading cells during entosis (homotypic cell-in-cell invasion). This process depends on G12/13 signaling and PDZ-RhoGEF activation .
Contrasting Functions with LPAR6:
While LPAR2 couples to Gi-mediated pathways to promote cell motility, LPAR6 preferentially couples to the Gα12/13-RhoA pathway, driving cytoskeletal contraction and suppression of cell motility . This differential G-protein coupling explains their opposing effects on cell movement.
Downstream Effector Activation:
LPAR2 plays a key role in phospholipase C-beta (PLC-beta) signaling and stimulates PLC activity in a manner independent of RALA activation . This pathway is critical for calcium signaling and protein kinase C activation.
Actin Cytoskeletal Regulation:
G12/13 signaling downstream of LPAR2 influences blebbing and actin dynamics, which are essential for the mechanical process of cell invasion .
Understanding these specific G-protein coupling preferences and downstream signaling cascades is crucial for developing targeted interventions that modulate LPAR2-mediated cellular processes in disease contexts.
Researchers seeking to integrate LPAR2 expression data with clinical outcomes should consider these methodological approaches:
This integrated approach provides robust evidence for the clinical relevance of LPAR2 in specific cancer contexts and may identify patient subgroups most likely to benefit from therapies targeting the LPAR2 signaling axis.
Researchers working with biotin-conjugated LPAR2 antibodies should be aware of these common pitfalls:
Endogenous Biotin Interference:
Tissues may contain endogenous biotin that can cause non-specific binding
Solution: Use biotin blocking steps (streptavidin followed by biotin) before applying the biotin-conjugated primary antibody
Alternative: Consider using avidin-biotin blocking kits specifically designed for this purpose
Storage-Related Degradation:
Cross-Reactivity Challenges:
Detection System Sensitivity Issues:
Background in Highly Biotin-Rich Tissues:
Tissues like liver, kidney, and brain contain high endogenous biotin
Solution: Use alternative non-biotin detection methods for these tissues
Alternative: Increase washing steps and blocking concentrations
Competitive Binding with Lysophosphatidic Acid:
Native LPA may compete with antibody binding in certain sample preparations
Solution: Consider fixation methods that preserve epitope accessibility while removing endogenous ligands
Addressing these challenges proactively will improve experimental outcomes and data reliability when working with biotin-conjugated LPAR2 antibodies.
When faced with contradictory results between LPAR2 expression and functional outcomes, researchers should consider:
Context-Dependent Signaling:
LPAR2 couples to multiple G-protein families (G(i)/G(o), G(12)/G(13), G(q)) , enabling different functional outcomes depending on the cellular context
Examine which G-protein coupling predominates in your specific experimental system
Consider the balance between different LPA receptors, as LPAR2 and LPAR6 can have opposing effects
Cancer-Type Specificity:
Methodological Considerations:
Antibody detection may not reflect functional receptor status
Evaluate both protein expression (IHC, Western blot) and functional assays (signaling, migration)
Consider receptor internalization, which may reduce membrane expression despite active signaling
Competing Pathways:
Analysis Framework:
Use multivariate analyses that control for confounding factors
Consider non-linear relationships between expression and function
Employ multiple statistical approaches (parametric and non-parametric)
Validation Strategies:
Use genetic approaches (knockout, knockdown) to confirm antibody-based findings
Apply pharmacological tools (specific agonists/antagonists) to distinguish receptor-specific effects
Consider the temporal dynamics of receptor expression and downstream signaling
Through systematic consideration of these factors, researchers can better interpret apparently contradictory results and develop a more nuanced understanding of LPAR2 biology.
Comprehensive control strategies for evaluating LPAR2 expression in tumor samples should include:
Technical Controls:
Positive Controls: Known LPAR2-expressing tissues/cell lines (preferably matched to tumor type)
Negative Controls: Primary antibody omission on serial sections
Isotype Controls: Rabbit IgG at the same concentration as the primary antibody
Absorption Controls: Primary antibody pre-incubated with immunizing peptide
Biological Reference Controls:
Adjacent Normal Tissue: For comparison with tumor expression
Tissue Microarrays: Multiple tumor and normal samples processed identically
Gradient of Expression: Include samples with known high, medium, and low LPAR2 expression
Validation Controls:
Orthogonal Methods: Validate IHC findings with Western blot or RT-PCR
Multiple Antibodies: Use antibodies targeting different epitopes of LPAR2
In Situ Hybridization: Confirm protein expression with mRNA detection
Contextual Controls:
Clinical Correlation Controls:
Quantification Controls:
Inter-observer Validation: Multiple pathologists scoring the same samples
Digital Pathology Standards: Calibration controls for automated scoring
Batch Controls: Reference samples included in each staining batch
Several cutting-edge methodologies show promise for advancing LPAR2 research:
Spatial Transcriptomics and Proteomics:
Techniques like 10x Genomics Visium or GeoMx DSP allow simultaneous visualization of LPAR2 expression and spatial context
These approaches can reveal microenvironmental relationships between LPAR2-expressing cells and immune infiltrates
Integration with multiplexed immunofluorescence can map receptor distribution relative to signaling nodes
Single-Cell Multi-Omics:
Single-cell RNA sequencing combined with protein detection (CITE-seq) can correlate LPAR2 transcript levels with surface protein expression
This can identify cellular subpopulations with unique LPAR2 signaling properties
Trajectory analysis can reveal how LPAR2 expression changes during cellular differentiation or tumor progression
Live Cell Biosensors:
FRET-based biosensors for detecting LPAR2 activation and G-protein coupling in real-time
Optogenetic approaches to spatiotemporally control LPAR2 signaling
Luminescent biosensors to track downstream second messenger production
Advanced Microscopy Techniques:
Super-resolution microscopy (STORM, PALM) to visualize LPAR2 nanoclustering at the membrane
Lattice light-sheet microscopy for long-term 3D imaging of LPAR2-mediated cell dynamics
Correlative light and electron microscopy to connect receptor distribution with ultrastructural features
Engineered Antibody Formats:
Nanobodies or single-domain antibodies for improved tissue penetration
Bispecific antibodies targeting LPAR2 and downstream effectors
Photoswitchable antibodies for selective visualization in specific regions
Computational Approaches:
Machine learning algorithms for pattern recognition in LPAR2 distribution
Systems biology modeling of LPAR2 signaling networks
Digital pathology tools for automated quantification across large tissue cohorts
These innovative approaches will provide deeper insights into LPAR2 biology and potentially reveal new therapeutic opportunities in cancer and immune modulation.
