LPAR2 Antibody, HRP conjugated

Shipped with Ice Packs
In Stock

Description

Key Functional Roles of LPAR2

  • Endothelial Barrier Protection: LPAR2 activation stabilizes vascular endothelial permeability, mitigating conditions like deep vein thrombosis (DVT) and radiation-induced colonic mucosal damage .

  • Radiation Mitigation: LPAR2 agonists reduce γ-irradiation-induced tight junction disruption and endotoxemia by modulating Rho-ROCK signaling pathways .

  • NSAID Enteropathy Modulation: LPAR2 deficiency exacerbates indomethacin-induced intestinal apoptosis, while LPAR2 agonists attenuate mucosal injury .

Experimental Use of HRP-Conjugated Antibody

  • ELISA Quantification: Used to measure LPAR2 expression in human leukocytes and testicular tissues, where the receptor is most abundant .

  • Mechanistic Studies: Facilitated the identification of LPAR2 upregulation in endothelial cells under ischemic or inflammatory conditions .

Comparative Analysis with Other LPAR2 Antibodies

The HRP-conjugated variant is part of a broader toolkit for LPAR2 detection. Key comparisons include:

ConjugateApplicationsSensitivitySupplier Examples
HRPELISAHighAbbexa , Assay Genie
UnconjugatedWB, IHC, IFModerateBiomatik , Leading Biology
BiotinELISA, Flow CytometryHighCusabio

References to Key Studies

  • Radiation Protection: LPAR2 activation via HRP-linked antibody studies demonstrated reduced oxidative stress and cofilin activation in irradiated colonic epithelium .

  • Vascular Research: HRP-conjugated antibodies confirmed LPAR2’s role in endothelial permeability regulation using murine DVT models .

  • Drug Toxicity: Highlighted LPAR2’s dual role in NSAID-induced enteropathy, balancing mucosal integrity and inflammation .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch products within 1-3 business days of receiving your order. Delivery times may vary depending on your location and chosen shipping method. For specific delivery estimates, please consult your local distributor.
Synonyms
LPAR2; EDG4; LPA2; Lysophosphatidic acid receptor 2; LPA receptor 2; LPA-2; Lysophosphatidic acid receptor Edg-4
Target Names
Uniprot No.

