In Vitro Efficacy Testing Services for Eosinophilic Esophagitis
Drug R&D Solutions

In Vitro Efficacy Testing Services for Eosinophilic Esophagitis

Inquiry

We provide robust and sensitive in vitro screening and characterization platforms for accelerating the discovery and screening of potential therapies for Eosinophilic Esophagitis. Our services enable the evaluation of drug candidates targeting key immune pathways, cytokines, and cellular responses implicated in the pathogenesis of this disease. We focus on relevant targets such as eosinophil activation, Th2 cytokine signaling, and epithelial barrier function. Our assays assess processes including inflammation, apoptosis, cellular proliferation, cytokine release, and receptor-ligand interactions to support comprehensive preclinical profiling.

Our in vitro efficacy testing portfolio for Eosinophilic Esophagitis covers a diverse range of biochemical, cellular, and molecular assays. These methods are designed to measure activity, binding affinity, functional impact, and mechanistic action of therapeutic candidates. Collectively, they provide detailed insights into efficacy, potency, and mechanism of action.

ATP assay: Quantifies cellular ATP levels as a measure of cell viability, proliferation, or cytotoxicity.

Annexin V binding assay: Detects early apoptotic cells by binding to phosphatidylserine residues exposed on the cell membrane.

Arrestin protease recruitment assay: Assesses receptor activation by measuring arrestin recruitment, indicative of GPCR signaling.

Biolayer interferometry assay: Measures real-time biomolecular interactions to determine binding kinetics and affinities.

Chemiluminescent assay: Utilizes light emission from chemical reactions to quantify specific analytes or enzymatic activities.

Competitive binding assay: Evaluates the ability of test compounds to compete with labeled ligands for binding to target receptors.

Displacement of [125I]-[Sar1,Ile8]-angiotensin II: Measures the displacement of radiolabeled angiotensin II analogs from receptor binding sites.

Displacement of [125I]-[Tyr4]-angiotensin II: Assesses competition for angiotensin II receptor binding via radioligand displacement.

Displacement of [125I]-angiotensin II: Quantifies compound affinity by measuring the displacement of radiolabeled angiotensin II.

Displacement of [3H]-dexamethasone: Evaluates the ability of compounds to compete for glucocorticoid receptor binding.

Displacement of [3H]-prostaglandin D2: Assesses competitive binding at prostaglandin D2 receptors using tritiated ligand.

Displacement of [3H]-valsartan: Measures receptor-ligand interactions through displacement of radiolabeled valsartan.

Dye assay (alamar blue): Fluorescent dye-based assay for assessing cell viability and metabolic activity.

ELISA assay: Quantitative detection of proteins, cytokines, or antibodies using enzyme-linked immunosorbent techniques.

Flow cytometry assay: Multiparametric analysis of cell populations based on fluorescent labeling of surface or intracellular markers.

Fluorescence resonance energy transfer (FRET) assay: Detects molecular interactions via energy transfer between fluorescent tags.

Fluorescent assay: Utilizes fluorescence-based detection to quantify cellular or molecular events.

Fluorescent-activated cell sorting (FACS) assay: Enables sorting and analysis of specific cell subsets based on fluorescent markers.

Gene reporter assay: Measures gene expression changes by quantifying reporter gene activity under specific promoters.

Homogeneous Time Resolved Fluorescence (HTRF) assay: Non-radioactive, high-sensitivity assay to detect biomolecular interactions or analytes.

Luciferine/luciferase assay: Quantifies gene expression or ATP levels using bioluminescence generated by luciferase enzyme.

Nuclear translocation assay: Measures the movement of signaling molecules or transcription factors into the nucleus.

RNA assay: Detects and quantifies RNA expression levels to assess gene regulation or target engagement.

Radioactivity assay: Employs radiolabeled compounds to quantify binding, uptake, or enzymatic activity.

Saturation binding assay: Determines the maximal binding capacity (Bmax) and affinity (Kd) of compounds for target receptors.

Sensitization with ovalbumin: Models immune sensitization by exposing cells to ovalbumin to assess allergic responses.

Surface plasmon resonance assay: Provides real-time analysis of binding kinetics and affinities between biomolecules.

Thymidine incorporation assay: Measures cell proliferation based on incorporation of radiolabeled thymidine into DNA.

[35S]-GTPgammaS binding assay: Assesses GPCR activation by measuring binding of radiolabeled GTP analogs.

Our assays measure a comprehensive set of pharmacological parameters, including potency, efficacy, affinity, and inhibitory concentrations. These parameters are critical for ranking candidate compounds, optimizing lead selection, and supporting dose-response modeling. Accurate quantification of these metrics enables informed decision-making throughout the drug development process.

