Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Med Chem ; 65(12): 8303-8331, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35696646

ABSTRACT

The perinucleolar compartment (PNC) is a dynamic subnuclear body found at the periphery of the nucleolus. The PNC is enriched with RNA transcripts and RNA-binding proteins, reflecting different states of genome organization. PNC prevalence positively correlates with cancer progression and metastatic capacity, making it a useful marker for metastatic cancer progression. A high-throughput, high-content assay was developed to identify novel small molecules that selectively reduce PNC prevalence in cancer cells. We identified and further optimized a pyrrolopyrimidine series able to reduce PNC prevalence in PC3M cancer cells at submicromolar concentrations without affecting cell viability. Structure-activity relationship exploration of the structural elements necessary for activity resulted in the discovery of several potent compounds. Analysis of in vitro drug-like properties led to the discovery of the bioavailable analogue, metarrestin, which has shown potent antimetastatic activity with improved survival in rodent models and is currently being evaluated in a first-in-human phase 1 clinical trial.


Subject(s)
Cell Nucleus , Neoplasms , Biomarkers/metabolism , Cell Nucleolus/metabolism , Cell Nucleolus/pathology , Cell Nucleus/metabolism , Humans , Neoplasms/metabolism , Pyrimidines , Pyrroles
2.
Mol Pharmacol ; 94(4): 1197-1209, 2018 10.
Article in English | MEDLINE | ID: mdl-30068735

ABSTRACT

The D1 dopamine receptor is linked to a variety of neuropsychiatric disorders and represents an attractive drug target for the enhancement of cognition in schizophrenia, Alzheimer disease, and other disorders. Positive allosteric modulators (PAMs), with their potential for greater selectivity and larger therapeutic windows, may represent a viable drug development strategy, as orthosteric D1 receptor agonists possess known clinical liabilities. We discovered two structurally distinct D1 receptor PAMs, MLS6585 and MLS1082, via a high-throughput screen of the NIH Molecular Libraries program small-molecule library. Both compounds potentiate dopamine-stimulated G protein- and ß-arrestin-mediated signaling and increase the affinity of dopamine for the D1 receptor with low micromolar potencies. Neither compound displayed any intrinsic agonist activity. Both compounds were also found to potentiate the efficacy of partial agonists. We tested maximally effective concentrations of each PAM in combination to determine if the compounds might act at separate or similar sites. In combination, MLS1082 + MLS6585 produced an additive potentiation of dopamine potency beyond that caused by either PAM alone for both ß-arrestin recruitment and cAMP accumulation, suggesting diverse sites of action. In addition, MLS6585, but not MLS1082, had additive activity with the previously described D1 receptor PAM "Compound B," suggesting that MLS1082 and Compound B may share a common binding site. A point mutation (R130Q) in the D1 receptor was found to abrogate MLS1082 activity without affecting that of MLS6585, suggesting this residue may be involved in the binding/activity of MLS1082 but not that of MLS6585. Together, MLS1082 and MLS6585 may serve as important tool compounds for the characterization of diverse allosteric sites on the D1 receptor as well as the development of optimized lead compounds for therapeutic use.


Subject(s)
Allosteric Regulation/physiology , Allosteric Site/physiology , Receptors, Dopamine/metabolism , Animals , CHO Cells , Cricetulus , Cyclic AMP/metabolism , Dopamine/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Signal Transduction/physiology , beta-Arrestins/metabolism
3.
Sci Transl Med ; 10(441)2018 05 16.
Article in English | MEDLINE | ID: mdl-29769289

ABSTRACT

Metastasis remains a leading cause of cancer mortality due to the lack of specific inhibitors against this complex process. To identify compounds selectively targeting the metastatic state, we used the perinucleolar compartment (PNC), a complex nuclear structure associated with metastatic behaviors of cancer cells, as a phenotypic marker for a high-content screen of over 140,000 structurally diverse compounds. Metarrestin, obtained through optimization of a screening hit, disassembles PNCs in multiple cancer cell lines, inhibits invasion in vitro, suppresses metastatic development in three mouse models of human cancer, and extends survival of mice in a metastatic pancreatic cancer xenograft model with no organ toxicity or discernable adverse effects. Metarrestin disrupts the nucleolar structure and inhibits RNA polymerase (Pol) I transcription, at least in part by interacting with the translation elongation factor eEF1A2. Thus, metarrestin represents a potential therapeutic approach for the treatment of metastatic cancer.


