Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Med Chem ; 64(15): 11527-11542, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34260228

ABSTRACT

The orphan G-protein-coupled receptor GPR139 is highly expressed in the habenula, a small brain nucleus that has been linked to depression, schizophrenia (SCZ), and substance-use disorder. High-throughput screening and a medicinal chemistry structure-activity relationship strategy identified a novel series of potent and selective benzotriazinone-based GPR139 agonists. Herein, we describe the chemistry optimization that led to the discovery and validation of multiple potent and selective in vivo GPR139 agonist tool compounds, including our clinical candidate TAK-041, also known as NBI-1065846 (compound 56). The pharmacological characterization of these GPR139 agonists in vivo demonstrated GPR139-agonist-dependent modulation of habenula cell activity and revealed consistent in vivo efficacy to rescue social interaction deficits in the BALB/c mouse strain. The clinical GPR139 agonist TAK-041 is being explored as a novel drug to treat negative symptoms in SCZ.


Subject(s)
Drug Discovery , Nerve Tissue Proteins/agonists , Receptors, G-Protein-Coupled/agonists , Schizophrenia/drug therapy , Animals , Cell Line , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Structure , Nerve Tissue Proteins/deficiency , Receptors, G-Protein-Coupled/deficiency , Structure-Activity Relationship
2.
Bioorg Med Chem Lett ; 27(4): 1099-1104, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28082036

ABSTRACT

Axl has been a target of interest in the oncology field for several years based on its role in various oncogenic processes. To date, no wild-type Axl crystal structure has been reported. Herein, we describe the structure-based optimization of a novel chemotype of Axl inhibitors, 1H-imidazole-2-carboxamide, using a mutated kinase homolog, Mer(I650M), as a crystallographic surrogate. Iterative optimization of the initial lead compound (1) led to compound (21), a selective and potent inhibitor of wild-type Axl. Compound (21) will serve as a useful compound for further in vivo studies.


Subject(s)
Imidazoles/chemistry , Imidazoles/pharmacology , Mutation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Crystallography, X-Ray , Molecular Structure , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
3.
Assay Drug Dev Technol ; 4(5): 545-53, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17115925

ABSTRACT

In this paper we have explored the utility of the real-time cell electronic sensing (RTCES, ACEA Biosciences Inc., San Diego, CA) system for monitoring the quality of live cells in cell-based assays as well as for assay development. We have demonstrated that each cell type displays unique growth kinetic profiles that provide a quantitative account of cell behavior and can be used as a diagnostic tool for cellular quality control. The utility of the specific signature patterns was shown by demonstrating the significant differences in primary cell behavior depending on the supplier. In addition, the RT-CES system was able to differentiate cell behavior depending on the passage stage of the cells. The utility of the RT-CES system as an assay development tool was demonstrated in cytotoxicity assays. The RT-CES system not only provides information regarding the potency of cytotoxic compounds, but in addition relates potency to the rate of the response for each concentration of the compound tested, which is important for understanding the mechanism of compound action. Moreover, real-time display of cytotoxicity data by the RT-CES system allows for calculation of real-time 50% inhibitory concentration (IC50) values or determination of optimal IC(50) value. In summary, the RT-CES system provides high content and information-rich data that are beyond the scope of single-point assays.


Subject(s)
Biological Assay/methods , Biosensing Techniques/methods , Cell Culture Techniques/methods , Cytotoxins/toxicity , Endothelial Cells/drug effects , Endothelial Cells/physiology , Toxicity Tests/methods , Biological Assay/instrumentation , Biosensing Techniques/instrumentation , Cell Culture Techniques/instrumentation , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Computer Systems , Electronics , Endothelial Cells/cytology , Humans , Quality Control , Toxicity Tests/instrumentation
4.
Assay Drug Dev Technol ; 4(5): 597-607, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17115930

ABSTRACT

Cell-based assays have become an integral part of the preclinical drug development process. Recently, noninvasive label-free cell-based assay technologies have taken center stage, offering important and distinct advantages over and in addition to traditional label-based endpoint assays. Dynamic monitoring of live cells, the preclusion of label, and kinetics are some of the fundamental features of cell-based label-free technologies. In this article we will discuss the real-time cell electronic sensing (RT-CES, ACEA Biosciences Inc., San Diego, CA) system and some of its key applications for cell-based assays such as cell proliferation and cytotoxicity, functional assays for receptor-ligand analysis, cell adhesion and spreading assays, dynamic monitoring of endothelial barrier function, and dynamic monitoring of cell migration and invasion. Also, where appropriate we will briefly discuss other label-free technologies in an application-specific manner.