EC-300: The concentration of a compound required to achieve 300% of the baseline effect, useful for detailed dose-response analysis.

EC-50: The concentration at which a compound elicits 50% of its maximal effect, indicating potency.

ED-50: The dose required to produce 50% of the maximal therapeutic effect, important for in vitro to in vivo translation.

IC-50: The concentration needed to inhibit a biological process by 50%, commonly used for antagonist evaluation.

Kd: The equilibrium dissociation constant, reflecting the affinity between ligand and receptor.

Ki: The inhibition constant, quantifying the binding affinity of an inhibitor for its target.

MEC: Minimum effective concentration, the lowest concentration at which a compound produces a detectable effect.

MED: Minimum effective dose, the smallest dose achieving a desired therapeutic effect.

MIC: Minimum inhibitory concentration, the lowest concentration inhibiting visible growth of a microorganism or cell population.

pA-2: Negative logarithm of the antagonist dose ratio, used to characterize competitive antagonism.

pEC-50: The negative logarithm of EC-50, providing a more convenient scale for comparing potencies.

pIC-50: The negative logarithm of IC-50, facilitating precise potency comparisons among inhibitors.

pKb: Negative logarithm of the equilibrium constant for antagonist binding, useful for receptor pharmacology.

pKi: Negative logarithm of Ki, commonly used to compare binding affinities across compounds.

Recommended In Vitro Efficacy Tests

Angiotensin Ii Receptor Type 1

The Angiotensin II Receptor Type 1 (AT1R) is implicated in Eosinophilic Esophagitis (EoE) pathogenesis via inflammatory and fibrotic pathways. Testing AT1R is crucial for EoE drug development to assess candidate efficacy and specificity. Our service employs fluorescent and radioactivity assays, including displacement of [125I]- and [3H]-labeled ligands and arrestin recruitment assays. Key parameters measured are pA2, pKi, pIC50, IC50, and Ki, ensuring accurate characterization of AT1R interactions.

Pharmacological Activity Material Method Parameter
Angiotensin AT1 receptor affinity Adrenal gland (cortex), rat Displacement of [125I]-angiotensin II IC-50
Angiotensin AT1 receptor affinity CHO Chinese hamster ovary cells transfected with human receptor Displacement of [125I]-[Sar1,Ile8]-angiotensin II IC-50
Angiotensin AT1 receptor affinity CHO Chinese hamster ovary cells transfected with human receptor Displacement of [125I]-[Tyr4]-angiotensin II Ki
Angiotensin AT1 receptor affinity CHO Chinese hamster ovary cells transfected with human receptor Displacement of [3H]-valsartan Ki
Angiotensin AT1 receptor affinity Cells transfected with receptor Displacement of [125I]-[Sar1,Ile8]-angiotensin II IC-50
Angiotensin AT1 receptor affinity HEK293 human embryonic kidney cells transfected with human receptor Displacement of [125I]-[Sar1,Ile8]-angiotensin II pIC-50
Angiotensin AT1 receptor affinity HEK293T human embryonic kidney cells transfected with human receptor Displacement of [125I]-[Sar1,Ile8]-angiotensin II pKi
Angiotensin AT1 receptor affinity Human receptor Radioactivity assay Ki
Angiotensin AT1 receptor affinity Liver, rat Displacement of [125I]-angiotensin II IC-50
Angiotensin AT1 receptor affinity Myocytes (aorta, thoracic), rat Displacement of [125I]-[Sar1,Ile8]-angiotensin II IC-50
Angiotensin AT1 receptor affinity Myocytes (vascular), rat Displacement of [125I]-[Sar1,Ile8]-angiotensin II IC-50
Angiotensin AT1 receptor affinity Myocytes (vascular), rat Displacement of [125I]-angiotensin II IC-50
Angiotensin AT1 receptor affinity Rat receptor Displacement of [125I]-[Sar1,Ile8]-angiotensin II IC-50
Angiotensin AT1 receptor affinity Smooth muscle cells (vascular), rat Displacement of [125I]-angiotensin II IC-50
Calcium mobilization (angiotensin II-induced), inhibition HEK293 human embryonic kidney cells transfected with AT1 receptor Fluorescent assay IC-50
G-Protein (receptor-linked) activation (angiotensin II-induced), inhibition Cells transfected with human AT1 receptor Arrestin protease recruitment assay IC-50
Muscle contraction (angiotensin II-induced), inhibition Uterus, rat pA-2

Interleukin 13

Interleukin 13 (IL-13) plays a central role in the pathogenesis of Eosinophilic Esophagitis (EoE) by promoting eosinophilic inflammation. Accurate IL-13 testing is crucial for EoE drug development to evaluate therapeutic efficacy and mechanism of action. Key methods include flow cytometry, ELISA, fluorescent, chemiluminescent, surface plasmon resonance, radioactivity assays, and ovalbumin sensitization. Main parameters assessed are ED-50, IC-50, and Kd, providing insights into drug potency and binding affinity.