Target Background

Function
The LPAR2 receptor binds to lysophosphatidic acid (LPA), a signaling molecule involved in a wide range of cellular processes. LPAR2 is known to interact with the G(i)/G(o), G(12)/G(13), and G(q) families of heterotrimeric G proteins. It plays a crucial role in the phospholipase C-beta (PLC-beta) signaling pathway, stimulating phospholipase C (PLC) activity independently of RALA activation.
Gene References Into Functions
  1. Research suggests that LPA2 and LPA3 receptors exert opposing effects during red blood cell differentiation. PMID: 27244685
  2. Studies indicate that LPA signaling through LPA2 may play a significant role in regulating cellular functions in HT1080 cells treated with cisplatin. PMID: 28205098
  3. Elevated LPA2 mRNA levels are associated with poorer differentiation, while higher LPA6 levels are linked to microvascular invasion in hepatocellular carcinoma (HCC). When combined with increased serum ATX levels, both factors become risk indicators for recurrence after surgical treatment. PMID: 27583415
  4. Epithelial dysplasia was observed in founder mouse intestine, indicating a correlation between LPA2 overexpression and epithelial dysplasia. This study demonstrates that overexpression of LPA2 alone can lead to intestinal dysplasia. PMID: 27124742
  5. LPA2 expression was found to be associated with HIF-1alpha expression. High levels of LPA2 were linked to shorter overall survival and served as an independent prognostic predictor for breast cancer in Chinese women. PMID: 27805252
  6. LPAR2 mRNA is upregulated in colorectal cancer. PMID: 26937138
  7. Data suggest that high expression levels of LPAR2 and LPAR1 in endometrial cancer tissue, positively correlated with FIGO stage, indicate their potential as biomarkers for endometrial cancer progression. PMID: 26327335
  8. Evidence suggests that LPA2 and LPA3 may act as a molecular switch and play opposing roles during megakaryopoiesis of K562 cells. PMID: 25463482
  9. The RhoA-regulated formin Dia1 is implicated in entosis downstream of LPAR2. PMID: 24950964
  10. The crystal structure of the NHERF2 PDZ1 domain complex with the C-terminal LPA2 sequence has been determined. Binding specificity between PDZ1 and LPA2 is achieved through hydrogen bonds and hydrophobic interactions, with the last four LPA2 residues contributing to specific interactions. PMID: 24613836
  11. LPA1 and LPA2 are the primary LPA receptor subtypes compared to the low expression of LPA3 in PANC-1 tumor cells. PMID: 24061591
  12. Lysophosphatidic acid (LPA) increased hepatocellular carcinoma cells' cell invasion, which was dependent on LPA receptors. PMID: 23569130
  13. LPA2 and LPA6 receptor subtypes are predominant in both human pulmonary artery endothelial cells (HPAECs) and human microvascular endothelial cells (HMVECs). PMID: 23084965
  14. Research found that LPA receptor 2/3-mediated IL-8 expression occurs through Gi/PI3K/AKT, Gi/PKC, and IkappaB/NF-kappaB signaling pathways. PMID: 21964883
  15. This study demonstrates for the first time that key components of the LPA pathway are modulated following traumatic brain injuries in humans. PMID: 21234797
  16. MAGI-3 competes with NHERF-2 to negatively regulate LPA2 receptor signaling in colon cancer cells. PMID: 21134377
  17. LPA2 gene mutations may play a role in the pathogenesis of colon cancer. PMID: 20890765
  18. Studies show that human microglia express LPA receptor subtypes LPA(1), LPA(2), and LPA(3) at both mRNA and protein levels. LPA activation of C13NJ cells induced Rho and extracellular signal-regulated kinase activation, enhancing cellular ATP production. PMID: 19899077
  19. Research demonstrates that two biological fluids, blood plasma and seminal plasma, differentially activate LPA receptors. PMID: 12123830
  20. Findings suggest that LPA(2) and LPA(3) may be involved in VEGF expression mediated by LPA signals during human ovarian oncogenesis. PMID: 12668280
  21. LPA may directly increase the level of cyclin D1 in ovarian cancer cells, promoting their proliferation. PMID: 12759391
  22. Upregulation of LPA2 may play a role in carcinogenesis, particularly in postmenopausal breast cancer. PMID: 15535846
  23. LPA2 is the primary LPA receptor in colon cancer cells, and cellular signals by LPA2 are largely mediated through its interaction with NHERF2. PMID: 15728708
  24. The formation of the LPA receptor/PDZ domain-containing RhoGEF complex plays a pivotal role in LPA-induced RhoA activation. PMID: 15755723
  25. These findings demonstrate that MAGI-3 interacts directly with LPA(2) and regulates its ability to activate Erk and RhoA. PMID: 16904289
  26. EDG4 and EDG2 cooperate to enhance LPA-stimulated chemotaxis in breast tumor cell lines. PMID: 17496233
  27. Data suggest that LPA receptor-dependent expression of CTGF and CYR61 represents a common host response following interaction with bacteria. PMID: 17765657
  28. The lysophosphatidic acid 2 receptor mediates down-regulation of Siva-1 to promote cell survival. PMID: 17965021
  29. A role for the transgenic lysophosphatidic acid (LPA)2 receptor has been identified in regulating smooth muscle cell migratory responses in the context of vascular injury. PMID: 18703779
  30. LPA and LPA receptors, including LPA(2) and LPA(1), represent potential therapeutic targets for patients with mesothelioma. PMID: 18754873
  31. Expression of LPA2 during ovarian carcinogenesis contributes to ovarian cancer aggressiveness, suggesting that targeting LPA production and action may hold potential for treating ovarian cancer. PMID: 19001604
  32. Switching of LPA receptor expression from LPA3 to LPA1 may be involved in prostate cancer progression and/or androgen independence. PMID: 19025891
  33. LPA(1) receptor, LPA(2), and LPA(3) receptors-induced VASP phosphorylation is a critical mediator of tumor cell migration initiation. PMID: 19081821
  34. LPA2 and Gi/Src pathways are significant for LPA-induced COX-2 expression and cell migration, which could be a promising drug target for ovarian cancer cell metastasis. PMID: 19116446

Show More

Hide All

Database Links

HGNC: 3168

OMIM: 605110

KEGG: hsa:9170

STRING: 9606.ENSP00000384665

UniGene: Hs.122575

Protein Families
G-protein coupled receptor 1 family
Subcellular Location
Cell surface. Cell membrane; Multi-pass membrane protein. Note=Prior to LPA treatment found predominantly at the cell surface but in the presence of LPA colocalizes with RALA in the endocytic vesicles.
Tissue Specificity
Expressed most abundantly in testes and peripheral blood leukocytes with less expression in pancreas, spleen, thymus and prostate. Little or no expression in heart, brain, placenta, lung, liver, skeletal muscle, kidney, ovary, small intestine, or colon.

