LPAR2 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery timelines may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery details.
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
LPAR2 is a receptor for lysophosphatidic acid (LPA), a signaling molecule involved in a wide range of cellular activities. It is coupled to the G(i)/G(o), G(12)/G(13), and G(q) families of heterotrimeric G proteins, playing a crucial role in the phospholipase C-beta (PLC-beta) signaling pathway. LPAR2 stimulates phospholipase C (PLC) activity independently of RALA activation.
Gene References Into Functions
  1. Research suggests that LPA2 and LPA3 receptors have opposing roles 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. Higher levels of LPA6 are associated with microvascular invasion in hepatocellular carcinoma (HCC), while increased LPA2 mRNA levels correlate with poorer differentiation. Both factors, when combined with elevated serum ATX levels, are associated with a higher risk of recurrence after surgical treatment. PMID: 27583415
  4. Overexpression of LPA2 has been linked to epithelial dysplasia in the intestines of founder mice, suggesting a direct role in intestinal dysplasia development. PMID: 27124742
  5. LPA2 expression has been correlated with HIF-1alpha expression and shorter overall survival in breast cancer patients. High LPA2 levels have been identified as an independent prognostic predictor for breast cancer in Chinese women. PMID: 27805252
  6. LPAR2 mRNA is upregulated in colorectal cancer. PMID: 26937138
  7. High expression levels of LPAR2 and LPAR1 in endometrial cancer tissue, along with positive correlations with FIGO stage, suggest these receptors as potential biomarkers for endometrial cancer progression. PMID: 26327335
  8. LPA2 and LPA3 may act as a molecular switch, playing opposing roles during megakaryopoiesis of K562 cells. PMID: 25463482
  9. The RhoA-regulated formin Dia1 is involved in entosis downstream of LPAR2. PMID: 24950964
  10. The crystal structure of the NHERF2 PDZ1 domain complex with the C-terminal LPA2 sequence reveals that binding specificity is achieved through hydrogen bonds and hydrophobic contacts. The last four LPA2 residues contribute significantly to these interactions. PMID: 24613836
  11. LPA1 and LPA2 are the dominant LPA receptor subtypes in PANC-1 tumor cells, with LPA3 exhibiting low expression. PMID: 24061591
  12. Lysophosphatidic acid (LPA) has been shown to increase hepatocellular carcinoma cell invasion, an effect mediated by LPA receptors. PMID: 23569130
  13. LPA2 and LPA6 receptor subtypes are predominantly expressed in both human pulmonary artery endothelial cells (HPAECs) and human microvascular endothelial cells (HMVECs). PMID: 23084965
  14. LPA receptor 2/3-mediated IL-8 expression is known to occur through Gi/PI3K/AKT, Gi/PKC, and IkappaB/NF-kappaB signaling pathways. PMID: 21964883
  15. Research demonstrates that key components of the LPA pathway are modulated after 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. Mutations in the LPA2 gene may play a role in the pathogenesis of colon cancer. PMID: 20890765
  18. Human microglia express LPA receptor subtypes LPA(1), LPA(2), and LPA(3) at both the mRNA and protein levels. LPA activation of C13NJ cells induces Rho and extracellular signal-regulated kinase activation, enhancing cellular ATP production. PMID: 19899077
  19. Blood plasma and seminal plasma differentially activate LPA receptors. PMID: 12123830
  20. Studies suggest that LPA(2) and LPA(3) may be involved in VEGF expression mediated by LPA signals in human ovarian oncogenesis. PMID: 12668280
  21. LPA can directly increase cyclin D1 levels in ovarian cancer cells, promoting their proliferation. PMID: 12759391
  22. Upregulation of LPA2 may contribute to carcinogenesis, particularly in postmenopausal breast cancer. PMID: 15535846
  23. LPA2 is the primary LPA receptor in colon cancer cells, and its signaling is largely mediated through its interaction with NHERF2. PMID: 15728708
  24. The formation of the LPA receptor/PDZ domain-containing RhoGEF complex plays a vital role in LPA-induced RhoA activation. PMID: 15755723
  25. MAGI-3 interacts directly with LPA(2) and regulates its ability to activate Erk and RhoA. PMID: 16904289
  26. EDG4 and EDG2 collaborate to promote LPA-stimulated chemotaxis in breast tumor cell lines. PMID: 17496233
  27. Data suggests that LPA receptor-dependent expression of CTGF and CYR61 represents a common host response after interaction with bacteria. PMID: 17765657
  28. The lysophosphatidic acid 2 receptor mediates the downregulation of Siva-1 to promote cell survival. PMID: 17965021
  29. The transgenic lysophosphatidic acid (LPA)2 receptor plays a role in regulating smooth muscle cell migratory responses in the context of vascular injury. PMID: 18703779
  30. LPA and LPA receptors, particularly LPA(2) and LPA(1), represent potential therapeutic targets for patients with malignant pleural mesothelioma (MPM). PMID: 18754873
  31. LPA2 expression during ovarian carcinogenesis contributes to ovarian cancer aggressiveness, suggesting that targeting LPA production and action could potentially be used to treat ovarian cancer. PMID: 19001604
  32. The switch in 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 crucial for LPA-induced COX-2 expression and cell migration, making them promising drug targets 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 regulate?

