Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Int J Mol Sci ; 22(9)2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33922601

ABSTRACT

Many human diseases are the result of abnormal expression or activation of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs). Not surprisingly, more than 30 tyrosine kinase inhibitors (TKIs) are currently in clinical use and provide unique treatment options for many patients. PTPs on the other hand have long been regarded as "undruggable" and only recently have gained increased attention in drug discovery. Striatal-enriched tyrosine phosphatase (STEP) is a neuron-specific PTP that is overactive in Alzheimer's disease (AD) and other neurodegenerative and neuropsychiatric disorders, including Parkinson's disease, schizophrenia, and fragile X syndrome. An emergent model suggests that the increase in STEP activity interferes with synaptic function and contributes to the characteristic cognitive and behavioral deficits present in these diseases. Prior efforts to generate STEP inhibitors with properties that warrant clinical development have largely failed. To identify novel STEP inhibitor scaffolds, we developed a biophysical, label-free high-throughput screening (HTS) platform based on the protein thermal shift (PTS) technology. In contrast to conventional HTS using STEP enzymatic assays, we found the PTS platform highly robust and capable of identifying true hits with confirmed STEP inhibitory activity and selectivity. This new platform promises to greatly advance STEP drug discovery and should be applicable to other PTP targets.


Subject(s)
Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Humans , Molecular Structure
2.
Biochem Pharmacol ; 219: 115937, 2024 01.
Article in English | MEDLINE | ID: mdl-37995979

ABSTRACT

Mitochondrial uridine insertion/deletion RNA editing, catalyzed by a multiprotein complex (editosome), is essential for gene expression in trypanosomes and Leishmania parasites. As this process is absent in the human host, a drug targeting this mechanism promises high selectivity and reduced toxicity. Here, we successfully miniaturized our FRET-based full-round RNA editing assay, which replicates the complete RNA editing process, adapting it into a 1536-well format. Leveraging this assay, we screened over 100,000 compounds against purified editosomes derived from Trypanosoma brucei, identifying seven confirmed primary hits. We sourced and evaluated various analogs to enhance the inhibitory and parasiticidal effects of these primary hits. In combination with secondary assays, our compounds marked inhibition of essential catalytic activities, including the RNA editing ligase and interactions of editosome proteins. Although the primary hits did not exhibit any growth inhibitory effect on parasites, we describe eight analog compounds capable of effectively killing T. brucei and/or Leishmania donovani parasites within a low micromolar concentration. Whether parasite killing is - at least in part - due to inhibition of RNA editing in vivo remains to be assessed. Our findings introduce novel molecular scaffolds with the potential for broad antitrypanosomal effects.


Subject(s)
Trypanosoma brucei brucei , Humans , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/metabolism , High-Throughput Screening Assays , RNA Editing , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA/metabolism
3.
Bioorg Med Chem Lett ; 22(21): 6656-60, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23010269

ABSTRACT

The recently discovered apelin/APJ system has emerged as a critical mediator of cardiovascular homeostasis and is associated with the pathogenesis of cardiovascular disease. A role for apelin/APJ in energy metabolism and gastrointestinal function has also recently emerged. We disclose the discovery and characterization of 4-oxo-6-((pyrimidin-2-ylthio)methyl)-4H-pyran-3-yl 4-nitrobenzoate (ML221), a potent APJ functional antagonist in cell-based assays that is >37-fold selective over the closely related angiotensin II type 1 (AT1) receptor. ML221 was derived from an HTS of the ~330,600 compound MLSMR collection. This antagonist showed no significant binding activity against 29 other GPCRs, except to the κ-opioid and benzodiazepinone receptors (<50/<70%I at 10 µM). The synthetic methodology, development of structure-activity relationship (SAR), and initial in vitro pharmacologic characterization are also presented.