Pharmacological Activity Material Method Parameter
Chemokine (C-C motif) ligand 26 [CCL26] production (interleukin-13-induced), inhibition HaCaT human keratinocytes Chemiluminescent assay IC-50
Gene (interleukin-13) transcription (antigen-induced), inhibition Bronchoalveolar lavage fluid, rat (sensitized) Sensitization with ovalbumin ED-50
Integrin CD23 expression (interleukin-13-induced), inhibition Lymphocytes, human IC-50
Interleukin-13 affinity Cynomolgus monkey protein Surface plasmon resonance assay Kd
Interleukin-13 affinity Human protein Surface plasmon resonance assay Kd
Interleukin-13 affinity Recombinant cynomolgus monkey protein Surface plasmon resonance assay Kd
Interleukin-13 affinity Recombinant human protein Surface plasmon resonance assay Kd
Interleukin-13 affinity IC-50
Interleukin-13 production, inhibition B9 mouse hybridoma cells IC-50
Interleukin-13/Interleukin-13 receptor subunit alpha-1/Interleukin-4alpha receptor interaction, inhibition HEK293 human embryonic kidney cells transfected with human protein Flow cytometry assay IC-50
Interleukin-13/Interleukin-13 receptor subunit alpha-2 interaction, inhibition COS7 african green monkey kidney cells Radioactivity assay IC-50
Interleukin-13/Interleukin-13 receptor subunit alpha-2 interaction, inhibition Recombinant human receptor ELISA assay IC-50
Mitogenesis (interleukin-13-induced), inhibition TF1 human erythroleukemia cells Chemiluminescent assay IC-50
Mitogenesis (interleukin-13-induced), inhibition TF1 human erythroleukemia cells Fluorescent assay IC-50
Monocyte chemoattractant protein-1 production (interleukin-13-induced), inhibition HaCaT human keratinocytes Chemiluminescent assay IC-50
Protein (NTRK1) expression (interleukin-13-induced), inhibition HaCaT human keratinocytes Chemiluminescent assay IC-50
Signal transducer and activator of transcription-6 (STAT6) phosphorylation (interleukin-13-induced), inhibition HT29 human colon adenocarcinoma cells IC-50
Signal transducer and activator of transcription-6 (STAT6) phosphorylation (interleukin-13-induced), inhibition Lymphocytes, human IC-50
Thymus and activation-regulated chemokine (TARC; CCL17) production (interleukin-13-induced), inhibition A549 human non-small-cell lung carcinoma cells ELISA assay IC-50

Interleukin 15

Interleukin 15 (IL-15) is implicated in Eosinophilic Esophagitis (EoE) by promoting eosinophil survival and inflammation. Accurate IL-15 testing is crucial for drug development targeting EoE. We offer chemiluminescent, surface plasmon resonance, and ELISA assays to quantify IL-15 activity and drug interactions. Key parameters measured include IC-50 (inhibitory concentration) and Kd (binding affinity), providing essential data for therapeutic candidate evaluation.

Pharmacological Activity Material Method Parameter
Interleukin-15 affinity Human protein ELISA assay IC-50
Interleukin-15 affinity Monkey protein ELISA assay IC-50
Interleukin-15 affinity Recombinant protein Surface plasmon resonance assay Kd
Interleukin-15/Interleukin-15 receptor alpha complex affinity Recombinant cynomolgus monkey protein Surface plasmon resonance assay Kd
Interleukin-15/Interleukin-15 receptor alpha complex affinity Recombinant human protein Surface plasmon resonance assay Kd
Mitogenesis (human interleukin-15-induced), inhibition Kit225 human T-cell chronic lymphocytic leukemia cells (interleukin-2-dependent) Chemiluminescent assay IC-50
Mitogenesis (human interleukin-15-induced), inhibition M07e human acute myeloid leukemia cells Chemiluminescent assay IC-50
Mitogenesis (monkey interleukin-15-induced), inhibition Kit225 human T-cell chronic lymphocytic leukemia cells (interleukin-2-dependent) Chemiluminescent assay IC-50
Mitogenesis (monkey interleukin-15-induced), inhibition M07e human acute myeloid leukemia cells Chemiluminescent assay IC-50

Interleukin 4

Interleukin 4 (IL-4) is a key cytokine driving inflammation in Eosinophilic Esophagitis (EoE). Accurate IL-4 testing is crucial for evaluating drug candidates targeting this pathway. Our service utilizes chemiluminescent assays to quantify IL-4 activity and determine inhibitory concentration 50 (IC-50) values, enabling precise assessment of therapeutic efficacy in EoE drug development.