Q&A

What is LPAR2 and what cellular functions does it mediate?

LPAR2 (lysophosphatidic acid receptor 2) is a G-protein coupled receptor that functions as a receptor for lysophosphatidic acid (LPA), mediating diverse cellular activities. In humans, the canonical protein has 348 amino acid residues with a molecular mass of 38.7 kDa and is primarily localized in the cell membrane . LPAR2 belongs to the G-protein coupled receptor 1 family and is also known by several synonyms including EDG4, LPA-2, and LPA2 . The receptor plays critical roles in cell proliferation, migration, and survival pathways, with recent research highlighting its importance in maintaining endothelial barrier integrity in acute lung injury models .

What is the tissue distribution pattern of LPAR2?

LPAR2 exhibits a distinct expression pattern across human tissues. It is most abundantly expressed in testes and peripheral blood leukocytes, with lower expression levels detected in the pancreas, spleen, thymus, and prostate . This differential expression pattern suggests tissue-specific functions of LPAR2 and should be considered when designing experiments targeting this receptor. When evaluating antibody performance in specific tissue types, researchers should account for these varying expression levels to establish appropriate detection thresholds.

What are the primary applications for LPAR2 Antibody, HRP conjugated?

The HRP-conjugated LPAR2 antibody is specifically optimized for several immunodetection techniques:

ApplicationSample TypeTypical Dilution RangeKey Considerations
Western BlotCell/tissue lysates1:500-1:2000Reducing conditions may affect epitope recognition
ELISAPurified protein, serum1:1000-1:5000Direct detection without secondary antibody
ImmunohistochemistryFFPE tissue sections1:100-1:500May require antigen retrieval optimization
ImmunofluorescenceFixed cells1:100-1:500HRP substrate selection crucial for sensitivity

The HRP conjugation eliminates the need for secondary antibody incubation, reducing experimental time and potential cross-reactivity issues . For each application, optimization of antibody concentration against your specific sample type is recommended to maximize signal-to-noise ratio.

How should researchers optimize LPAR2 Antibody, HRP conjugated for Western blot applications?

Optimization of Western blot protocols for LPAR2 detection requires careful consideration of several parameters:

  • Sample preparation: LPAR2 is a membrane protein, requiring effective membrane protein extraction methods. Lysis buffers containing 1-2% non-ionic detergents (Triton X-100 or NP-40) effectively solubilize LPAR2 while preserving epitope integrity.

  • Reducing conditions: As LPAR2 contains multiple cysteine residues that may form disulfide bonds, compare reducing and non-reducing conditions to determine optimal epitope exposure.

  • Transfer parameters: Extended transfer times (90-120 minutes) or semi-dry transfer systems may improve transfer efficiency of this transmembrane protein.

  • Blocking optimization: Test 5% BSA versus 5% non-fat dry milk in TBS-T to minimize background while preserving specific binding.

  • Signal development: For HRP-conjugated antibodies, chemiluminescent substrates with varying sensitivity levels should be tested to optimize detection without signal saturation.

When troubleshooting, compare results against positive control lysates from tissues known to express high LPAR2 levels (testes, leukocytes) and consider using recombinant LPAR2 protein as a standard .

What considerations should researchers take when using LPAR2 Antibody, HRP conjugated in immunohistochemistry?

For optimal immunohistochemical detection of LPAR2 using HRP-conjugated antibodies:

  • Fixation evaluation: Compare results between formalin-fixed paraffin-embedded (FFPE) and frozen sections, as membrane protein epitopes can be differentially preserved.

  • Antigen retrieval: Test multiple antigen retrieval methods (citrate buffer pH 6.0, EDTA buffer pH 9.0, enzymatic retrieval) to determine optimal epitope exposure.

  • Permeabilization: Since LPAR2 is a membrane protein, mild permeabilization (0.1-0.3% Triton X-100) may enhance antibody accessibility without disrupting tissue architecture.