Subject(s)
Cell Nucleolus/pathology , Neoplasm Metastasis/drug therapy , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Animals , Cell Line, Tumor , Cell Nucleolus/drug effects , Cell Nucleolus/metabolism , Cell Nucleolus/ultrastructure , Cell Proliferation/drug effects , Chromatin/metabolism , DNA, Ribosomal/genetics , Humans , Male , Mice , Neoplasm Invasiveness , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Peptide Elongation Factor 1/metabolism , Promoter Regions, Genetic/genetics , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , RNA Polymerase I/metabolism , RNA Precursors/biosynthesis , Survival Analysis , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
4.
Cell Death Dis ; 7(12): e2492, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27906188

ABSTRACT

The discovery of chemotherapeutic agents for the treatment of cancer commonly uses cell proliferation assays in which cells grow as two-dimensional (2D) monolayers. Compounds identified using 2D monolayer assays often fail to advance during clinical development, most likely because these assays do not reproduce the cellular complexity of tumors and their microenvironment in vivo. The use of three-dimensional (3D) cellular systems have been explored as enabling more predictive in vitro tumor models for drug discovery. To date, small-scale screens have demonstrated that pharmacological responses tend to differ between 2D and 3D cancer cell growth models. However, the limited scope of screens using 3D models has not provided a clear delineation of the cellular pathways and processes that differentially regulate cell survival and death in the different in vitro tumor models. Here we sought to further understand the differences in pharmacological responses between cancer tumor cells grown in different conditions by profiling a large collection of 1912 chemotherapeutic agents. We compared pharmacological responses obtained from cells cultured in traditional 2D monolayer conditions with those responses obtained from cells forming spheres versus cells already in 3D spheres. The target annotation of the compound library screened enabled the identification of those key cellular pathways and processes that when modulated by drugs induced cell death in all growth conditions or selectively in the different cell growth models. In addition, we also show that many of the compounds targeting these key cellular functions can be combined to produce synergistic cytotoxic effects, which in many cases differ in the magnitude of their synergism depending on the cellular model and cell type. The results from this work provide a high-throughput screening framework to profile the responses of drugs both as single agents and in pairwise combinations in 3D sphere models of cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Culture Techniques/methods , Models, Biological , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Spheroids, Cellular/drug effects
5.
JCI Insight ; 1(19): e88427, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27882347

ABSTRACT

Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disease and one of the leading inherited causes of infant mortality. SMA results from insufficient levels of the survival motor neuron (SMN) protein, and studies in animal models of the disease have shown that increasing SMN protein levels ameliorates the disease phenotype. Our group previously identified and optimized a new series of small molecules, with good potency and toxicity profiles and reasonable pharmacokinetics, that were able to increase SMN protein levels in SMA patient-derived cells. We show here that ML372, a representative of this series, almost doubles the half-life of residual SMN protein expressed from the SMN2 locus by blocking its ubiquitination and subsequent degradation by the proteasome. ML372 increased SMN protein levels in muscle, spinal cord, and brain tissue of SMA mice. Importantly, ML372 treatment improved the righting reflex and extended survival of a severe mouse model of SMA. These results demonstrate that slowing SMN degradation by selectively inhibiting its ubiquitination can improve the motor phenotype and lifespan of SMA model mice.