Subject(s)
Drug Discovery , Nitrobenzoates/chemical synthesis , Pyrans/chemical synthesis , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Apelin Receptors , Cardiovascular Agents/chemistry , Cardiovascular Agents/pharmacology , Dose-Response Relationship, Drug , Hepatocytes/drug effects , Inhibitory Concentration 50 , Mice , Molecular Structure , Nitrobenzoates/chemistry , Nitrobenzoates/pharmacology , Protein Binding/drug effects , Pyrans/chemistry , Pyrans/pharmacology , Structure-Activity Relationship
4.
Cancer Res ; 81(6): 1472-1485, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33414172

ABSTRACT

Invadopodia are actin-based proteolytic membrane protrusions required for invasive behavior and tumor growth. In this study, we used our high-content screening assay to identify kinases whose activity affects invadopodia formation. Among the top hits selected for further analysis was TAO3, an STE20-like kinase of the GCK subfamily. TAO3 was overexpressed in many human cancers and regulated invadopodia formation in melanoma, breast, and bladder cancers. Furthermore, TAO3 catalytic activity facilitated melanoma growth in three-dimensional matrices and in vivo. A novel, potent catalytic inhibitor of TAO3 was developed that inhibited invadopodia formation and function as well as tumor cell extravasation and growth. Treatment with this inhibitor demonstrated that TAO3 activity is required for endosomal trafficking of TKS5α, an obligate invadopodia scaffold protein. A phosphoproteomics screen for TAO3 substrates revealed the dynein subunit protein LIC2 as a relevant substrate. Knockdown of LIC2 or expression of a phosphomimetic form promoted invadopodia formation. Thus, TAO3 is a new therapeutic target with a distinct mechanism of action. SIGNIFICANCE: An unbiased screening approach identifies TAO3 as a regulator of invadopodia formation and function, supporting clinical development of this class of target.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Endosomes/metabolism , Neoplasm Invasiveness/pathology , Podosomes/drug effects , Protein Serine-Threonine Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cytoplasmic Dyneins/genetics , Cytoplasmic Dyneins/metabolism , Datasets as Topic , Extracellular Matrix , Female , Gene Expression Profiling , Gene Knockdown Techniques , High-Throughput Screening Assays , Humans , Male , Melanoma/drug therapy , Melanoma/pathology , Mice , Neoplasm Invasiveness/prevention & control , Podosomes/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Time-Lapse Imaging , Xenograft Model Antitumor Assays
5.
Biochem Biophys Res Commun ; 391(1): 1049-55, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19995556

ABSTRACT

Patients with alveolar rhabdomyosarcoma (ARMS) have poorer response to conventional chemotherapy and lower survival rates than those with embryonal RMS (ERMS). To identify compounds that preferentially block the growth of ARMS, we conducted a small-scale screen of 160 kinase inhibitors against the ARMS cell line Rh30 and ERMS cell line RD and identified inhibitors of glycogen synthase kinase 3 (GSK3), including TWS119 as ARMS-selective inhibitors. GSK3 inhibitors inhibited cell proliferation and induced apoptosis more effectively in Rh30 than RD cells. Ectopic expression of fusion protein PAX3-FKHR in RD cells significantly increased their sensitivity to TWS119. Down-regulation of GSK3 by GSK3 inhibitors or siRNA significantly reduced the transcriptional activity of PAX3-FKHR. These results suggest that GSK3 is directly involved in regulating the transcriptional activity of PAX3-FKHR. Also, GSK3 phosphorylated PAX3-FKHR in vitro, suggesting that GSK3 might regulate PAX3-FKHR activity via phosphorylation. These findings support a novel mechanism of PAX3-FKHR regulation by GSK3 and provide a novel strategy to develop GSK inhibitors as anti-ARMS therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Forkhead Transcription Factors/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Paired Box Transcription Factors/metabolism , Protein Kinase Inhibitors/pharmacology , Rhabdomyosarcoma, Alveolar/enzymology , Antineoplastic Agents/isolation & purification , Drug Screening Assays, Antitumor , Forkhead Box Protein O1 , Glycogen Synthase Kinase 3/metabolism , Humans , PAX3 Transcription Factor , Phosphorylation , Protein Kinase Inhibitors/isolation & purification , Pyrimidines/pharmacology , Pyrroles/pharmacology , Rhabdomyosarcoma, Alveolar/pathology
6.
Elife ; 82019 10 22.
Article in English | MEDLINE | ID: mdl-31637999