LPAR2 (Lysophosphatidic acid receptor 2) functions as a receptor for lysophosphatidic acid (LPA), which mediates diverse cellular activities. It appears to couple with multiple G protein families, including G(i)/G(o), G(12)/G(13), and G(q), enabling signal transduction through various pathways. LPAR2 plays a crucial role in the phospholipase C-beta (PLC-beta) signaling pathway and stimulates phospholipase C activity in a manner independent of RALA activation . This receptor has been implicated in cellular processes including myogenic cell growth and proliferation, where it activates pro-mitogenic ERK1/2 and AKT signaling pathways . Additionally, LPAR2 has demonstrated dual roles in maintaining mucosal integrity and orchestrating inflammatory responses in models of NSAID-induced enteropathy .

What are the characteristics of commercially available LPAR2 Antibody, FITC conjugated products?

Currently available LPAR2 Antibody, FITC conjugated products are typically polyclonal antibodies raised in rabbits against synthetic peptide sequences from human LPAR2 protein. Specific characteristics include:

PropertyDetails
TargetLysophosphatidic Acid Receptor 2 (LPAR2)
HostRabbit
ClonalityPolyclonal
Species ReactivityHuman
ImmunogenPeptide sequence from Human LPAR2 protein (8-27 AA)
ConjugationFITC (Fluorescein isothiocyanate)
Excitation/Emission499/515 nm
Laser Line488 nm
FormLiquid
Storage Buffer0.01M PBS, pH 7.4, 50% Glycerol, 0.03% Proclin 300
Purity>95%, Protein G purified
Storage-20°C or -80°C, avoid repeated freeze-thaw cycles

This information provides researchers with essential parameters for experimental planning and execution .

How should LPAR2 Antibody, FITC conjugated be stored to maintain optimal activity?

For optimal retention of activity, LPAR2 Antibody, FITC conjugated should be stored at -20°C or -80°C immediately upon receipt. The antibody should be aliquoted into smaller volumes to minimize freeze-thaw cycles, as repeated freezing and thawing significantly compromises antibody integrity and performance. When working with the antibody, thaw aliquots on ice and protect from prolonged exposure to light, as FITC is photosensitive and can photobleach with extended light exposure . For short-term storage (up to one month), the reconstituted antibody can be kept at 2-8°C under sterile conditions, but for longer-term storage (up to six months), maintain at -20°C to -70°C under sterile conditions . When storing diluted working solutions, include carrier proteins (like BSA) to prevent adsorption to tube walls and stabilize the antibody. Always centrifuge briefly before opening the vial to collect all liquid at the bottom, especially after thawing.

What are the optimal sample preparation methods for flow cytometry using LPAR2 Antibody, FITC conjugated?

When preparing samples for flow cytometry using LPAR2 Antibody, FITC conjugated, researchers should follow these methodological steps:

  • Cell preparation: Harvest cells (1×10^6 cells per sample) and wash twice with ice-cold PBS containing 1% BSA.