Subject(s)
Muscular Atrophy, Spinal/drug therapy , Survival of Motor Neuron 1 Protein/chemistry , Ubiquitination , Animals , Disease Models, Animal , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic
6.
J Biomol Screen ; 21(10): 1112-1124, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27647668

ABSTRACT

Human neuronal cells differentiated from induced pluripotent cells have emerged as a new model system for the study of disease pathophysiology and evaluation of drug efficacy. Differentiated neuronal cells are more similar in genetics and biological content to human brain cells than other animal disease models. However, culture of neuronal cells in assay plates requires a labor-intensive procedure of plate precoating, hampering its applications in high-throughput screening (HTS). We developed a simplified method with one-step seeding of neural stem cells in assay plates by supplementing the medium with a recombinant human vitronectin (VTN), thus avoiding plate precoating. Robust results were obtained from cell viability, calcium response, and neurite outgrowth assays using this new method. Our data demonstrate that this approach greatly simplifies high-throughput assays using neuronal cells differentiated from human stem cells for translational research.


Subject(s)
Cell Culture Techniques/methods , High-Throughput Screening Assays/methods , Neural Stem Cells/cytology , Vitronectin/pharmacology , Cell Differentiation/genetics , Culture Media/pharmacology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Neural Stem Cells/drug effects
7.
Mol Pharmacol ; 86(1): 96-105, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24755247

ABSTRACT

A high-throughput screening campaign was conducted to interrogate a 380,000+ small-molecule library for novel D2 dopamine receptor modulators using a calcium mobilization assay. Active agonist compounds from the primary screen were examined for orthogonal D2 dopamine receptor signaling activities including cAMP modulation and ß-arrestin recruitment. Although the majority of the subsequently confirmed hits activated all signaling pathways tested, several compounds showed a diminished ability to stimulate ß-arrestin recruitment. One such compound (MLS1547; 5-chloro-7-[(4-pyridin-2-ylpiperazin-1-yl)methyl]quinolin-8-ol) is a highly efficacious agonist at D2 receptor-mediated G protein-linked signaling, but does not recruit ß-arrestin as demonstrated using two different assays. This compound does, however, antagonize dopamine-stimulated ß-arrestin recruitment to the D2 receptor. In an effort to investigate the chemical scaffold of MLS1547 further, we characterized a set of 24 analogs of MLS1547 with respect to their ability to inhibit cAMP accumulation or stimulate ß-arrestin recruitment. A number of the analogs were similar to MLS1547 in that they displayed agonist activity for inhibiting cAMP accumulation, but did not stimulate ß-arrestin recruitment (i.e., they were highly biased). In contrast, other analogs displayed various degrees of G protein signaling bias. These results provided the basis to use pharmacophore modeling and molecular docking analyses to build a preliminary structure-activity relationship of the functionally selective properties of this series of compounds. In summary, we have identified and characterized a novel G protein-biased agonist of the D2 dopamine receptor and identified structural features that may contribute to its biased signaling properties.


Subject(s)
Arrestins/antagonists & inhibitors , GTP-Binding Proteins/metabolism , Receptors, Dopamine D2/metabolism , Animals , Arrestins/metabolism , CHO Cells , Cell Line , Cricetulus , Cyclic AMP/metabolism , HEK293 Cells , Humans , Protein Binding/physiology , Signal Transduction/physiology , Small Molecule Libraries , Structure-Activity Relationship , beta-Arrestins
8.
PLoS One ; 9(2): e89335, 2014.
Article in English | MEDLINE | ID: mdl-24586696