ABSTRACT

High-content phenotypic screening has become the approach of choice for drug discovery due to its ability to extract drug-specific multi-layered data. In the field of epigenetics, such screening methods have suffered from a lack of tools sensitive to selective epigenetic perturbations. Here we describe a novel approach, Microscopic Imaging of Epigenetic Landscapes (MIEL), which captures the nuclear staining patterns of epigenetic marks and employs machine learning to accurately distinguish between such patterns. We validated the MIEL platform across multiple cells lines and using dose-response curves, to insure the fidelity and robustness of this approach for high content high throughput drug discovery. Focusing on noncytotoxic glioblastoma treatments, we demonstrated that MIEL can identify and classify epigenetically active drugs. Furthermore, we show MIEL was able to accurately rank candidate drugs by their ability to produce desired epigenetic alterations consistent with increased sensitivity to chemotherapeutic agents or with induction of glioblastoma differentiation.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Drug Discovery/methods , Epigenesis, Genetic/drug effects , High-Throughput Screening Assays , Histones/genetics , Neoplasm Proteins/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/metabolism , Dose-Response Relationship, Drug , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Histones/metabolism , Humans , Image Processing, Computer-Assisted/statistics & numerical data , Machine Learning , Microscopy, Fluorescence , Neoplasm Proteins/metabolism
7.
Cancer Lett ; 449: 145-162, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30771432

ABSTRACT

Inhibition of ubiquitin ligases with small molecule remains a very challenging task, given the lack of catalytic activity of the target and the requirement of disruption of its interactions with other proteins. Siah1/2, which are E3 ubiquitin ligases, are implicated in melanoma and prostate cancer and represent high-value drug targets. We utilized three independent screening approaches in our efforts to identify small-molecule Siah1/2 inhibitors: Affinity Selection-Mass Spectrometry, a protein thermal shift-based assay and an in silico based screen. Inhibitors were assessed for their effect on viability of melanoma and prostate cancer cultures, colony formation, prolyl-hydroxylase-HIF1α signaling, expression of selected Siah2-related transcripts, and Siah2 ubiquitin ligase activity. Several analogs were further characterized, demonstrating improved efficacy. Combination of the top hits identified in the different assays demonstrated an additive effect, pointing to complementing mechanisms that underlie each of these Siah1/2 inhibitors.


Subject(s)
Melanoma/drug therapy , Nuclear Proteins/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Small Molecule Libraries/administration & dosage , Ubiquitin-Protein Ligases/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Computer Simulation , Down-Regulation , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mass Spectrometry , Melanoma/genetics , Mice , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Small Molecule Libraries/isolation & purification , Small Molecule Libraries/pharmacology , Ubiquitin-Protein Ligases/genetics , Xenograft Model Antitumor Assays
8.
J Biomol Screen ; 20(10): 1232-45, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26265713

ABSTRACT

Endoplasmic reticulum (ER) stress activates three distinct signal transducers on the ER membrane. Inositol-requiring protein 1 (IRE1), the most conserved signal transducer, plays a key role in ER stress-mediated signaling. During ER stress, IRE1 initiates two discrete signaling cascades: the "adaptive" signaling cascade mediated by the XBP1 pathway and the "alarm" signaling cascade mediated by stress-activated protein kinase pathways. Fine-tuning of the balance between the adaptive and alarm signals contributes significantly to cellular fate under ER stress. Thus, we propose that the design of high-throughput screening (HTS) assays to selectively monitor IRE1 mediated-signaling would be desirable for drug discovery. To this end, we report the generation of stable human neural cell lines and development of cell-based HTS luciferase (Luc) reporter gene assays for the identification of pathway-specific chemical modulators of IRE1. We implemented a cell-based Luc assay using a chimeric CHOP-Gal4 transcription factor in 384-well format for monitoring IRE1 kinase-mediated p38MAPK activation and an unfolded response pathway element (URPE)-Luc cell-based assay in 1536-well format for monitoring IRE1's RNase-mediated activation of XBP1. Chemical library screening was successfully conducted with both the CHOP/Gal4-Luc cells and UPRE-Luc engineered cells. The studies demonstrate the feasibility of using these HTS assays for discovery of pathway-selective modulators of IRE1.