  • Fixation: If intracellular staining is required, fix cells with 4% paraformaldehyde for 10-15 minutes at room temperature, then permeabilize with 0.1% Triton X-100 in PBS for 5 minutes. For membrane-bound LPAR2, fixation may be optional or performed after staining.

  • Blocking: Incubate cells with blocking buffer (PBS containing 5% normal serum from the same species as the secondary antibody) for 30 minutes to reduce non-specific binding.

  • Antibody staining: Dilute the LPAR2 Antibody, FITC conjugated to the optimal concentration (typically starting with manufacturer's recommendations, often 1:100 to 1:500) in PBS with 1% BSA. Incubate cells with the diluted antibody for 30-60 minutes at 4°C in the dark.

  • Washing: Wash cells thoroughly 3 times with PBS containing 1% BSA to remove unbound antibody.

  • Final preparation: Resuspend cells in 300-500 μL PBS with 1% BSA for flow cytometric analysis. Include appropriate controls, such as unstained cells, isotype controls, and positive and negative controls .

For optimal results, always determine the ideal antibody concentration through titration experiments for your specific cell type and experimental conditions.

How can I perform Western blot analysis using unlabeled LPAR2 antibodies when studying signaling pathways?

For Western blot analysis of LPAR2 when studying associated signaling pathways, follow this detailed protocol:

  • Sample preparation: Extract total protein from cells or tissues using RIPA buffer supplemented with protease and phosphatase inhibitors. Quantify protein concentration using Bradford or BCA assay.

  • Gel electrophoresis: Load 20-30 μg of protein per lane on an SDS-PAGE gel (10-12% is typically suitable for detecting LPAR2, which has a predicted band size of 39 kDa).

  • Transfer: Transfer proteins to a PVDF membrane using standard wet or semi-dry transfer methods.

  • Blocking: Block the membrane with 5% non-fat dry milk or BSA in TBST for 1 hour at room temperature.

  • Primary antibody incubation: Dilute the LPAR2 antibody to 1:500 in blocking buffer and incubate overnight at 4°C. For parallel analysis of signaling molecules, strip and reprobe membranes or run multiple gels with antibodies against ERK1/2, phospho-ERK1/2, AKT, phospho-AKT, and other relevant pathway components .

  • Washing: Wash the membrane 3-4 times with TBST, 5-10 minutes each.

  • Secondary antibody: Incubate with HRP-conjugated secondary antibody (e.g., Anti-Rabbit IgG for rabbit polyclonal primary antibodies) at 1:2000-1:5000 dilution for 1 hour at room temperature.

  • Detection: Develop using enhanced chemiluminescence (ECL) reagent and document on a digital imaging system.

This methodology has been validated in studies investigating LPAR2-mediated signaling in various cell contexts .

What controls should be included when using LPAR2 Antibody, FITC conjugated for immunofluorescence microscopy?

When conducting immunofluorescence microscopy with LPAR2 Antibody, FITC conjugated, the following controls are essential for proper result interpretation:

  • Negative controls:

    • Isotype control: Use FITC-conjugated non-specific IgG from the same host species (rabbit) at the same concentration as the LPAR2 antibody to assess background and non-specific binding.

    • Secondary antibody only control (when using indirect methods): Omit primary antibody to evaluate non-specific binding of secondary reagents.

    • Unstained cells: To establish autofluorescence levels.

  • Specificity controls:

    • Blocking peptide control: Pre-incubate the antibody with the immunizing peptide (residues 8-27 of Human LPAR2) before staining to confirm signal specificity.

    • LPAR2-negative cells or tissues: Use cells known not to express LPAR2 to confirm specificity.

    • siRNA knockdown: Compare staining in cells where LPAR2 expression has been reduced via siRNA to validate antibody specificity.

  • Positive controls:

    • Known LPAR2-expressing cells: Include cells with confirmed LPAR2 expression, such as certain human peripheral blood lymphocytes or COLO205 cells .

  • Co-localization markers:

    • Plasma membrane markers: As LPAR2 is a transmembrane receptor, include markers such as Na⁺/K⁺-ATPase to confirm proper localization.

    • G-protein signaling components: Include markers for associated signaling molecules to evaluate functional contexts.