ABSTRACT

Recent genetic and clinical evidence has implicated glucokinase regulatory protein (GKRP) in the pathogenesis of type 2 diabetes and related traits. The primary role of GKRP is to bind and inhibit hepatic glucokinase (GCK), a critically important protein in human health and disease that exerts a significant degree of control over glucose metabolism. As activation of GCK has been associated with improved glucose tolerance, perturbation of the GCK-GKRP interaction represents a potential therapeutic target for pharmacological modulation. Recent structural and kinetic advances are beginning to provide insight into the interaction of these two proteins. However, tools to comprehensively assess the GCK-GKRP interaction, particularly in the context of small molecules, would be a valuable resource. We therefore developed three robust and miniaturized assays for assessing the interaction between recombinant human GCK and GKRP: an HTRF assay, a diaphorase-coupled assay, and a luciferase-coupled assay. The assays are complementary, featuring distinct mechanisms of detection (luminescence, fluorescence, FRET). Two assays rely on GCK enzyme activity modulation by GKRP while the FRET-based assay measures the GCK-GKRP protein-protein interaction independent of GCK enzymatic substrates and activity. All three assays are scalable to low volumes in 1536-well plate format, with robust Z' factors (>0.7). Finally, as GKRP sequesters GCK in the hepatocyte nucleus at low glucose concentrations, we explored cellular models of GCK localization and translocation. Previous findings from freshly isolated rat hepatocytes were confirmed in cryopreserved rat hepatocytes, and we further extended this study to cryopreserved human hepatocytes. Consistent with previous reports, there were several key differences between the rat and human systems, with our results suggesting that human hepatocytes can be used to interrogate GCK translocation in response to small molecules. The assay panel developed here should help direct future investigation of the GCK-GKRP interaction in these or other physiologically relevant human systems.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Enzyme Assays/methods , Glucokinase/metabolism , Hepatocytes/cytology , Animals , Cryopreservation/methods , Data Interpretation, Statistical , Fluorescence , Fluorescence Resonance Energy Transfer , Histological Techniques , Humans , Image Processing, Computer-Assisted , Luciferases , NADH Dehydrogenase , Rats
9.
J Med Chem ; 57(8): 3450-63, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24666157

ABSTRACT

The D2 dopamine receptor (D2 DAR) is one of the most validated drug targets for neuropsychiatric and endocrine disorders. However, clinically approved drugs targeting D2 DAR display poor selectivity between the D2 and other receptors, especially the D3 DAR. This lack of selectivity may lead to undesirable side effects. Here we describe the chemical and pharmacological characterization of a novel D2 DAR antagonist series with excellent D2 versus D1, D3, D4, and D5 receptor selectivity. The final probe 65 was obtained through a quantitative high-throughput screening campaign, followed by medicinal chemistry optimization, to yield a selective molecule with good in vitro physical properties, metabolic stability, and in vivo pharmacokinetics. The optimized molecule may be a useful in vivo probe for studying D2 DAR signal modulation and could also serve as a lead compound for the development of D2 DAR-selective druglike molecules for the treatment of multiple neuropsychiatric and endocrine disorders.


Subject(s)
Dopamine Antagonists/chemical synthesis , Dopamine D2 Receptor Antagonists , Dopamine Antagonists/pharmacology , Drug Discovery , HEK293 Cells , High-Throughput Screening Assays , Humans , Structure-Activity Relationship
10.
Dev Cell ; 26(5): 511-24, 2013 Sep 16.
Article in English | MEDLINE | ID: mdl-23993788

ABSTRACT

Phagocytosis of large extracellular particles such as apoptotic bodies requires delivery of the intracellular endosomal and lysosomal membranes to form plasmalemmal pseudopods. Here, we identified mucolipin TRP channel 1 (TRPML1) as the key lysosomal Ca2+ channel regulating focal exocytosis and phagosome biogenesis. Both particle ingestion and lysosomal exocytosis are inhibited by synthetic TRPML1 blockers and are defective in macrophages isolated from TRPML1 knockout mice. Furthermore, TRPML1 overexpression and TRPML1 agonists facilitate both lysosomal exocytosis and particle uptake. Using time-lapse confocal imaging and direct patch clamping of phagosomal membranes, we found that particle binding induces lysosomal PI(3,5)P2 elevation to trigger TRPML1-mediated lysosomal Ca2+ release specifically at the site of uptake, rapidly delivering TRPML1-resident lysosomal membranes to nascent phagosomes via lysosomal exocytosis. Thus phagocytic ingestion of large particles activates a phosphoinositide- and Ca2+-dependent exocytosis pathway to provide membranes necessary for pseudopod extension, leading to clearance of senescent and apoptotic cells in vivo.