Subject(s)
Endoribonucleases/antagonists & inhibitors , High-Throughput Screening Assays , Protein Serine-Threonine Kinases/antagonists & inhibitors , Small Molecule Libraries , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Stress , Endoribonucleases/physiology , Enzyme Activation , Genes, Reporter , HeLa Cells , Humans , Luciferases/analysis , Luciferases/genetics , MAP Kinase Signaling System , Neurons , Protein Serine-Threonine Kinases/physiology , Regulatory Factor X Transcription Factors , Thapsigargin/metabolism , Transcription Factors/metabolism , X-Box Binding Protein 1
9.
J Biomol Screen ; 19(8): 1201-11, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24870016

ABSTRACT

Antiapoptotic Bcl-2 family proteins are validated cancer targets composed of six related proteins. From a drug discovery perspective, these are challenging targets that exert their cellular functions through protein-protein interactions (PPIs). Although several isoform-selective inhibitors have been developed using structure-based design or high-throughput screening (HTS) of synthetic chemical libraries, no large-scale screen of natural product collections has been reported. A competitive displacement fluorescence polarization (FP) screen of nearly 150,000 natural product extracts was conducted against all six antiapoptotic Bcl-2 family proteins using fluorochrome-conjugated peptide ligands that mimic functionally relevant PPIs. The screens were conducted in 1536-well format and displayed satisfactory overall HTS statistics, with Z'-factor values ranging from 0.72 to 0.83 and a hit confirmation rate between 16% and 64%. Confirmed active extracts were orthogonally tested in a luminescent assay for caspase-3/7 activation in tumor cells. Active extracts were resupplied, and effort toward the isolation of pure active components was initiated through iterative bioassay-guided fractionation. Several previously described altertoxins were isolated from a microbial source, and the pure compounds demonstrate activity in both Bcl-2 FP and caspase cellular assays. The studies demonstrate the feasibility of ultra-high-throughput screening using natural product sources and highlight some of the challenges associated with this approach.


Subject(s)
Biological Products/chemistry , High-Throughput Screening Assays/methods , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Caco-2 Cells , Caspase 3/metabolism , Caspase 7/metabolism , Drug Screening Assays, Antitumor/methods , Fluorescence Polarization/methods , High-Throughput Screening Assays/instrumentation , Humans , Miniaturization , Molecular Targeted Therapy/methods , Mycotoxins/isolation & purification , Mycotoxins/pharmacology , Solid Phase Extraction , bcl-X Protein/antagonists & inhibitors
10.
PLoS One ; 8(3): e59045, 2013.
Article in English | MEDLINE | ID: mdl-23527084

ABSTRACT

Bone Morphogenetic Proteins (BMPs) are morphogens that play a major role in regulating development and homeostasis. Although BMPs are used for the treatment of bone and kidney disorders, their clinical use is limited due to the supra-physiological doses required for therapeutic efficacy causing severe side effects. Because recombinant BMPs are expensive to produce, small molecule activators of BMP signaling would be a cost-effective alternative with the added benefit of being potentially more easily deliverable. Here, we report our efforts to identify small molecule activators of BMP signaling. We have developed a cell-based assay to monitor BMP signaling by stably transfecting a BMP-responsive human cervical carcinoma cell line (C33A) with a reporter construct in which the expression of luciferase is driven by a multimerized BMP-responsive element from the Id1 promoter. A BMP-responsive clone C33A-2D2 was used to screen a bioactive library containing ∼5,600 small molecules. We identified four small molecules of the family of flavonoids all of which induced luciferase activity in a dose-dependent manner and ventralized zebrafish embryos. Two of the identified compounds induced Smad1, 5 phosphorylation (P-Smad), Id1 and Id2 expression in a dose-dependent manner demonstrating that our assays identified small molecule activators of BMP signaling.


Subject(s)
Bone Morphogenetic Proteins/agonists , Bone Morphogenetic Proteins/metabolism , Drug Discovery , Signal Transduction/drug effects , Small Molecule Libraries , Animals , Cell Line, Tumor , Chalcone/pharmacology , Embryo, Nonmammalian/drug effects , Embryonic Development/drug effects , Flavones/pharmacology , Genes, Reporter , High-Throughput Screening Assays , Humans , Mice , Mice, Knockout , Myoblasts/cytology , Myoblasts/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Zebrafish
11.
Chem Biol ; 17(8): 892-902, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20797618