These controls collectively ensure reliable and interpretable immunofluorescence data when working with LPAR2 Antibody, FITC conjugated .

How can LPAR2 Antibody, FITC conjugated be used to investigate the role of LPAR2 in myogenic differentiation and proliferation?

To investigate LPAR2's role in myogenic differentiation and proliferation using FITC-conjugated antibodies, researchers can implement the following comprehensive approach:

  • Expression profiling during differentiation:

    • Culture C2C12 myoblasts and induce differentiation.

    • At defined timepoints (proliferating myoblasts, early differentiation, mature myotubes), harvest cells for flow cytometry using LPAR2 Antibody, FITC conjugated.

    • Quantify changes in LPAR2 expression levels throughout the differentiation process.

  • Co-labeling experiments:

    • Perform flow cytometry or immunofluorescence with LPAR2 Antibody, FITC conjugated alongside markers of proliferation (Ki67, BrdU incorporation) and differentiation (MyoD, myogenin).

    • Analyze co-expression patterns to determine if LPAR2-positive cells are predominantly proliferating or differentiating.

  • Functional studies with pharmacological modulators:

    • Treat cells with LPA (lysophosphatidic acid) or PA (phosphatidic acid) at concentrations known to activate LPAR2 (typically 1-10 μM).

    • Assess activation of downstream ERK1/2 and AKT signaling pathways through phosphorylation state.

    • Quantify DNA synthesis (via EdU incorporation) and cell proliferation rates.

    • Compare these responses to LPAR1/LPAR2 antagonist (DBIBB) treatment .

  • Single-cell analysis:

    • Sort LPAR2-high and LPAR2-low populations using the FITC-conjugated antibody.

    • Perform single-cell transcriptomics to identify gene expression signatures associated with LPAR2 expression levels.

    • Focus on genes involved in the Autotaxin-LPA axis, which regulates myogenic cell behavior .

This methodological approach has revealed that LPA activates pro-mitogenic signaling pathways like ERK1/2 and AKT in C2C12 myogenic cells and increases intracellular Ca²⁺ concentration, ultimately inducing DNA synthesis and promoting myoblast proliferation .

What methodologies can be employed to study LPAR2 involvement in NSAID-induced gastric and intestinal injury?

To investigate LPAR2's role in NSAID-induced gastric and intestinal injury, researchers can employ the following methodological approaches:

  • Animal models with genetic manipulation:

    • Compare wild-type (WT) and Lpar2 knockout (Lpar2^-/-^) mice in response to NSAID treatment.

    • Administer indomethacin (20-30 mg/kg via gastric gavage) to induce enteropathy.

    • Evaluate tissue damage at multiple timepoints (6h and 24h post-treatment) to capture both early and late events .

  • Pharmacological intervention studies:

    • Treat animals with LPAR2-selective agonists (e.g., DBIBB at 10 mg/kg) prior to NSAID exposure.

    • Assess protective effects against mucosal damage through histological scoring.

    • Include immunocompromised models (NOD/SCID mice) to evaluate immune-dependent versus immune-independent mechanisms .

  • Molecular and cellular analyses:

    • Quantify LPAR2 expression in gastric and intestinal tissues using flow cytometry with FITC-conjugated antibodies.

    • Correlate expression patterns with tissue damage and inflammatory markers.

    • Assess neutrophil recruitment and inflammatory cytokine production.

    • Monitor autotaxin (ATX) activity and expression, as it is the main LPA-producing enzyme .

  • Ex vivo and in vitro validation:

    • Culture primary gastric or intestinal epithelial cells from WT and Lpar2^-/-^ mice.

    • Treat with NSAIDs and evaluate cellular responses including barrier function, apoptosis, and wound healing.

    • Use LPAR2 Antibody, FITC conjugated to confirm receptor expression in cultured cells.

This approach has revealed a dual role for LPAR2 in NSAID enteropathy: it contributes to maintaining mucosal integrity after NSAID exposure while also orchestrating inflammatory responses associated with ulceration. Furthermore, indomethacin treatment reduces both ATX activity and LPAR2 expression, suggesting inhibition of the ATX-LPAR2 axis as an early event in enteropathy pathogenesis .