Pharmacological Activity Material Method Parameter
Interleukin-4 production, inhibition HT2 mouse T-lymphocytes (helper) IC-50
Mitogenesis (interleukin-4-induced), inhibition TF1 human erythroleukemia cells Chemiluminescent assay IC-50

Janus Kinase 1

Janus Kinase 1 (JAK1) is a key mediator of inflammatory signaling in Eosinophilic Esophagitis (EoE). JAK1 testing is crucial for identifying and optimizing drug candidates targeting this pathway. Our service employs a sensitive fluorescence resonance energy transfer (FRET) assay to assess compound binding affinity, reporting the primary parameter pKi. This enables precise evaluation of potential JAK1 inhibitors for EoE drug development.

Pharmacological Activity Material Method Parameter
Protein-tyrosine kinase (JAK1), inhibition Recombinant enzyme Fluorescence resonance energy transfer (FRET) assay pKi
Protein-tyrosine kinase (JAK1), inhibition Recombinant human enzyme Fluorescence resonance energy transfer (FRET) assay pKi

Kit Proto-Oncogene, Receptor Tyrosine Kinase

The Kit Proto-Oncogene, Receptor Tyrosine Kinase is implicated in the pathogenesis of Eosinophilic Esophagitis by mediating eosinophil activation and tissue remodeling. Testing its activity is critical for identifying and optimizing targeted therapies. Our service employs chemiluminescent assays to quantify kinase inhibition, providing precise IC-50 values essential for drug candidate evaluation and decision-making in Eosinophilic Esophagitis drug development.

Pharmacological Activity Material Method Parameter
Mitogenesis (stem cell factor-induced), inhibition MO7e human promegakaryocytic leukemia cells Chemiluminescent assay IC-50

Mechanistic Target Of Rapamycin Kinase

Our Mechanistic Target Of Rapamycin (mTOR) Kinase testing service supports Eosinophilic Esophagitis (EoE) drug development by evaluating mTOR’s key role in regulating immune cell proliferation and inflammation in EoE. This testing identifies compounds that inhibit mTOR, crucial for therapeutic targeting. We employ ATP, ELISA, fluorescent, FRET, chemiluminescent, thymidine incorporation, and RNA assays to determine critical parameters such as IC-50, MEC, Kd, and MIC, ensuring robust candidate profiling.

Pharmacological Activity Material Method Parameter
Gene (mTOR) transcription, induction UMB1949 human angiomyolipoma cells (immortalized) (TSC2-mutated) RNA assay MEC
Mammalian target of rapamycin mTOR affinity Kd
Mammalian target of rapamycin mTOR phosphorylation (glucose-induced), inhibition INS1 human pancreatic beta-cells MIC
Mammalian target of rapamycin mTOR phosphorylation (high glucose-induced), inhibition INS1 human pancreatic beta-cells MIC
Mammalian target of rapamycin mTOR phosphorylation, inhibition A549 human non-small-cell lung carcinoma cells (cisplatin-resistant) Chemiluminescent assay MIC
Mammalian target of rapamycin mTOR phosphorylation, inhibition HepG2 human hepatoblastoma cells Chemiluminescent assay MIC
Mammalian target of rapamycin mTOR phosphorylation, inhibition Nasopharyngeal carcinoma cells, human Chemiluminescent assay MIC
Mammalian target of rapamycin mTOR, inhibition HEK293 human embryonic kidney cells transfected with human enzyme Fluorescent assay IC-50
Mammalian target of rapamycin mTOR, inhibition HeLa human cervix adenocarcinoma cells ELISA assay IC-50
Mammalian target of rapamycin mTOR, inhibition Kelly human neuroblastoma cells (ALK-mutated) (MYCN-overexpressing) IC-50
Mammalian target of rapamycin mTOR, inhibition SKBr3 human breast adenocarcinoma cells (HER2 [ERBB2]-overexpressing) (trastuzumab-resistant) IC-50
Mammalian target of rapamycin mTOR, inhibition T-lymphocytes, human (concanavalin A-stimulated) IC-50
Mammalian target of rapamycin mTOR, inhibition U87MG human glioblastoma cells Thymidine incorporation assay IC-50
Mammalian target of rapamycin mTOR, inhibition ATP assay IC-50
Mammalian target of rapamycin mTOR, inhibition Fluorescence resonance energy transfer (FRET) assay IC-50
Mammalian target of rapamycin mTOR, inhibition IC-50
Mammalian target of rapamycin mTOR/FKBP12 interaction, inhibition Human enzyme ATP assay IC-50
Ribosomal protein S6 kinase phosphorylation, inhibition Fibroblasts (embryonic), mouse (TSC1-null) Fluorescent assay IC-50