The relationship between LPAR2 signaling and cancer immunotherapy presents several intriguing research directions:
Modulation of T Cell Infiltration:
Since LPAR2 promotes T cell motility , targeting this receptor could potentially enhance T cell trafficking into tumors
Conversely, the ATX-LPA axis can have chemorepulsive effects on tumor-infiltrating lymphocytes
Combination approaches targeting both LPAR2 and LPAR6 might optimize T cell infiltration patterns
Cancer Type-Specific Approaches:
Combination with Immune Checkpoint Inhibitors:
LPAR2 modulation could potentially sensitize tumors to anti-PD-1/PD-L1 therapy
Research should investigate whether LPAR2 expression correlates with checkpoint inhibitor response
Sequential approaches might optimize immune infiltration before checkpoint blockade
Targeting the ATX-LPA-LPAR2 Axis:
Autotaxin inhibitors could reduce LPA production, affecting both tumor cells and immune cells
Selective LPAR2 modulators might enhance beneficial immune functions while limiting pro-tumor effects
Antibody-drug conjugates targeting LPAR2 could deliver immunomodulatory compounds directly to the tumor microenvironment
Biomarker Development:
LPAR2 expression patterns could serve as predictive biomarkers for immunotherapy response
The ratio of different LPA receptors might provide more comprehensive predictive power
Monitoring changes in LPAR2 expression during treatment could indicate developing resistance mechanisms
Cell-Based Therapeutic Approaches:
Engineered T cells with modified LPAR2 signaling might demonstrate enhanced tumor infiltration
Dendritic cell vaccines targeting LPAR2-expressing tumors could generate more specific immune responses
Ex vivo modification of tumor-infiltrating lymphocytes to optimize LPAR2-mediated motility
These approaches highlight the potential of LPAR2-targeted strategies to enhance existing immunotherapies and develop novel treatment paradigms in cancer.
Beyond cancer, LPAR2 presents exciting research opportunities in several disease areas:
Inflammatory and Autoimmune Disorders:
Given LPAR2's role in T cell motility , investigating its contribution to inflammatory cell recruitment in autoimmune diseases
Examining the balance between pro-inflammatory and regulatory T cell migration mediated by LPAR2
Developing targeted therapies that modulate LPAR2 signaling to restore immune homeostasis
Cardiovascular Disease:
Exploring LPAR2's role in atherosclerosis development and progression
Investigating potential connections between LPAR2 signaling and cardiac remodeling after injury
Examining relationships between LPAR2 and lipoprotein metabolism, particularly given the existence of antibodies against both LPAR2 and lipoprotein a
Fibrotic Disorders:
Investigating LPAR2's contribution to fibroblast activation and extracellular matrix deposition
Examining LPAR2-mediated signaling in organ-specific fibrosis (liver, lung, kidney)
Developing therapeutic strategies targeting the ATX-LPA-LPAR2 axis to mitigate fibrotic processes
Neurological Disorders:
Analyzing LPAR2 expression patterns in neurodegenerative diseases
Exploring the receptor's role in neuroinflammation and blood-brain barrier integrity
Investigating potential neuroprotective or neurotoxic effects of LPAR2 signaling
Metabolic Diseases:
Examining LPAR2's role in adipose tissue inflammation and insulin resistance
Investigating connections between LPAR2 signaling and hepatic metabolism
Exploring potential links to obesity-related immune dysfunction
Reproductive Biology:
Analyzing LPAR2's contribution to embryo implantation and placental development
Investigating its role in reproductive tract immunity
Exploring connections to fertility disorders and pregnancy complications
Tissue Repair and Regeneration:
Examining how LPAR2-mediated cell migration contributes to wound healing
Investigating the receptor's role in stem cell mobilization and differentiation
Developing LPAR2-targeted approaches to enhance tissue regeneration
Infectious Disease Responses:
Analyzing how LPAR2 signaling affects immune responses to pathogens
Investigating whether infectious agents exploit or modulate LPAR2 signaling
Exploring the receptor as a potential target for immunomodulatory anti-infective strategies
These diverse research directions highlight LPAR2's significance beyond cancer and suggest potential therapeutic applications across multiple disease areas.