  • Signal amplification: For low-abundance expression, consider tyramide signal amplification (TSA) systems compatible with HRP-conjugated antibodies.

  • Counterstaining compatibility: Certain counterstains may mask HRP-developed signals; optimize counterstaining intensity accordingly.

Given LPAR2's variable tissue expression, include positive control tissues (testes, leukocyte-rich tissues) and negative controls (antibody diluent only) in each experiment to validate staining specificity .

How can researchers effectively validate the specificity of LPAR2 Antibody, HRP conjugated?

Comprehensive validation of LPAR2 antibody specificity involves multiple complementary approaches:

  • Genetic validation: Compare staining patterns between wild-type samples and those from Lpar2 knockout models. Recent studies utilizing Lpar2-/- mice demonstrate the utility of this approach for validation .

  • Peptide competition: Pre-incubate antibody with excess immunizing peptide to confirm signal reduction in positive samples.

  • Orthogonal detection methods: Correlate protein detection with mRNA expression using RT-PCR or RNA-seq data from the same samples.

  • Multiple antibody validation: Compare results with antibodies targeting different LPAR2 epitopes to confirm consistent localization patterns.

  • Heterologous expression: Test antibody reactivity in cell lines with controlled LPAR2 overexpression versus non-expressing controls.

How can LPAR2 Antibody, HRP conjugated be employed to study endothelial barrier function in acute lung injury models?

Recent research demonstrates that LPAR2 plays a protective role in maintaining endothelial barrier integrity during septic acute lung injury (ALI). When designing experiments to investigate this pathway:

  • Experimental models: Compare lipopolysaccharide (LPS)-induced ALI models in wild-type versus Lpar2-/- mice, assessing parameters such as vascular permeability, inflammatory cell infiltration, and cytokine production .

  • Endothelial permeability assays: Implement in vivo permeability assays using Evans blue dye extravasation to quantify barrier function differences, as documented in recent studies showing increased leakage in Lpar2-deficient mice .

  • Mechanistic pathway analysis: Employ the HRP-conjugated LPAR2 antibody in combination with other pathway-specific antibodies to investigate the PLC-PKC-FAK signaling cascade through which LPAR2 mediates barrier protection .

  • Therapeutic intervention assessment: Evaluate potential LPAR2-targeting therapeutic compounds (such as DBIBB) using the antibody to correlate receptor engagement with functional outcomes in lung injury models .

Data interpretation should account for the complex interplay between LPAR2 activation and downstream effects on tight junction proteins (ZO-1, claudin-5) and adherens junction proteins (VE-cadherin) as demonstrated in recent publications .

What are the considerations for multiplexed detection using LPAR2 Antibody, HRP conjugated alongside other markers?

Multiplexed protein detection involving LPAR2 requires careful experimental design:

  • Sequential detection strategies: For HRP-conjugated antibodies in multiplexed IHC, implement sequential detection with complete HRP inactivation between rounds using hydrogen peroxide treatment.

  • Chromogen selection: When using multiple HRP-conjugated antibodies, select chromogens with distinct spectral properties (DAB, AEC, TMB) for clear signal discrimination.

  • Compatible fixation methods: Identify fixation protocols that preserve epitopes for all target proteins in the multiplexed panel.

  • Cross-reactivity testing: Validate the absence of cross-reactivity between antibodies in the multiplex panel through single-staining controls.

  • Digital analysis parameters: For quantitative assessment, establish non-overlapping spectral parameters for each chromogen to allow accurate digital separation of signals.

This approach enables simultaneous visualization of LPAR2 with binding partners or downstream signaling components in the same tissue section.

How can researchers optimize detection sensitivity for low-abundance LPAR2 expression?

For tissues with low LPAR2 expression (e.g., pancreas, spleen), sensitivity enhancement strategies include:

  • Signal amplification systems: Implement tyramide signal amplification (TSA) compatible with HRP-conjugated antibodies, which can increase detection sensitivity 10-100 fold.

  • Extended primary antibody incubation: Overnight incubation at 4°C may enhance binding to low-abundance targets without increasing background.

  • Substrate selection: Compare standard DAB with more sensitive alternatives like DAB-Ni or Nova Red for chromogenic applications, or use enhanced chemiluminescent substrates for Western blotting.