ABSTRACT

Pantothenate kinase (PanK) catalyzes the rate-controlling step in coenzyme A (CoA) biosynthesis. PanK3 is stringently regulated by acetyl-CoA and uses an ordered kinetic mechanism with ATP as the leading substrate. Biochemical analysis of site-directed mutants indicates that pantothenate binds in a tunnel adjacent to the active site that is occupied by the pantothenate moiety of the acetyl-CoA regulator in the PanK3acetyl-CoA binary complex. A high-throughput screen for PanK3 inhibitors and activators was applied to a bioactive compound library. Thiazolidinediones, sulfonylureas and steroids were inhibitors, and fatty acyl-amides and tamoxifen were activators. The PanK3 activators and inhibitors either stimulated or repressed CoA biosynthesis in HepG2/C3A cells. The flexible allosteric acetyl-CoA regulatory domain of PanK3 also binds the substrates, pantothenate and pantetheine, and small molecule inhibitors and activators to modulate PanK3 activity.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Small Molecule Libraries/metabolism , Acetyl Coenzyme A/metabolism , Allosteric Regulation/drug effects , Binding Sites , Coenzyme A/biosynthesis , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Hep G2 Cells , High-Throughput Screening Assays , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Binding , Protein Structure, Tertiary , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Substrate Specificity , Sulfonylurea Compounds/metabolism , Thiazolidinediones/metabolism
12.
Cancer Lett ; 284(2): 157-64, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19442434

ABSTRACT

Patients with alveolar rhabdomyosarcoma (ARMS) have poorer response to conventional chemotherapy and lower survival rates than those with embryonal RMS (ERMS). By high-throughput screening, we identified camptothecin as an ARMS-selective inhibitor. Camptothecin more efficiently inhibited proliferation and induced apoptosis in Rh30 (ARMS) than RD (ERMS) cells. Ectopic expression of the PAX3-FKHR (PF) fusion protein in RD cells significantly increased sensitivity, whereas siRNA knockdown of PF decreased sensitivity of Rh30 cells to camptothecin. The sensitization required a transcriptionally active PF, and camptothecin downregulated levels of PF protein. These findings suggest that it is feasible to develop agents that preferentially block the growth of ARMS.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Forkhead Transcription Factors/physiology , Rhabdomyosarcoma, Alveolar/metabolism , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Tumor/drug effects , Drug Resistance, Neoplasm/genetics , Forkhead Box Protein O1 , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Humans , RNA Interference , RNA, Small Interfering/pharmacology , Rhabdomyosarcoma, Alveolar/drug therapy , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/pathology , Transcription, Genetic
13.
J Biol Chem ; 283(45): 30650-7, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18784074

ABSTRACT

The human pregnane X receptor (hPXR) regulates the expression of critical drug metabolism enzymes. One of such enzymes, cytochrome P450 3A4 (CYP3A4), plays critical roles in drug metabolism in hepatocytes that are either quiescent or passing through the cell cycle. It has been well established that the expression of P450, such as CYP3A4, is markedly reduced during liver development or regeneration. Numerous studies have implicated cellular signaling pathways in modulating the functions of nuclear receptors, including hPXR. Here we report that inhibition of cyclin-dependent kinases (Cdks) by kenpaullone and roscovitine (two small molecule inhibitors of Cdks that we identified in a screen for compounds that activate hPXR) leads to activation of hPXR-mediated CYP3A4 gene expression in HepG2 human liver carcinoma cells. Consistent with this finding, activation of Cdk2 attenuates the activation of CYP3A4 gene expression. In vitro kinase assays revealed that Cdk2 directly phosphorylates hPXR. A phosphomimetic mutation of a putative Cdk phosphorylation site, Ser(350), significantly impairs the function of hPXR, whereas a phosphorylation-deficient mutation confers resistance to Cdk2. Using HepG2 that has been stably transfected with hPXR and the CYP3A4-luciferase reporter, enriched in different phases of the cell cycle, we found that hPXR-mediated CYP3A4 expression is greatly reduced in the S phase. Our results indicate for the first time that Cdk2 negatively regulates the activity of hPXR, and suggest an important role for Cdk2 in regulating hPXR activity and CYP3A4 expression in hepatocytes passing through the cell cycle, such as those in fetal or regenerating adult liver.