Subject(s)
Lysosomes/genetics , Phagocytosis/genetics , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism , Aging/genetics , Animals , Calcium/metabolism , Exocytosis/genetics , Gene Expression Regulation , Mice , Particle Size , Phosphatidylinositol Phosphates/metabolism , Transient Receptor Potential Channels/agonists , Transient Receptor Potential Channels/antagonists & inhibitors
11.
J Med Chem ; 54(18): 6215-33, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21819082

ABSTRACT

Spinal muscular atrophy (SMA) is an autosomal recessive disorder affecting the expression or function of survival motor neuron protein (SMN) due to the homozygous deletion or rare point mutations in the survival motor neuron gene 1 (SMN1). The human genome includes a second nearly identical gene called SMN2 that is retained in SMA. SMN2 transcripts undergo alternative splicing with reduced levels of SMN. Up-regulation of SMN2 expression, modification of its splicing, or inhibition of proteolysis of the truncated protein derived from SMN2 have been discussed as potential therapeutic strategies for SMA. In this manuscript, we detail the discovery of a series of arylpiperidines as novel modulators of SMN protein. Systematic hit-to-lead efforts significantly improved potency and efficacy of the series in the primary and orthogonal assays. Structure-property relationships including microsomal stability, cell permeability, and in vivo pharmacokinetics (PK) studies were also investigated. We anticipate that a lead candidate chosen from this series may serve as a useful probe for exploring the therapeutic benefits of SMN protein up-regulation in SMA animal models and a starting point for clinical development.


Subject(s)
Muscular Atrophy, Spinal/metabolism , Piperidines/chemical synthesis , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein/metabolism , Thiadiazoles/chemical synthesis , Thiazoles/chemical synthesis , Alternative Splicing , Caco-2 Cells , Cell Membrane Permeability , Drug Design , Exons , Fibroblasts/drug effects , Fibroblasts/metabolism , Genes, Reporter , HEK293 Cells , Humans , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Male , Microsomes, Liver/metabolism , Muscular Atrophy, Spinal/drug therapy , Piperidines/pharmacokinetics , Piperidines/pharmacology , Promoter Regions, Genetic , Structure-Activity Relationship , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/genetics , Thiadiazoles/pharmacokinetics , Thiadiazoles/pharmacology , Thiazoles/pharmacokinetics , Thiazoles/pharmacology , Transcription, Genetic
12.
Endocrinology ; 151(7): 3454-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20427476

ABSTRACT

Small molecule inverse agonists for the TSH receptor (TSHR) may be used as probes of the role of basal (or agonist-independent or constitutive) signaling and may have therapeutic potential as orally active drugs to inhibit basal signaling in patients with thyroid cancer and in some patients with hyperthyroidism. We describe the first small-molecule ligand [1;2-(3-((2,6-dimethylphenoxy)methyl)-4-methoxyphenyl)-3-(furan-2-ylmethyl)-2,3-dihydroquinazolin-4(1H)-one] that exhibits inverse agonist properties at TSHR. 1 inhibits basal and TSH-stimulated signaling, measured as cAMP production, by TSHRs in HEK-EM 293 cells stably expressing wild-type TSHRs; the antagonism of TSH-mediated signaling is competitive. 1 also inhibits basal signaling by wild-type TSHRs, and four constitutively active mutants of TSHR expressed transiently in HEK-EM 293 cells. 1 was active under more physiologically relevant conditions in primary cultures of human thyrocytes expressing endogenous TSHRs where it inhibited basal levels of mRNA transcripts for thyroglobulin, thyroperoxidase, sodium iodide symporter, and TSHR. These data serve as proof of principle that small, drug-like molecules can inhibit basal signaling by TSHR. We suggest that this small molecule is a lead compound for the development of higher-potency inverse agonists that can be used as probes of TSHR biology with therapeutic potential.


Subject(s)
Receptors, Thyrotropin/agonists , Animals , CHO Cells , Cell Line , Cells, Cultured , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Mutagenesis, Site-Directed , Receptors, Thyrotropin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Thyrotropin/pharmacology
13.
Assay Drug Dev Technol ; 8(3): 367-79, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20230302

ABSTRACT

Activation of G(q) protein-coupled receptors can be monitored by measuring the increase in intracellular calcium with fluorescent dyes. Recent advances in fluorescent kinetic plate readers and liquid-handling technology have made it possible to follow these transient changes in intracellular calcium in a 1,536-well plate format for high-throughput screening (HTS). Here, we have applied the latest generation of fluorescence kinetic plate readers to multiplex the agonist and antagonist screens of a G protein-coupled receptor (GPCR). This multiplexed assay format provides an efficient and cost-effective method for HTS of G(q)-coupled GPCR targets.