How can phosphorylation status of downstream signaling molecules be correlated with LPAR2 activation in flow cytometry experiments?

To correlate phosphorylation status of downstream signaling molecules with LPAR2 activation in flow cytometry experiments, researchers should implement the following methodological approach:

  • Cell stimulation and fixation protocol:

    • Culture cells of interest (e.g., human peripheral blood lymphocytes or COLO205 cells) in serum-free medium for 6-12 hours to reduce baseline signaling.

    • Stimulate cells with LPA (1-10 μM) for varying durations (5, 15, 30, 60 minutes) to capture signaling kinetics.

    • Immediately fix cells with 4% paraformaldehyde for 15 minutes to preserve phosphorylation states.

    • Permeabilize with ice-cold 90% methanol or 0.1% Triton X-100 to allow antibody access to intracellular targets.

  • Multiplex antibody staining:

    • Perform simultaneous staining with LPAR2 Antibody, FITC conjugated (detected in FL1 channel) and PE-conjugated antibodies against phosphorylated signaling molecules:

      • Phospho-ERK1/2 (Thr202/Tyr204)

      • Phospho-AKT (Ser473)

      • Phospho-PLC-β

    • Include appropriate isotype controls for each fluorochrome.

  • Pharmacological intervention strategy:

    • Pretreat cells with LPAR2-specific agonists (DBIBB) or antagonists before LPA stimulation.

    • Include inhibitors of specific downstream pathways (U0126 for MEK/ERK, LY294002 for PI3K/AKT, U73122 for PLC) to validate signaling specificity.

  • Data analysis framework:

    • Gate on LPAR2-positive and LPAR2-negative populations.

    • For each population, quantify the phosphorylation status of downstream molecules.

    • Calculate the phosphorylation index (fold change in MFI of stimulated vs. unstimulated cells).

    • Correlate LPAR2 expression level (MFI in FL1) with phosphorylation indices of signaling molecules.

This methodology allows researchers to establish direct relationships between LPAR2 receptor expression levels, activation status, and downstream signaling events at the single-cell level, providing insights into the heterogeneity of cellular responses to LPA stimulation .

What are common issues encountered when using LPAR2 Antibody, FITC conjugated, and how can they be resolved?

When working with LPAR2 Antibody, FITC conjugated, researchers may encounter several technical challenges. Here are common issues and their methodological solutions:

  • Low signal intensity:

    • Cause: Insufficient antibody concentration, degraded antibody, or low LPAR2 expression.

    • Solution: Titrate antibody concentrations (try 1:50 - 1:500 dilutions); ensure proper storage conditions; confirm LPAR2 expression in your sample with positive controls; use signal amplification systems; optimize cell fixation and permeabilization conditions .

  • High background/non-specific staining:

    • Cause: Inadequate blocking, excessive antibody concentration, or cell autofluorescence.

    • Solution: Extend blocking time (60 minutes with 5% serum); optimize antibody dilution; include 0.1% Triton X-100 in wash buffers; implement quenching steps for autofluorescence; use FC receptor blocking reagents for immune cells .

  • Photobleaching:

    • Cause: FITC is susceptible to photobleaching with prolonged light exposure.

    • Solution: Minimize sample exposure to light during all steps; prepare samples in amber tubes; add anti-fade reagents to mounting media for microscopy; consider immediate acquisition after staining .

  • Inconsistent or unexpected molecular weight in Western blots:

    • Cause: Post-translational modifications, protein degradation, or non-specific binding.

    • Solution: Include protein denaturation controls; optimize lysate preparation with appropriate protease inhibitors; validate with alternative LPAR2 antibodies; compare with recombinant LPAR2 protein as a standard; note that LPAR2 has a predicted band size of 39 kDa .

  • Poor reproducibility between experiments:

    • Cause: Variability in cell preparation, antibody handling, or instrument settings.

    • Solution: Standardize protocols; use calibration beads for flow cytometry; implement detailed SOPs; maintain consistent instrument settings between experiments .

These troubleshooting approaches have been validated across multiple experimental systems and can significantly improve the reliability and reproducibility of experiments utilizing LPAR2 Antibody, FITC conjugated .