Subject(s)
Cell Cycle , Cyclin-Dependent Kinase 2/metabolism , Cytochrome P-450 CYP3A/biosynthesis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Liver Neoplasms/enzymology , Receptors, Steroid/metabolism , Benzazepines/pharmacology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cytochrome P-450 CYP3A/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Hepatocytes/enzymology , Humans , Indoles/pharmacology , Liver Neoplasms/genetics , Mutation , Phosphorylation/drug effects , Phosphorylation/genetics , Pregnane X Receptor , Regeneration/drug effects , Regeneration/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
14.
Mol Pharmacol ; 62(3): 747-55, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12181453

ABSTRACT

Constitutive activity of wild-type and mutant forms of human beta(1)- and beta(2)-adrenoceptors was measured by guanosine 5'-O-(3-[(35)S]thio)triphosphate ([(35)S]GTPgammaS) binding assays using fusion proteins between these receptors and G(s)alpha. Constitutive activity of the beta(1)-adrenoceptor is enhanced by mutation of Leu(322). The ability of ligands to suppress receptor instability and produce up-regulation is often associated with constitutively active mutants. Leu(322)Lysbeta(1)-adrenoceptor, but not wild type, was up-regulated by exposure to the beta(1)-adrenoceptor selective blocker betaxolol. More extensive sequence alterations of the beta(1)-adrenoceptor were generated to mimic the initially described constitutively active mutant (CAM) of the beta(2)-adrenoceptor that is up-regulated strongly by betaxolol. Substitution of amino acids 316 to 324 of the beta(1)-adrenoceptor with the equivalent alpha(1b)-adrenoceptor sequence did not result in up-regulation by betaxolol. However, these forms of both beta(1)- and beta(2)-adrenoceptors displayed substantial and equivalent constitutive activity. The addition of the Leu(322)Lys mutation into the alpha(1b)-adrenoceptor substituted beta(1)-adrenoceptor to produce the CAMKbeta(1)-adrenoceptor allowed substantially greater levels of up-regulation by betaxolol without enhancement of constitutive [(35)S]GTPgammaS binding. Arg(156)Alabeta(1)-adrenoceptor was up-regulated strongly by betaxolol but displayed lower constitutive activity than did other mutants. Binding of [(35)S]GTPgammaS binding to all the fusion proteins was increased substantially by isoprenaline. Despite the ability of betaxolol to cause up-regulation of many mutants, only for the CAMbeta(2)-adrenoceptor-G(s)alpha and CAMKbeta(1)-adrenoceptor-G(s)alpha fusion proteins was the basal binding of [(35)S]GTPgammaS decreased by betaxolol. Clear resolution between receptor constitutive activity and ligand suppression of receptor instability can be obtained for mutant beta-adrenoceptors, and potential inverse agonists do not function equally at phenotypically apparently equivalent CAM receptors.


Subject(s)
Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Receptors, Adrenergic, beta-1/metabolism , Adrenergic beta-Antagonists/pharmacology , Betaxolol/pharmacology , GTP-Binding Protein alpha Subunits, Gs/metabolism , Guanine Nucleotides/metabolism , Humans , Mutation , Receptors, Adrenergic, beta-1/genetics , Sulfur Radioisotopes , Transfection , Up-Regulation
15.
J Biol Chem ; 278(32): 30248-60, 2003 Aug 08.
Article in English | MEDLINE | ID: mdl-12750375

ABSTRACT

The M3 muscarinic receptor is a prototypical member of the class I family of G protein-coupled receptors (GPCRs). To facilitate studies on the structural mechanisms governing M3 receptor activation, we generated an M3 receptor-expressing yeast strain (Saccharomyces cerevisiae) that requires agonist-dependent M3 receptor activation for cell growth. By using receptor random mutagenesis followed by a genetic screen in yeast, we initially identified a point mutation at the cytoplasmic end of transmembrane domain (TM) VI (Q490L) that led to robust agonist-independent M3 receptor signaling in both yeast and mammalian cells. To explore further the molecular mechanisms by which point mutations can render GPCRs constitutively active, we subjected a region of the Q490L mutant M3 receptor that included TM V-VII to random mutagenesis. We then applied a yeast genetic screen to identify second-site mutations that could suppress the activating effects of the Q490L mutation and restore wild-type receptor-like function to the Q490L mutant receptor. This analysis led to the identification of 12 point mutations that allowed the Q490L mutant receptor to function in a fashion similar to the wild-type receptor. These amino acid substitutions mapped to two distinct regions of the M3 receptor, the exofacial segments of TM V and VI and the cytoplasmic ends of TM V-VII. Strikingly, in the absence of the activating Q490L mutation, all recovered point mutations severely reduced the efficiency of receptor/G protein coupling, indicating that the targeted residues play important roles in receptor activation and/or receptor/G protein coupling. This strategy should be generally applicable to identify sites in GPCRs that are critically involved in receptor function.