Nuclear Receptor Subfamily 3 Group C Member 1

Nuclear Receptor Subfamily 3 Group C Member 1 (NR3C1), the glucocorticoid receptor, modulates inflammation in Eosinophilic Esophagitis (EoE). Testing NR3C1 activity is crucial for evaluating drug efficacy and safety in EoE therapy development. Our service employs luciferase, ELISA, fluorescent, FRET, [3H]-dexamethasone displacement, chemiluminescent, HTRF, nuclear translocation, and gene reporter assays. We deliver key pharmacological parameters: MEC, EC-50, MED, EC-300, pEC-50, IC-50, and Ki, supporting robust candidate assessment.

Pharmacological Activity Material Method Parameter
Gene (glucocorticoid receptor) transcription, induction A549 human non-small-cell lung carcinoma cells EC-50
Gene (glucocorticosteroid response element) transcription, induction A549 human non-small-cell lung carcinoma cells ELISA assay EC-300
Gene (glucocorticosteroid response element) transcription, induction A549 human non-small-cell lung carcinoma cells transfected with glucocorticoid response element/luciferase Luciferine/luciferase assay pEC-50
Gene (glucocorticosteroid response element) transcription, induction BEAS2B human bronchial epithelial cells ELISA assay EC-300
Gene (glucocorticosteroid response element) transcription, induction BEAS2B human bronchial epithelial cells transfected with glucocorticosteroid response element Luciferine/luciferase assay MEC
Gene (glucocorticosteroid response element) transcription, induction BEAS2B human bronchial epithelial cells transfected with glucocorticosteroid response element/small interfering RNA for M2 receptor Luciferine/luciferase assay MEC
Gene (glucocorticosteroid response element) transcription, induction HEK293 human embryonic kidney cells transfected with human GR receptor Luciferine/luciferase assay EC-50
Gene (glucocorticosteroid response element) transcription, induction SW1353 human chondrosarcoma cells transfected with human GR receptor Gene reporter assay EC-50
Gene transcription, induction A549 human non-small-cell lung carcinoma cells transfected with glucocorticosteroid-response element Gene reporter assay EC-50
Gene transcription, induction BEAS2B human bronchial epithelial cells transfected with glucocorticosteroid response element Luciferine/luciferase assay pEC-50
Gene transcription, induction BEAS2B human bronchial epithelial cells transfected with glucocorticosteroid response element (TNF alpha-treated) Luciferine/luciferase assay pEC-50
Gene transcription, induction HEK293 human embryonic kidney cells transfected with human GR receptor Luciferine/luciferase assay EC-50
Glucocorticosteroid GR receptor activation, induction CHO-K1 Chinese hamster ovary cells transfected with receptor Nuclear translocation assay EC-50
Glucocorticosteroid GR receptor activation, induction Human receptor Chemiluminescent assay EC-50
Glucocorticosteroid GR receptor activation, induction Nuclear translocation assay MEC
Glucocorticosteroid GR receptor activation, induction EC-50
Glucocorticosteroid GR receptor affinity Human receptor Fluorescent assay Ki
Glucocorticosteroid GR receptor affinity Human receptor Ki
Glucocorticosteroid GR receptor affinity Recombinant human receptor Displacement of [3H]-dexamethasone Ki
Glucocorticosteroid GR receptor affinity Recombinant receptor Fluorescence resonance energy transfer (FRET) assay IC-50
Glucocorticosteroid GR receptor affinity Thymus, rat Displacement of [3H]-dexamethasone IC-50
Glucocorticosteroid GR receptor affinity Homogeneous Time Resolved Fluorescence (HTRF) assay IC-50
Glucocorticosteroid GR receptor expression, induction Brain, rat Chemiluminescent assay MED

Prostaglandin D2 Receptor 2

Prostaglandin D2 Receptor 2 (DP2/CRTH2) mediates eosinophil activation and recruitment in Eosinophilic Esophagitis, driving inflammation. Testing DP2 is crucial for developing targeted therapies. Key assays include saturation binding, fluorescent, [3H]-prostaglandin D2 displacement, [35S]-GTPγS binding, FACS, and Annexin V binding, enabling assessment of ligand-receptor interactions and cellular responses. Main parameters measured are pA-2, Kd, pKb, IC-50, and Ki, critical for evaluating drug potency and receptor affinity.