How can researchers optimize LPAR2 Antibody, FITC conjugated concentration for different experimental techniques?

Optimizing LPAR2 Antibody, FITC conjugated concentration is critical for generating reliable, reproducible results across different experimental techniques. Here's a methodological approach for each technique:

  • Flow Cytometry Titration:

    • Prepare a single-cell suspension from LPAR2-expressing samples.

    • Set up a dilution series (typically 1:10, 1:50, 1:100, 1:200, 1:500, 1:1000).

    • Stain cells with each dilution using identical protocols.

    • Analyze signal-to-noise ratio by comparing mean fluorescence intensity (MFI) of positive population to negative/isotype controls.

    • Calculate staining index: (MFI positive - MFI negative)/2 × standard deviation of negative population.

    • Select the dilution that provides the highest staining index while minimizing background .

  • Fluorescence Microscopy Optimization:

    • Prepare serial dilutions (1:50 to 1:1000) of antibody.

    • Stain identical samples with each dilution.

    • Compare signal intensity, specificity, and background across conditions.

    • Optimal concentration typically provides distinct membrane staining for LPAR2 with minimal cytoplasmic background.

    • Include blocking peptide controls to confirm specificity at the selected concentration .

  • Western Blot Optimization:

    • For using unconjugated LPAR2 antibodies as comparison:

    • Test antibody concentrations ranging from 0.1-10 μg/mL (typically 1 μg/mL is a good starting point).

    • Load constant amounts of protein from LPAR2-expressing samples.

    • The optimal concentration should yield a clear band at approximately 39 kDa with minimal non-specific binding.

    • Validate with positive controls such as human peripheral blood lymphocytes or COLO205 cell lysates .

TechniqueRecommended Starting DilutionOptimization RangeKey Assessment Criteria
Flow Cytometry1:1001:10 - 1:1000Staining index, population separation
Fluorescence Microscopy1:1001:50 - 1:500Signal specificity, membrane localization
Western Blot (unconjugated)1 μg/mL0.1-10 μg/mLBand specificity, signal-to-noise ratio

This systematic approach ensures optimal antibody performance across multiple experimental platforms while conserving valuable reagent .

What are the considerations for using LPAR2 Antibody, FITC conjugated in multicolor flow cytometry panels?

When incorporating LPAR2 Antibody, FITC conjugated into multicolor flow cytometry panels, researchers must consider several methodological factors to ensure optimal panel performance:

  • Spectral properties and panel design:

    • FITC has excitation/emission peaks at 499/515 nm and is optimally excited by a 488 nm laser .

    • Avoid fluorochromes with significant spectral overlap with FITC (such as PE-Cy5, GFP).

    • Use brightener fluorochromes (APC, PE) for markers with low expression levels and dimmer fluorochromes (FITC) for highly expressed targets.

    • Place FITC in a channel where compensation requirements are minimized.

    • Consider brightness hierarchy: if LPAR2 expression is low, substitute FITC with brighter alternatives like Alexa Fluor 488 if available .

  • Compensation controls and panel validation:

    • Prepare single-stained controls for each fluorochrome in your panel using the same cells or compensation beads.

    • Include FMO (Fluorescence Minus One) controls that contain all fluorochromes except FITC to accurately determine LPAR2-positive populations.

    • Validate that LPAR2 staining is not affected by the presence of other antibodies through comparison of single-stained vs. full panel results.

    • Establish appropriate voltages where FITC negative and positive populations are clearly distinguished .

  • Biological and experimental considerations:

    • When studying signaling pathways, include markers for phosphorylated signaling molecules (pERK1/2, pAKT) with fluorochromes that have minimal spectral overlap with FITC.

    • For co-expression studies with other receptors (like LPAR1), ensure antibody combinations do not cause steric hindrance.

    • Include appropriate cellular markers to identify specific populations where LPAR2 function is being investigated.

    • When examining LPAR2 in contexts like myogenic differentiation or NSAID-induced damage, include relevant markers of cell state or damage .

  • Data analysis strategies:

    • Implement appropriate gating strategies that account for potential autofluorescence in the FITC channel.

