We provide robust and sensitive in vitro screening and characterization platforms for accelerating the discovery and screening of potential therapies for Prader-Willi Syndrome. Our service offers comprehensive efficacy testing of candidate molecules targeting key receptors and signaling pathways implicated in Prader-Willi Syndrome, such as the oxytocin receptor and related neuroendocrine modulators. These targets are central to the regulation of appetite, metabolism, and neurobehavioral symptoms characteristic of the disease. Our assays are designed to elucidate the molecular mechanisms underlying pathological processes including hormone dysregulation and abnormal signal transduction.
Our portfolio encompasses a diverse set of in vitro assay methodologies, including binding, functional, and signaling-based assays. These methods enable the assessment of compound activity, potency, and mechanism of action at critical targets relevant to Prader-Willi Syndrome. This comprehensive approach allows for rigorous evaluation of therapeutic candidates in early-stage drug discovery.
Arrestin protease recruitment assay: Measures the recruitment of arrestin proteins to receptors, providing insights into receptor activation and downstream signaling relevant to neuroendocrine function.
Arrestin protease recruitment assay (with calcium): Evaluates arrestin recruitment in the presence of calcium, allowing for the assessment of calcium-dependent signaling pathways often dysregulated in Prader-Willi Syndrome.
Chemiluminescent assay: Utilizes light emission as a readout for detecting specific biomolecular interactions or enzymatic activity, supporting high-sensitivity quantification of pathway modulation.
Displacement of [3H]-oxytocin: Assesses the binding affinity of test compounds to the oxytocin receptor by measuring their ability to displace radiolabeled oxytocin, which is central to appetite and social behavior regulation.
Fluorescent assay: Detects changes in fluorescence to monitor receptor activation, ligand binding, or intracellular signaling events, providing rapid and quantitative data.
Homogeneous Time Resolved Fluorescence (HTRF) assay: Employs energy transfer between fluorophores to measure molecular interactions or second messenger levels with high sensitivity and throughput.
Luciferine/luciferase assay: Monitors gene expression or cellular signaling via bioluminescence, enabling detection of functional responses to candidate therapies.
Surface plasmon resonance assay: Provides real-time, label-free analysis of biomolecular binding kinetics and affinities, useful for characterizing drug-target interactions.
With calcium: Assays incorporating calcium as a co-factor to evaluate calcium-mediated signaling, which is implicated in the pathophysiology of Prader-Willi Syndrome.
[35S]-GTPgammaS binding assay: Measures G-protein activation following receptor engagement, offering mechanistic insights into GPCR signaling modulation.
cAMP accumulation assay: Quantifies changes in intracellular cAMP levels as a readout of GPCR activity, relevant to neuroendocrine and metabolic regulation.
We measure a range of key pharmacological parameters to provide quantitative assessments of compound potency, efficacy, and binding characteristics. These parameters are instrumental in guiding medicinal chemistry optimization and compound prioritization. By establishing benchmarks such as EC-50, IC-50, Kd, Ki, and MIC, we enable informed decision-making throughout the drug discovery process.
EC-50: The concentration of a compound that produces 50% of its maximal effect, crucial for comparing compound potencies.
IC-50: The concentration required to inhibit a biological process by 50%, valuable for identifying effective inhibitors.
Kd: The dissociation constant reflecting the binding affinity between a drug and its target, essential for understanding target engagement.
Ki: The inhibition constant that quantifies the potency of a compound as an inhibitor, important for ranking and optimizing lead candidates.
MIC: The minimum inhibitory concentration needed to prevent biological activity, used for determining the threshold of efficacy for candidate molecules.
Growth Hormone Receptor (GHR) plays a crucial role in mediating growth hormone effects, often disrupted in Prader-Willi Syndrome (PWS). Accurate GHR testing is essential for PWS drug development to assess drug-target interactions. Our service utilizes surface plasmon resonance (SPR) assays to measure real-time binding between candidate drugs and GHR, providing precise affinity parameters such as the dissociation constant (Kd), critical for evaluating therapeutic potential.
| Pharmacological Activity | Material | Method | Parameter |
|---|---|---|---|
| Growth hormone receptor affinity | Human receptor | Surface plasmon resonance assay | Kd |
Histamine Receptor H3 is implicated in appetite regulation and cognitive function, making it a key target in Prader-Willi Syndrome drug development. Our testing service evaluates H3 receptor modulation using luciferin/luciferase, HTRF, and [35S]-GTPγS binding assays. These methods accurately determine IC-50 values, enabling precise assessment of compound efficacy and potency. This testing is vital for identifying promising drug candidates for Prader-Willi Syndrome.