Pharmacological Activity Material Method Parameter
Apoptosis (prostaglandin D2-induced), inhibition T-lymphocytes (Th2), human Annexin V binding assay IC-50
Calcium mobilization (prostaglandin D2-induced), inhibition CHO Chinese hamster ovary cells transfected with human DP2 (CRTH2) receptor Fluorescent assay IC-50
Cell shape change (prostaglandin D2-induced), inhibition Blood, human Fluorescent-activated cell sorting (FACS) assay pKb
Cell shape change (prostaglandin D2-induced), inhibition Blood, human IC-50
Cell shape change (prostaglandin D2-induced), inhibition Eosinophils, human Fluorescent-activated cell sorting (FACS) assay IC-50
Cell shape change (prostaglandin D2-induced), inhibition Eosinophils, human IC-50
Chemotaxis (prostaglandin D2-induced), inhibition T-lymphocytes (Th2), human Fluorescent-activated cell sorting (FACS) assay IC-50
G-Protein (receptor-linked) activation (prostaglandin D2-induced), inhibition CHO Chinese hamster ovary cells transfected with human DP2 (CRTH2) receptor [35S]-GTPgammaS binding assay pA-2
Prostanoid DP2 (CRTH2) receptor affinity CHO Chinese hamster ovary cells transfected with human receptor Displacement of [3H]-prostaglandin D2 Ki
Prostanoid DP2 (CRTH2) receptor affinity CHO Chinese hamster ovary cells transfected with human receptor Saturation binding assay Kd
Prostanoid DP2 (CRTH2) receptor affinity CHO Chinese hamster ovary cells transfected with rat receptor Displacement of [3H]-prostaglandin D2 Ki
Prostanoid DP2 (CRTH2) receptor affinity CHO Chinese hamster ovary cells transfected with receptor Displacement of [3H]-prostaglandin D2 Ki
Prostanoid DP2 (CRTH2) receptor affinity T-lymphocytes (CD4+), human Displacement of [3H]-prostaglandin D2 Ki
Prostanoid DP2 (CRTH2) receptor internalization (prostaglandin D2-induced), inhibition Blood, human IC-50

Sphingosine-1-Phosphate Receptor 1

The Sphingosine-1-Phosphate Receptor 1 (S1P1) is implicated in Eosinophilic Esophagitis by mediating immune cell trafficking and inflammation. Testing S1P1 activity is crucial for developing targeted therapies. Our service utilizes arrestin protease recruitment and fluorescent assays to assess drug effects on S1P1. Key pharmacological parameters determined include EC50 (potency) and IC50 (inhibition), enabling precise evaluation of candidate compounds for Eosinophilic Esophagitis treatment.

Pharmacological Activity Material Method Parameter
G-Protein (receptor-linked) activation (sphingosine 1-phosphate receptor agonist-induced), inhibition Cells overexpressing S1P1 receptor Arrestin protease recruitment assay IC-50
G-Protein (receptor-linked) activation, induction Cells transfected with human S1P1 receptor Arrestin protease recruitment assay EC-50
G-Protein (receptor-linked) activation, induction HEK293 human embryonic kidney cells transfected with dog S1P1 receptor Arrestin protease recruitment assay EC-50
G-Protein (receptor-linked) activation, induction HEK293 human embryonic kidney cells transfected with human S1P1 receptor Arrestin protease recruitment assay EC-50
G-Protein (receptor-linked) activation, induction HEK293 human embryonic kidney cells transfected with monkey S1P1 receptor Arrestin protease recruitment assay EC-50
G-Protein (receptor-linked) activation, induction HEK293 human embryonic kidney cells transfected with mouse S1P1 receptor Arrestin protease recruitment assay EC-50
Sphingosine 1-phosphate S1P1 receptor internalization, induction CHO Chinese hamster ovary cells transfected with human receptor EC-50
cAMP production (forskolin-induced), inhibition CHO Chinese hamster ovary cells transfected with human S1P1 receptor Fluorescent assay IC-50
cAMP production (forskolin-induced), inhibition Cells transfected with dog S1P1 receptor Fluorescent assay IC-50
cAMP production (forskolin-induced), inhibition Cells transfected with human S1P1 receptor Fluorescent assay IC-50
cAMP production (forskolin-induced), inhibition Cells transfected with monkey S1P1 receptor Fluorescent assay IC-50
cAMP production (forskolin-induced), inhibition Cells transfected with mouse S1P1 receptor Fluorescent assay IC-50
cAMP production (forskolin-induced), inhibition Cells transfected with rat S1P1 receptor Fluorescent assay IC-50
cAMP production (forskolin/IBMX-induced), inhibition HEK293 human embryonic kidney cells transfected with dog S1P1 receptor Fluorescent assay IC-50
cAMP production (forskolin/IBMX-induced), inhibition HEK293 human embryonic kidney cells transfected with human S1P1 receptor Fluorescent assay IC-50
cAMP production (forskolin/IBMX-induced), inhibition HEK293 human embryonic kidney cells transfected with monkey S1P1 receptor Fluorescent assay IC-50
cAMP production (forskolin/IBMX-induced), inhibition HEK293 human embryonic kidney cells transfected with mouse S1P1 receptor Fluorescent assay IC-50
cAMP production (forskolin/IBMX-induced), inhibition HEK293 human embryonic kidney cells transfected with rat S1P1 receptor Fluorescent assay IC-50