    • Consider visualization methods like t-SNE or UMAP for high-dimensional data that can reveal relationships between LPAR2 expression and other markers.

    • Quantify not just percent positive cells but also expression level (MFI) of LPAR2 across different cell populations.

By carefully considering these factors, researchers can develop robust multicolor panels that provide reliable data on LPAR2 expression and its relationship to other cellular parameters .

How can LPAR2 Antibody, FITC conjugated be utilized in single-cell analysis to understand heterogeneity in LPAR2 signaling?

LPAR2 Antibody, FITC conjugated offers significant potential for single-cell analysis to unravel heterogeneity in LPAR2 signaling through several methodological approaches:

  • Single-cell sorting based on LPAR2 expression:

    • Use LPAR2 Antibody, FITC conjugated for FACS to isolate cells with varying LPAR2 expression levels (negative, low, medium, high).

    • Subject sorted populations to single-cell RNA sequencing to identify transcriptional signatures associated with different LPAR2 expression levels.

    • This approach can reveal co-regulated genes within the ATX-LPA signaling axis and identify previously unknown heterogeneity in receptor expression patterns .

  • Mass cytometry (CyTOF) integration:

    • Develop panels incorporating anti-LPAR2 antibodies alongside markers for downstream signaling molecules.

    • Simultaneously measure up to 40 parameters at the single-cell level.

    • Analyze with algorithms like SPADE, viSNE, or PhenoGraph to identify cell subpopulations with distinct LPAR2 signaling characteristics.

    • This methodology can reveal rare cell populations with unique LPAR2-mediated responses that might be missed in bulk analyses.

  • Live-cell imaging for signaling dynamics:

    • Use LPAR2 Antibody, FITC conjugated to identify LPAR2-expressing cells.

    • Combine with fluorescent reporters for second messengers (Ca²⁺, cAMP) or signaling events (FRET-based kinase activity sensors).

    • Perform time-lapse microscopy to capture temporal dynamics of signaling responses at the single-cell level.

    • This approach can reveal asynchronous or oscillatory signaling patterns and cell-to-cell variability in response magnitude or kinetics.

  • Spatial transcriptomics and proteomics:

    • Apply LPAR2 Antibody, FITC conjugated in combination with multiplexed in situ hybridization or imaging mass cytometry.

    • Map LPAR2 expression and activation states within tissue microenvironments.

    • Correlate spatial distribution of LPAR2-positive cells with tissue architecture and pathological features.

    • This methodology is particularly valuable for understanding LPAR2's role in complex tissues, such as in NSAID-induced intestinal injury models .

These approaches collectively enable researchers to move beyond population averages and uncover how cellular heterogeneity in LPAR2 expression translates to diverse functional outcomes in physiological and pathological contexts .

What are the current challenges and future perspectives in studying LPAR2 using FITC-conjugated antibodies in the context of drug development?

In the context of drug development targeting LPAR2 pathways, researchers face several challenges when using FITC-conjugated antibodies, alongside promising future perspectives:

  • Current methodological challenges:

    • Receptor internalization dynamics: LPAR2 undergoes internalization upon ligand binding, making surface detection with antibodies challenging. Future methods need to account for receptor trafficking and distinguish surface from internalized receptors.

    • Conformational specificity: Current antibodies may not distinguish between different conformational states of LPAR2 (active vs. inactive). Developing conformation-specific antibodies would enable better assessment of drug effects on receptor activation.

    • Cross-reactivity concerns: Ensuring absolute specificity between closely related receptors (LPAR1-6) remains challenging. Rigorous validation in knockout systems is essential for antibody-based drug screening assays .

    • Photobleaching limitations: FITC's susceptibility to photobleaching limits long-term imaging studies. Alternative fluorophores with greater photostability might be preferred for extended drug-response monitoring .

  • Emerging methodologies and future directions:

    • High-throughput screening platforms: Developing flow cytometry-based assays using LPAR2 Antibody, FITC conjugated for screening compound libraries that modulate LPAR2 expression or internalization.

    • Biosensor development: Creating FRET-based biosensors incorporating LPAR2 antibody fragments to monitor receptor conformational changes in response to drug candidates.