  • Sample enrichment techniques: Consider membrane protein enrichment protocols or immunoprecipitation prior to Western blot analysis.

  • Digital enhancement: Employ computational image analysis with background correction algorithms to reveal subtle expression patterns.

These approaches should be systematically evaluated to determine the optimal combination for specific experimental contexts while maintaining specificity.

What are common issues with LPAR2 Antibody, HRP conjugated and their solutions?

IssuePossible CausesSolutions
High backgroundExcessive antibody concentration; Insufficient blockingTitrate antibody; Extend blocking time; Test alternative blocking reagents
Weak/no signalLow target expression; Epitope masking; Inactive HRPInclude positive control tissue; Optimize antigen retrieval; Test freshly prepared antibody
Non-specific bands (WB)Cross-reactivity; Sample degradationIncrease washing stringency; Use freshly prepared samples with protease inhibitors
Variable resultsInconsistent protocol execution; Antibody degradationStandardize protocol; Aliquot antibody and store at -20°C; Avoid freeze-thaw cycles
Membrane staining not observedInadequate permeabilization; Epitope maskedOptimize detergent concentration; Try alternative fixation methods

When troubleshooting, implement systematic changes to one variable at a time while maintaining appropriate controls to identify the specific factor affecting performance .

How should researchers interpret conflicting results between LPAR2 detection methods?

When faced with discrepancies between different detection methods:

  • Epitope accessibility differences: Different techniques (WB, IHC, IF) expose epitopes differently. Western blotting denatures proteins, potentially revealing epitopes masked in native confirmation used in IF/IHC.

  • Isoform specificity: Confirm which LPAR2 isoforms or post-translational modifications your antibody recognizes, as these may vary between tissue types or experimental conditions.

  • Technical validation: Verify antibody performance in each application through positive and negative controls, especially when translating between techniques.

  • Cross-methodology correlation: When possible, correlate protein detection with mRNA expression data to resolve discrepancies.

  • Literature comparison: Review published findings using similar antibodies to identify consensus patterns and potential technical artifacts.

How is LPAR2 research advancing our understanding of septic acute lung injury?

Recent studies using Lpar2 knockout mice have revealed crucial insights into septic acute lung injury pathophysiology:

  • Barrier protection mechanism: Lpar2 deficiency significantly increased vascular endothelial permeability in LPS-induced septic models, with knockout mice showing:

    • Increased mortality (58.3% vs. 29.2% in wild-type mice)

    • Enhanced lung edema (higher wet-to-dry ratio)

    • Greater Evans blue dye extravasation

    • Elevated inflammatory cytokine production (IL-6, TNF-α, CXCL15)

  • Molecular pathway identification: LPA-LPA2 signaling protects endothelial barrier integrity via the PLC-PKC-FAK pathway, maintaining expression of tight junction proteins (ZO-1, claudin-5) and adherens junction protein VE-cadherin .

  • Therapeutic potential: Pharmacological activation of LPAR2 by the agonist DBIBB significantly reduced inflammation, edema, and hemorrhage while increasing expression of vascular endothelial junction proteins in septic models .

These findings suggest that LPAR2-targeted therapies may represent a novel approach for septic ALI treatment, focusing on endothelial barrier protection rather than purely anti-inflammatory mechanisms.

What novel methodological approaches are emerging for LPAR2 protein quantification and localization?

Emerging technologies enhancing LPAR2 research include:

  • Super-resolution microscopy: Techniques such as STORM and PALM enable nanoscale visualization of LPAR2 membrane distribution and colocalization with signaling partners at resolutions below the diffraction limit.

  • Proximity ligation assays (PLA): These methods can detect protein-protein interactions between LPAR2 and downstream effectors with single-molecule sensitivity in situ.

  • Mass spectrometry-based quantification: Targeted proteomics approaches allow absolute quantification of LPAR2 protein levels across different tissues or experimental conditions.

  • CRISPR-Cas9 engineered reporter systems: Endogenous tagging of LPAR2 enables live-cell imaging of receptor dynamics without overexpression artifacts.

  • Single-cell proteomics: Emerging methods for protein detection at single-cell resolution can reveal LPAR2 expression heterogeneity within tissues.

These methodological advances are expanding our understanding of LPAR2 biology beyond traditional antibody-based approaches, enabling more precise quantification and spatial analysis.

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.