Subject(s)
GTP-Binding Proteins/chemistry , Receptors, Muscarinic/chemistry , Receptors, Muscarinic/genetics , Amino Acid Sequence , Animals , Blotting, Western , COS Cells , Carbachol/pharmacology , Cattle , Cell Division , Cell Membrane/metabolism , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Kinetics , Ligands , Models, Molecular , Molecular Sequence Data , Mutagenesis , Mutagenesis, Site-Directed , Mutation , Plasmids/metabolism , Point Mutation , Polymerase Chain Reaction , Promoter Regions, Genetic , Protein Structure, Tertiary , Rats , Receptor, Muscarinic M3 , Rhodopsin/chemistry , Saccharomyces cerevisiae/metabolism , Signal Transduction
16.
Mol Pharmacol ; 64(6): 1474-84, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14645678

ABSTRACT

The rat muscarinic acetylcholine receptor subtype 3 was modified by swapping the third intracellular loop with the corresponding region of a constitutively active mutant human beta2-adrenergic receptor and attaching Renilla reniformis luciferase to its C terminus. The chimeric fusion receptor displayed constitutive Gq- and Gs-coupled activity as demonstrated in nuclear factor of activated T cell and cAMP response element reporter gene assays. The chimeric receptor displayed a pharmacological binding profile comparable with that of the wild-type receptor for agonists, antagonists, and inverse agonists but showed a large decrease in expression in both human embryonic kidney 293 and COS-7 cells. Long-term treatment of cells expressing the chimeric receptor with agonists, antagonists, and inverse agonists resulted in a concentration-dependent up-regulation in the steady-state levels that was not observed for the wild-type receptor. The EC50 of neutral antagonists and inverse agonists was significantly correlated to their binding affinities at the wild-type receptor, whereas agonists demonstrated greater EC50 values for the chimeric receptor. To validate the approach as a means of discovering novel receptor modulators, a cell-based, high-throughput screening assay was developed and used to screen a small molecule compound collection against the chimeric fusion receptor. Several novel hits were identified and confirmed by ligand binding assay and functional assays using the wild-type rat muscarinic acetylcholine receptor subtype 3.


Subject(s)
Receptor, Muscarinic M3/biosynthesis , Receptor, Muscarinic M3/genetics , Receptors, G-Protein-Coupled/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemical synthesis , Sulfotransferases/genetics , Up-Regulation/physiology , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Dose-Response Relationship, Drug , Humans , Ligands , Molecular Sequence Data , Muscarinic Agonists/metabolism , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/metabolism , Muscarinic Antagonists/pharmacology , Rats , Receptor, Muscarinic M3/chemistry , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Recombinant Fusion Proteins/genetics , Up-Regulation/drug effects
17.
Rouxs Arch Dev Biol ; 204(5): 344-349, 1995 May.
Article in English | MEDLINE | ID: mdl-28306129

ABSTRACT

Protein-carbohydrate interactions are supposed to play key roles in the mechanisms of cell adhesion, biosignalling and intracellular routing, warranting the analysis of the developmental course of expression of epitopes of this system. Thus, a panel of carrier-immobilized carbohydrate ligands was used as probes, namely lactose,N-acetylgalactosamine,N-acetylglucosamine, mannose, fucose and maltose. Additionally, an antibody to an endogenous ß-galactoside-binding lectin (anti-galectin-1), the biotinylated lectin and two further human lectins, namely the macrophage migration inhibitory factor-binding sarcolectin and serum amyloid P component (SAP) that displays selectivity for sulphated sugars and mannose-6-phosphate, were included. They enabled us to assess the extent of the presence of respective binding sites in fixed sections from human lungs (pulmonary epithelial cells), livers (hepatocytes) and hearts (myocard cells) of 10-50 weeks gestation. Invariably, specific binding was detected in the three organ types, at least in certain stages. In most of the cases, the intensity of staining exhibited developmental regulation. The apparent patterns reveal similarities between the different cell types, as seen with immobilizedN-acetylglucosamine as well as with labelled galectin-1 and sarcolectin. However, drastic differences among such patterns with nearly opposite developmental courses do also occur, as detected for carrier-attached mannose and maltose residues. These results point to a potential importance for the detected glycohistochemical features in human development and substantiate the possibility of differential regulation of the presence of binding sites for distinct sugars within a certain organ and between the individual cell types of the monitored organs.

SELECTION OF CITATIONS
SEARCH DETAIL