Subject(s)
Calcium/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , CHO Cells , Calcium/analysis , Calibration , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Fluorescent Dyes , Indicators and Reagents , Kinetics , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Radioligand Assay , Receptor, Muscarinic M1/drug effects
14.
Proc Natl Acad Sci U S A ; 106(30): 12471-6, 2009 Jul 28.
Article in English | MEDLINE | ID: mdl-19592511

ABSTRACT

Seven-transmembrane-spanning receptors (7TMRs) are prominent drug targets. However, small-molecule ligands for 7-transmembrane-spanning receptors for which the natural ligands are large, heterodimeric glycoprotein hormones, like thyroid-stimulating hormone (TSH; thyrotropin), have only recently been reported, and none are approved for human use. We have used quantitative high-throughput screening to identify a small-molecule TSH receptor (TSHR) agonist that was modified to produce a second agonist with increased potency. We show that these agonists are highly selective for human TSHR versus other glycoprotein hormone receptors and interact with the receptor's serpentine domain. A binding pocket within the transmembrane domain was defined by docking into a TSHR homology model and was supported by site-directed mutagenesis. In primary cultures of human thyrocytes, both TSH and the agonists increase mRNA levels for thyroglobulin, thyroperoxidase, sodium iodide symporter, and deiodinase type 2, and deiodinase type 2 enzyme activity. Moreover, oral administration of the agonist stimulated thyroid function in mice, resulting in increased serum thyroxine and thyroidal radioiodide uptake. Thus, we discovered a small molecule that activates human TSHR in vitro, is orally active in mice, and could be a lead for development of drugs to use in place of recombinant human TSH in patients with thyroid cancer.


Subject(s)
Organic Chemicals/pharmacology , Receptors, Thyrotropin/agonists , Thyroid Gland/drug effects , Acetamides/chemical synthesis , Acetamides/chemistry , Acetamides/pharmacology , Animals , Binding Sites , Cell Line , Cells, Cultured , Cyclic AMP/metabolism , Female , Gene Expression/drug effects , Humans , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Organic Chemicals/chemical synthesis , Organic Chemicals/chemistry , Protein Structure, Tertiary , Quinazolinones/chemical synthesis , Quinazolinones/chemistry , Quinazolinones/pharmacology , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thyroglobulin/genetics , Thyroglobulin/metabolism , Thyroid Gland/cytology , Thyroid Gland/physiology , Thyrotropin/pharmacology , Transfection
15.
Am J Nephrol ; 29(3): 153-63, 2009.
Article in English | MEDLINE | ID: mdl-18753740

ABSTRACT

BACKGROUND: Conditionally immortalized podocytes are valuable research tools but are difficult to efficiently transfect and do not provide graded transgene expression. METHODS: Conditionally immortalized mouse podocyte cell lines were established employing a tetracycline-inducible system. Glomerular cells, isolated from transgenic mice bear- ing two transgenes, NPHS2-reverse tetracycline-controlled transactivator, rtTA (A transgene) and H2-Kb-thermosensitive SV40 T, ts58A (I transgene), were cloned. One clone (AI podocytes) expressing WT1 and synaptopodin was transfected with pBI-EGFP (enhanced green fluorescent protein, G transgene) and separately with ptTS-Neo (transcriptional suppressor, T transgene) to produce stable transformants, AIG podocytes and AIT podocytes. RESULTS: AIG podocytes expressed EGFP at 33 and 37 degrees C after doxycycline treatment, and retained podocin and rtTA mRNA expression and temperature-sensitive growth regulation. AIT podocytes, transiently transfected with luciferase-BI-EGFP (LG transgene), showed reduced background expression of EGFP and luciferase in the absence of doxycycline. In AITLG podocytes, generated by stable transfection of AIT podocytes with the LG transgene, luciferase expression was tightly regulated by doxycycline in a time- and concentration-dependent manner both at 33 and 37 degrees C, although background expression was not entirely eliminated. These podocytes retained temperature-sensitive growth regulation and expression of podocyte differentiation markers. CONCLUSION: Mouse podocytes expressed tetracycline-induced transgenes efficiently while retaining differentiation markers.