Sphingosine-1-Phosphate Receptor 4

Sphingosine-1-Phosphate Receptor 4 (S1P4) regulates immune cell trafficking and inflammation in Eosinophilic Esophagitis (EoE). Testing S1P4 activity is crucial for identifying potential EoE therapeutics targeting this pathway. Our service utilizes an arrestin protease recruitment assay to measure receptor activation, providing EC-50 values as a key parameter for drug potency and efficacy evaluation. This enables precise candidate selection in EoE drug development.

Pharmacological Activity Material Method Parameter
G-Protein (receptor-linked) activation, induction Cells transfected with human S1P4 receptor Arrestin protease recruitment assay EC-50
G-Protein (receptor-linked) activation, induction HEK293 human embryonic kidney cells transfected with human S1P4 receptor Arrestin protease recruitment assay EC-50

Sphingosine-1-Phosphate Receptor 5

Our Sphingosine-1-Phosphate Receptor 5 (S1PR5) testing service supports Eosinophilic Esophagitis drug development by assessing S1PR5’s role in modulating immune cell trafficking and inflammation. This testing is crucial for identifying compounds that effectively target S1PR5. Using the Arrestin protease recruitment assay, we precisely measure compound potency, providing key data such as EC-50 values to guide candidate selection and optimization.

Pharmacological Activity Material Method Parameter
G-Protein (receptor-linked) activation, induction Cells transfected with human S1P5 receptor Arrestin protease recruitment assay EC-50
G-Protein (receptor-linked) activation, induction HEK293 human embryonic kidney cells transfected with human S1P5 receptor Arrestin protease recruitment assay EC-50

Thymic Stromal Lymphopoietin

Thymic Stromal Lymphopoietin (TSLP) is a key cytokine implicated in the pathogenesis of Eosinophilic Esophagitis (EoE) by driving Th2-mediated inflammation. Accurate TSLP testing is vital for EoE drug development to evaluate therapeutic efficacy and mechanism of action. We offer a comprehensive panel of assays—including luciferase, ELISA, FRET, chemiluminescent, flow cytometry, FACS, SPR, BLI, competitive binding, and alamar blue—to quantify TSLP activity and binding, delivering critical parameters such as IC-50 and Kd.