| Pharmacological Activity | Material | Method | Parameter |
|---|---|---|---|
| G-Protein (receptor-linked) activation (histamine-induced), inhibition | HEK293T human embryonic kidney cells transfected with human H3 receptor | Luciferine/luciferase assay | IC-50 |
| G-Protein (receptor-linked) activation, inhibition | CHO-K1 Chinese hamster ovary cells transfected with human H3 receptor | [35S]-GTPgammaS binding assay | IC-50 |
| cAMP production (forskolin-induced/alpha-methylhistamine-depressed), antagonism | HEK293 human embryonic kidney cells transfected with human H3 receptor | Homogeneous Time Resolved Fluorescence (HTRF) assay | IC-50 |
Melanocortin 1 Receptor (MC1R) testing is vital in Prader-Willi Syndrome drug development as MC1R is implicated in appetite and energy regulation, both key features of the disorder. This service utilizes a chemiluminescent assay to assess MC1R activity in response to drug candidates, providing precise EC-50 measurements. These data enable accurate evaluation of compound potency and help guide therapeutic optimization for Prader-Willi Syndrome.
| Pharmacological Activity | Material | Method | Parameter |
|---|---|---|---|
| Melanocortin MC1 receptor activation, induction | EC-50 | ||
| cAMP production, induction | HEK293T human embryonic kidney cells transfected with human MC1 receptor | Chemiluminescent assay | EC-50 |
The Melanocortin 3 Receptor (MC3R) is implicated in energy homeostasis and appetite regulation, making it a key target in Prader-Willi Syndrome drug development. MC3R testing is essential for evaluating candidate compounds’ efficacy. Our service utilizes a sensitive chemiluminescent assay to measure receptor activation, providing precise EC-50 values for compound potency assessment, thereby supporting informed decision-making in therapeutic development.
| Pharmacological Activity | Material | Method | Parameter |
|---|---|---|---|
| cAMP production, induction | HEK293T human embryonic kidney cells transfected with human MC3 receptor | Chemiluminescent assay | EC-50 |
Melanocortin 4 Receptor (MC4R) plays a key role in regulating appetite and energy balance, making it central to Prader-Willi Syndrome (PWS) drug development. MC4R testing is crucial for evaluating potential therapeutics. Our service utilizes luciferase reporter, cAMP accumulation, chemiluminescent, and arrestin recruitment assays (with/without calcium) to assess MC4R activity. The primary parameter measured is EC-50, providing precise data for drug efficacy profiling in PWS research.
| Pharmacological Activity | Material | Method | Parameter |
|---|---|---|---|
| G-Protein (receptor-linked) activation, induction | HEK293 human embryonic kidney cells transfected with MC4 receptor | Arrestin protease recruitment assay | EC-50 |
| G-Protein (receptor-linked) activation, induction | HEK293T human embryonic kidney cells transfected with human MC4 receptor | Arrestin protease recruitment assay | EC-50 |
| G-Protein (receptor-linked) activation, induction | HEK293T human embryonic kidney cells transfected with human MC4 receptor | Arrestin protease recruitment assay (with calcium) | EC-50 |
| G-Protein (receptor-linked) activation, induction | HEK293T human embryonic kidney cells transfected with human MC4 receptor | With calcium | EC-50 |
| G-Protein (receptor-linked) activation, induction | HEK293T human embryonic kidney cells transfected with human MC4 receptor | EC-50 | |
| Melanocortin MC4 receptor activation, induction | EC-50 | ||
| Serine/threonine protein kinase (Erk) activation, induction | HEK293 human embryonic kidney cells transfected with MC4 receptor | Luciferine/luciferase assay | EC-50 |
| cAMP production, induction | HEK293 human embryonic kidney cells transfected with MC4 receptor | cAMP accumulation assay | EC-50 |
| cAMP production, induction | HEK293T human embryonic kidney cells transfected with human MC4 receptor | Chemiluminescent assay | EC-50 |
Our Oxytocin Receptor testing service supports Prader-Willi Syndrome drug development by evaluating compounds targeting the oxytocin receptor, which is implicated in the syndrome’s social and metabolic symptoms. Utilizing [3H]-oxytocin displacement and fluorescent assays, we determine key parameters—EC-50, Ki, and MIC—to assess compound potency and binding affinity. This testing is essential for identifying effective therapeutic candidates and optimizing drug efficacy and safety profiles.
| Pharmacological Activity | Material | Method | Parameter |
|---|---|---|---|
| Calcium mobilization, induction | CHO Chinese hamster ovary cells transfected with human OT receptor | Fluorescent assay | EC-50 |
| Oxytocin OT receptor (R150S-mutated) affinity | HEK293 human embryonic kidney cells transfected with human receptor | Displacement of [3H]-oxytocin | Ki |
| Oxytocin OT receptor affinity | HEK293 human embryonic kidney cells transfected with human receptor | Displacement of [3H]-oxytocin | Ki |
| Proteolysis, inhibition | Muscle (soleus), rat | Fluorescent assay | MIC |
Make Order
Experimental Scheme
Implementation
Conclusion