Subject(s)
Cell Culture Techniques , Podocytes/metabolism , Protein Synthesis Inhibitors/pharmacology , Tetracycline/pharmacology , Transcriptional Activation/drug effects , Animals , Cell Line , Gene Expression , Mice , Mice, Transgenic , Recombinant Fusion Proteins/metabolism , Transgenes
16.
J Biomol Screen ; 13(2): 120-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18216391

ABSTRACT

The thyroid-stimulating hormone (TSH; thyrotropin) receptor belongs to the glycoprotein hormone receptor subfamily of 7-transmembrane spanning receptors. TSH receptor (TSHR) is expressed mainly in thyroid follicular cells and is activated by TSH, which regulates the growth and function of thyroid follicular cells. Recombinant TSH is used in diagnostic screens for thyroid cancer, especially in patients after thyroid cancer surgery. Currently, no selective small-molecule agonists of the TSHR are available. To screen for novel TSHR agonists, the authors miniaturized a commercially available cell-based cyclic adenosine 3',5' monophosphate (cAMP) assay into a 1536-well plate format. This assay uses an HEK293 cell line stably transfected with the TSHR coupled to a cyclic nucleotide gated ion channel as a biosensor. From a quantitative high-throughput screen of 73,180 compounds in parallel with a parental cell line (without the TSHR), 276 primary active compounds were identified. The activities of the selected active compounds were further confirmed in an orthogonal homogeneous time-resolved fluorescence cAMP-based assay. Forty-nine compounds in several structural classes have been confirmed as the small-molecule TSHR agonists that will serve as a starting point for chemical optimization and studies of thyroid physiology in health and disease.


Subject(s)
Cyclic AMP/analysis , Drug Evaluation, Preclinical/methods , Receptors, Thyrotropin/agonists , Small Molecule Libraries/analysis , Algorithms , Calibration , Cells, Cultured , False Positive Reactions , Humans , Miniaturization , Models, Biological , Quantitative Structure-Activity Relationship , Receptors, Thyrotropin/genetics , Transfection
17.
Curr Chem Genomics ; 1: 70-8, 2008 Jul 11.
Article in English | MEDLINE | ID: mdl-20161830

ABSTRACT

Cell-based functional assays used for compound screening and lead optimization play an important role in drug discovery for G-protein coupled receptors (GPCRs). Cell-based assays can define the role of a compound as an agonist, antagonist or inverse agonist and can provide detailed information about the potency and efficacy of a compound. In addition, cell-based screens can be used to identify allosteric modulators that interact with sites other than the binding site of the endogenous ligand. Intracellular calcium assays which use a fluorescent calcium binding dye (such as Fluo-3, Fluo-4 or Fura-2) have been used in compound screening campaigns to measure the activity of Gq-coupled GPCRs. However, such screening methodologies require a special instrumentation to record the rapid change in intracellular free calcium concentration over time. The radioactive inositol 1,4,5- triphosphate (IP(3)) assay measures (3)H-inositol incorporation and is another traditional assay for the assessment of Gq-coupled GPCR activity, but it is not suitable for screening of large size compound collections because it requires a cell wash step and generates radioactive waste. To avoid these limitations, we have optimized and miniaturized a TR-FRET based IP-One assay that measures inositol monophosphate in a 1536-well plate format. This assay is homogenous, non-radioactive and does not require a kinetic readout. It has been tested with the cell lines expressing M(1) acetylcholine, FFAR1, vasopressin V1b, or Neuropeptide S receptors. The activities of antagonists determined in the IP-One assay correlated well with these measured in the intracellular calcium assay while the correlation of agonist activities might vary from cell line to cell line. This IP-One assay offers an alternative method for high throughput screening of Gq-coupled GPCRs without using costly kinetic plate readers.

SELECTION OF CITATIONS
SEARCH DETAIL
...