Subject(s)
Biological Assay/instrumentation , Biosensing Techniques/instrumentation , Drug Design , Electrochemistry/instrumentation , Pharmacology/instrumentation , Technology, Pharmaceutical/instrumentation , Biological Assay/methods , Biosensing Techniques/methods , Computer Systems , Electrochemistry/methods , Pharmacology/methods , Staining and Labeling , Technology Assessment, Biomedical , Technology, Pharmaceutical/methods
5.
J Biomol Screen ; 11(6): 634-43, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16858006

ABSTRACT

Kinases are the 2nd largest group of therapeutic targets in the human genome. In this article, a label-free and real-time cell-based receptor tyrosine kinase (RTK) assay that addresses limitation of existing kinase assays and can be used for high-throughput screening and lead optimization studies was validated and characterized. Using impedance, growth factor-induced morphological changes were quantitatively assessed in real time and used as a measure of RTK activity. COS7 cells treated with epidermal growth factor (EGF) and insulin results in a rapid increase in cell impedance. Assessment of these growth factor-induced morphological changes and levels of receptor autophosphorylation using fluorescent microscopy and enzyme-linked immunosorbent assay, respectively, demonstrates that these changes correlate with changes in impedance. This assay was used to screen, identify, and characterize a potent EGF receptor inhibitor from a compound library. This report describes an assay that is simple in that it does not require intensive optimization or special reagents such as peptides, antibodies, or probes. More important, because the assay is cell based, the studies are done in a physiologically relevant environment, allowing for concurrent assessment of a compound's solubility, stability, membrane permeability, cytotoxicity, and off-target interaction effects.


Subject(s)
Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , COS Cells , Chlorocebus aethiops , Electric Impedance , Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/metabolism
6.
Arthritis Rheum ; 54(2): 524-32, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16447229

ABSTRACT

OBJECTIVE: To perform a large-scale association analysis of single-nucleotide polymorphisms (SNPs) in patients with radiographically defined osteoarthritis (OA) of the knee. METHODS: We examined >25,000 SNPs located within approximately 14,000 genes for associations with radiographically defined knee OA, using polymerase chain reaction and MassExtend amplification techniques. Allele frequencies were estimated initially in DNA pools from 335 female patients with knee OA and 335 asymptomatic and radiographically negative female control subjects. All were of northern European ancestry. Significant allele frequency differences were validated by genotyping of individual DNA samples. Confirmed significant findings were verified in 2 additional case-control samples from the UK (443 cases and 303 controls) and Newfoundland (346 cases and 264 controls). Chondrosarcoma cell lines were used to test for potential differences in gene expression. RESULTS: The marker most strongly associated with the risk of knee OA was rs912428, a C/T polymorphism in intron 1 of LRCH1, a gene on chromosome 13q14 that encodes a novel protein of as-yet-unknown function. The frequency of the T allele compared with controls was consistently increased by 40% across all 3 case-control groups. Additional subanalyses in case-control samples with hip OA and hand OA suggested similar trends, but did not reach statistical significance. Association fine-mapping using 10 additional SNPs in LRCH1 confirmed intron 1 as the region of highest association but failed to reveal variations with significance stronger than the marker SNP, as did the haplotype analysis. LRCH1 was not up-regulated or overexpressed in chondrosarcoma cell lines exposed to inflammatory stimuli, suggesting a possible structural role. CONCLUSION: A genetic variant in LRCH1 was consistently associated with knee OA in 3 samples from 2 populations. Our results also suggest that the same association with OA may exist at other sites. Additional genetic and experimental work is needed to elucidate the precise mechanism by which the LRCH1 gene influences OA risk.


Subject(s)
Genetic Predisposition to Disease , Genetic Variation , Genome, Human , Microfilament Proteins/genetics , Osteoarthritis, Knee/genetics , Polymorphism, Single Nucleotide/genetics , Aged , Case-Control Studies , Chondrocytes/metabolism , Chondrosarcoma/metabolism , Chromosome Mapping , Female , Gene Frequency , Genetic Markers , Humans , Male , Middle Aged , Osteoarthritis, Knee/diagnostic imaging , Radiography , Tumor Cells, Cultured
7.
Anal Chem ; 78(1): 35-43, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16383308

ABSTRACT

G protein-coupled receptors (GPCRs) constitute important targets for drug discovery against a wide range of ailments including cancer, inflammatory, and cardiovascular diseases. Efforts are underway to screen selective modulators of GPCRs and also to deorphanize GPCRs with unidentified natural ligands. Most GPCR-based cellular screens depend on labeling or recombinant expression of receptor or reporter proteins, which may not capture the true physiology or pharmacology of the GPCRs. In this paper, we describe a noninvasive and label-free assay for GPCRs that can be used with both engineered and nonengineered cell lines. The assay is based on using cell-electrode impedance to measure minute changes in cellular morphology as a result of ligand-dependent GPCR activation. We have used this technology to assay the functional activation of GPCRs coupled to different signaling pathways and have compared it to standard assays. We have used pharmacological modulators of GPCR signaling pathways to demonstrate the specificity of impedance-based measurements. Our data indicate that cell-electrode impedance measurements offer a convenient, sensitive, and quantitative method for assessing GPCR function. Moreover, the noninvasive nature of the readout offers the added advantage of performing multiple treatments in the same well to study events such as desensitization and receptor cross-talk.