Pharmacological Activity Material Method Parameter
Gene (STAT5) activation (Escherichia coli derived-thymic stromal lymphopoietin-induced), inhibition BAF3 mouse lymphoblasts transfected with human TSLP receptor/IL7R/STAT5/luciferase Luciferine/luciferase assay IC-50
Gene (STAT5) activation (thymic stromal lymphopoietin-induced), inhibition BAF3 mouse lymphoblasts transfected with human TSLP receptor/IL7R/STAT5/luciferase Luciferine/luciferase assay IC-50
Gene (TSLP) transcription (thymic stromal lymphopoietin-induced), inhibition Luciferine/luciferase assay IC-50
Gene (TSLP) transcription, inhibition HEK293 human embryonic kidney cells transfected with human TSLP receptor/IL7R/STAT5/luciferase Luciferine/luciferase assay IC-50
Mitogenesis (human thymic stromal lymphopoietin-induced), inhibition BAF3 mouse lymphoblasts transfected with human TSLP receptor/IL7R Chemiluminescent assay IC-50
Mitogenesis (thymic stromal lymphopoietin-induced), inhibition BAF3 mouse lymphoblasts (TSLP/IL7R-overexpressing) Chemiluminescent assay IC-50
Mitogenesis (thymic stromal lymphopoietin-induced), inhibition BAF3 mouse lymphoblasts transfected with human TSLP receptor/IL7R Chemiluminescent assay IC-50
Mitogenesis (thymic stromal lymphopoietin-induced), inhibition BAF3 mouse lymphoblasts transfected with human TSLP receptor/IL7R Dye assay (alamar blue) IC-50
Mitogenesis (thymic stromal lymphopoietin-induced), inhibition BAF3 mouse lymphoblasts transfected with human TSLP receptor/IL7R IC-50
Signal transducer and activator of transcription-5 (STAT5) phosphorylation (cynomolgus monkey thymic stromal lymphopoietin-induced), inhibition SW756 human cervix squamous-cell carcinoma cells Luciferine/luciferase assay IC-50
Signal transducer and activator of transcription-5 (STAT5) phosphorylation (human thymic stromal lymphopoietin-induced), inhibition BAF3 mouse lymphoblasts transfected with human TSLP receptor/IL7R Flow cytometry assay IC-50
Signal transducer and activator of transcription-5 (STAT5) phosphorylation (human thymic stromal lymphopoietin-induced), inhibition SW756 human cervix squamous-cell carcinoma cells Luciferine/luciferase assay IC-50
Signal transducer and activator of transcription-5 (STAT5) phosphorylation (thymic stromal lymphopoietin-induced), inhibition SW756 human cervix squamous-cell carcinoma cells Luciferine/luciferase assay IC-50
Thymic stromal lymphopoietin (TSLP) affinity BAF3 mouse lymphoblasts transfected with human TSLP receptor/IL7R Fluorescent-activated cell sorting (FACS) assay IC-50
Thymic stromal lymphopoietin (TSLP) affinity CHO-K1 Chinese hamster ovary cells transfected with receptor/IL7R Fluorescent-activated cell sorting (FACS) assay IC-50
Thymic stromal lymphopoietin (TSLP) affinity Cynomolgus monkey protein Surface plasmon resonance assay Kd
Thymic stromal lymphopoietin (TSLP) affinity Human protein Surface plasmon resonance assay Kd
Thymic stromal lymphopoietin (TSLP) affinity Recombinant cynomolgus monkey protein ELISA assay IC-50
Thymic stromal lymphopoietin (TSLP) affinity Recombinant cynomolgus monkey protein Surface plasmon resonance assay Kd
Thymic stromal lymphopoietin (TSLP) affinity Recombinant human protein Biolayer interferometry assay Kd
Thymic stromal lymphopoietin (TSLP) affinity Recombinant human protein Competitive binding assay IC-50
Thymic stromal lymphopoietin (TSLP) affinity Recombinant human protein ELISA assay IC-50
Thymic stromal lymphopoietin (TSLP) affinity Recombinant human protein Surface plasmon resonance assay Kd
Thymic stromal lymphopoietin (TSLP)/Cytokine receptor like factor 2 (CRLF2) interaction, inhibition Recombinant human protein ELISA assay IC-50
Thymic stromal lymphopoietin (TSLP)/Cytokine receptor like factor 2 (CRLF2) interaction, inhibition Recombinant human protein Fluorescence resonance energy transfer (FRET) assay IC-50
Thymic stromal lymphopoietin (TSLP)/Interleukin 7 receptor (IL7R) interaction, inhibition Human protein ELISA assay IC-50
Thymic stromal lymphopoietin (TSLP)/Interleukin 7 receptor (IL7R) interaction, inhibition Recombinant human protein ELISA assay IC-50
Thymic stromal lymphopoietin (TSLP)/Thymic stromal lymphopoietin receptor (TSLPR) interaction, inhibition BAF3 mouse lymphoblasts (TSLP/IL7R-overexpressing) Flow cytometry assay IC-50
Thymic stromal lymphopoietin (TSLP)/Thymic stromal lymphopoietin receptor (TSLPR) interaction, inhibition HEK293F human embryonic kidney cells transfected with TSLP receptor Flow cytometry assay IC-50
Thymic stromal lymphopoietin (TSLP)/Thymic stromal lymphopoietin receptor (TSLPR) interaction, inhibition Human protein Chemiluminescent assay IC-50
Thymic stromal lymphopoietin (TSLP)/Thymic stromal lymphopoietin receptor (TSLPR) interaction, inhibition Recombinant human protein ELISA assay IC-50
HOW WE WORK

Make Order

Make Order

Experimental Scheme

Experimental Scheme

Implementation

Implementation

Conclusion

Conclusion
Make an Inquiry