    • Organoid and patient-derived xenograft models: Applying LPAR2 antibodies to evaluate drug efficacy in more physiologically relevant systems, especially for conditions like NSAID-induced enteropathy where LPAR2 has shown dual roles .

    • Combination therapy assessment: Using multiparameter analysis with LPAR2 Antibody, FITC conjugated alongside markers of drug response to identify optimal combination therapies targeting LPAR2 signaling.

  • Translational applications:

    • Biomarker development: LPAR2 expression levels detected by flow cytometry could serve as predictive biomarkers for response to drugs targeting the ATX-LPA axis.

    • Pharmacodynamic monitoring: Using LPAR2 Antibody, FITC conjugated to track receptor expression changes during treatment as pharmacodynamic endpoints.

    • Patient stratification strategies: Implementing flow cytometry-based LPAR2 profiling to identify patient subgroups most likely to benefit from targeted therapies.

As research progresses, developing advanced antibody formats with improved specificity, stability, and functional reporting capabilities will be crucial for fully leveraging LPAR2 as a drug target and biomarker in various pathological contexts .

How does LPAR2 receptor expression correlate with disease progression in experimental models, and how can FITC-conjugated antibodies help monitor therapeutic interventions?

The correlation between LPAR2 receptor expression and disease progression, along with methodologies for monitoring therapeutic interventions using FITC-conjugated antibodies, represents an important frontier in LPAR2 research:

  • Expression dynamics in disease models:

    • In NSAID-induced enteropathy models, indomethacin treatment reduces intestinal expression of LPAR2 mRNA, preceding the development of mucosal damage. This suggests that LPAR2 downregulation is an early pathogenic event rather than a consequence of tissue damage .

    • LPAR2 expression patterns differ between wild-type mice and those lacking LPAR2 (Lpar2^-/-^), with knockout mice showing accelerated mucosal damage and neutrophil recruitment at early timepoints (6 hours) after NSAID administration .

    • In myogenic models, LPAR2 expression correlates with cellular proliferation states, with functional studies demonstrating that LPAR2 activation promotes DNA synthesis and cell proliferation via ERK1/2 and AKT signaling pathways .

  • Monitoring therapeutic interventions with FITC-conjugated antibodies:

    • Pharmacological intervention assessment: LPAR2 Antibody, FITC conjugated can be used in flow cytometry to measure receptor expression changes following treatment with LPAR2 agonists like DBIBB or inhibitors of the ATX-LPA axis.

    • Quantitative monitoring protocol:

      • Collect tissue or blood samples at defined intervals during treatment.

      • Process samples for single-cell suspensions and stain with LPAR2 Antibody, FITC conjugated alongside relevant lineage markers.

      • Quantify both percentage of LPAR2-positive cells and receptor density (MFI) by flow cytometry.

      • Correlate changes in LPAR2 expression with clinical outcomes and biomarkers of disease activity.

    • Bimodal assessment strategy: Combine flow cytometry for quantitative analysis with immunofluorescence microscopy to evaluate changes in LPAR2 localization and tissue distribution during disease progression and treatment .

  • Translational research applications:

    • Treatment response prediction: Baseline LPAR2 expression levels measured by flow cytometry with FITC-conjugated antibodies may predict response to therapies targeting the ATX-LPA signaling axis.

    • Pharmacodynamic marker development: Changes in LPAR2 expression can serve as pharmacodynamic markers for drug efficacy in clinical trials.

    • Therapeutic window identification: By tracking LPAR2 expression during different disease stages, researchers can identify optimal timing for therapeutic interventions targeting this receptor.

  • Methodological considerations for longitudinal monitoring:

    • Establish consistent protocols for sample collection, processing, and staining to ensure comparability across timepoints.

    • Include appropriate controls at each timepoint to account for technical variability.

    • Consider developing standardized quantification methods (such as molecules of equivalent soluble fluorochrome, MESF) to allow absolute quantification of LPAR2 expression levels.

This comprehensive approach enables researchers to establish LPAR2 as both a disease biomarker and a therapeutic target in conditions where the ATX-LPA signaling axis plays a pathogenic role .

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 2024 Thebiotek. All Rights Reserved.