Subject(s)
Electronics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Blotting, Western , CHO Cells/metabolism , CHO Cells/ultrastructure , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Dopamine/metabolism , Electric Impedance , HeLa Cells/metabolism , HeLa Cells/ultrastructure , Histamine/metabolism , Humans , Immunoprecipitation , Inositol Phosphates/metabolism , Kinetics , Ligands , Microscopy, Fluorescence , Receptors, Dopamine/genetics , Receptors, Dopamine/metabolism , Receptors, Histamine/genetics , Receptors, Histamine/metabolism , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , Spectrum Analysis , Vasopressins/metabolism
8.
J Biomol Screen ; 10(8): 795-805, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16234347

ABSTRACT

Cellular interaction with and adhesion on different biological surfaces is a dynamic and integrated process requiring the participation of specialized cell surface receptors, structural proteins, signaling proteins, and the cellular cytoskeleton. In this report, the authors describe a label-free and real-time method for measuring and monitoring cell adhesion on special microplates integrated with electronic cell sensor arrays. These plates were used in conjunction with the real-time cell electronic sensing (RT-CES) system to dynamically and quantitatively monitor the specific interaction of fibroblasts with extracellular matrix (ECM) proteins and compared with standard adhesion techniques. Cell adhesion on ECM-coated cell sensor arrays is dependent on the concentration of ECM proteins coated and is inhibited by agents that disrupt the interaction of ECM with cell surface receptors. Furthermore, the authors demonstrate that the integrity of the actin cytoskeleton is required for productive cell adhesion and spreading on ECM-coated microelectronic sensors. Confirming earlier results, it is shown that interfering with Src expression or activity, via siRNA or small molecule, results in the disruption of adhesion and spreading of Bx PC3 cells. The results indicate that the RT-CES system offers a convenient and quantitative means of assessing the kinetics of cell adhesion in a high-throughput manner.


Subject(s)
Biosensing Techniques/instrumentation , Cell Adhesion , Microelectrodes , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Line , Electrochemistry , Extracellular Matrix Proteins/metabolism , Fibroblasts , Fluorescent Antibody Technique , RNA, Small Interfering/genetics , Receptors, Cell Surface/metabolism , Transfection , src-Family Kinases/antagonists & inhibitors
9.
Mol Cancer Ther ; 4(3): 361-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15767545

ABSTRACT

A genome-wide case-control association study done in our laboratory has identified a single nucleotide polymorphism located in RAD21 as being significantly associated with breast cancer susceptibility. RAD21 is believed to function in sister chromatid alignment as part of the cohesin complex and also in double-strand break (DSB) repair. Following our initial finding, expression studies revealed a 1.25- to 2.5-fold increased expression of this gene in several human breast cancer cell lines as compared with normal breast tissue. To determine whether suppression of RAD21 expression influences cellular proliferation, RNA interference technology was used in breast cancer cell lines MCF-7 and T-47D. Proliferation of cells treated with RAD21-specific small inhibitory RNA (siRNA) was significantly reduced as compared with mock-transfected cells and cells transfected with a control siRNA (Lamin A/C). This inhibition of proliferation correlated with a significant reduction in the expression of RAD21 mRNA and with an increased level of apoptosis. Moreover, MCF-7 cell sensitivity to two DNA-damaging chemotherapeutic agents, etoposide and bleomycin, was increased after inhibition of RAD21 expression with a dose reduction factor 50 (DRF50) of 1.42 and 3.71, respectively. At the highest concentrations of etoposide and bleomycin administered, cells transfected with a single siRNA duplex targeted against RAD21 showed 57% and 60% survival as compared with control cells, respectively. Based on these findings, we conclude that RAD21 is a novel target for developing cancer therapeutics that can potentially enhance the antitumor activity of chemotherapeutic agents acting via induction of DNA damage.


Subject(s)
Bleomycin/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Etoposide/pharmacology , Gene Expression Regulation, Neoplastic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Breast/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Cell Survival , DNA Damage , DNA-Binding Proteins , Dose-Response Relationship, Drug , Drug Synergism , Genetic Predisposition to Disease , Genome , Humans , Neoplasms/metabolism , Polymorphism, Single Nucleotide , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL