Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cell ; 168(6): 1041-1052.e18, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28283060

ABSTRACT

Most secreted growth factors and cytokines are functionally pleiotropic because their receptors are expressed on diverse cell types. While important for normal mammalian physiology, pleiotropy limits the efficacy of cytokines and growth factors as therapeutics. Stem cell factor (SCF) is a growth factor that acts through the c-Kit receptor tyrosine kinase to elicit hematopoietic progenitor expansion but can be toxic when administered in vivo because it concurrently activates mast cells. We engineered a mechanism-based SCF partial agonist that impaired c-Kit dimerization, truncating downstream signaling amplitude. This SCF variant elicited biased activation of hematopoietic progenitors over mast cells in vitro and in vivo. Mouse models of SCF-mediated anaphylaxis, radioprotection, and hematopoietic expansion revealed that this SCF partial agonist retained therapeutic efficacy while exhibiting virtually no anaphylactic off-target effects. The approach of biasing cell activation by tuning signaling thresholds and outputs has applications to many dimeric receptor-ligand systems.


Subject(s)
Anaphylaxis/metabolism , Hematopoietic Stem Cells/immunology , Mast Cells/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction , Stem Cell Factor/metabolism , Anaphylaxis/immunology , Animals , Dimerization , Humans , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Engineering , Proto-Oncogene Proteins c-kit/agonists , Proto-Oncogene Proteins c-kit/chemistry , Stem Cell Factor/chemistry , Stem Cell Factor/genetics
2.
Cell ; 160(6): 1196-208, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25728669

ABSTRACT

Most cell-surface receptors for cytokines and growth factors signal as dimers, but it is unclear whether remodeling receptor dimer topology is a viable strategy to "tune" signaling output. We utilized diabodies (DA) as surrogate ligands in a prototypical dimeric receptor-ligand system, the cytokine Erythropoietin (EPO) and its receptor (EpoR), to dimerize EpoR ectodomains in non-native architectures. Diabody-induced signaling amplitudes varied from full to minimal agonism, and structures of these DA/EpoR complexes differed in EpoR dimer orientation and proximity. Diabodies also elicited biased or differential activation of signaling pathways and gene expression profiles compared to EPO. Non-signaling diabodies inhibited proliferation of erythroid precursors from patients with a myeloproliferative neoplasm due to a constitutively active JAK2V617F mutation. Thus, intracellular oncogenic mutations causing ligand-independent receptor activation can be counteracted by extracellular ligands that re-orient receptors into inactive dimer topologies. This approach has broad applications for tuning signaling output for many dimeric receptor systems.


Subject(s)
Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Cell Line , Crystallography, X-Ray , Dimerization , Erythropoietin/metabolism , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mice , Models, Molecular , Molecular Dynamics Simulation , Molecular Sequence Data , Point Mutation , Protein Engineering , Receptors, Erythropoietin/agonists , Receptors, Erythropoietin/antagonists & inhibitors , Sequence Alignment
3.
Proc Natl Acad Sci U S A ; 110(26): 10830-5, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23754417

ABSTRACT

µ-Opioid receptors are among the most studied G protein-coupled receptors because of the therapeutic value of agonists, such as morphine, that are used to treat chronic pain. However, these drugs have significant side effects, such as respiratory suppression, constipation, allodynia, tolerance, and dependence, as well as abuse potential. Efforts to fine tune pain control while alleviating the side effects of drugs, both physiological and psychological, have led to the development of a wide variety of structurally diverse agonist ligands for the µ-opioid receptor, as well as compounds that target κ- and δ-opioid receptors. In recent years, the identification of allosteric ligands for some G protein-coupled receptors has provided breakthroughs in obtaining receptor subtype-selectivity that can reduce the overall side effect profiles of a potential drug. However, positive allosteric modulators (PAMs) can also have the specific advantage of only modulating the activity of the receptor when the orthosteric agonist occupies the receptor, thus maintaining spatial and temporal control of receptor signaling in vivo. This second advantage of allosteric modulators may yield breakthroughs in opioid receptor research and could lead to drugs with improved side-effect profiles or fewer tolerance and dependence issues compared with orthosteric opioid receptor agonists. Here, we describe the discovery and characterization of µ-opioid receptor PAMs and silent allosteric modulators, identified from high-throughput screening using a ß-arrestin-recruitment assay.


Subject(s)
Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Sulfones/pharmacology , Thiazoles/pharmacology , Allosteric Regulation , Allosteric Site , Animals , Arrestins/metabolism , CHO Cells , Cell Line , Cricetinae , Cricetulus , Drug Discovery , High-Throughput Screening Assays , Humans , Ligands , Rats , Sulfones/chemistry , Thiazoles/chemistry , beta-Arrestins
4.
J Biol Chem ; 288(49): 35039-48, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24145037

ABSTRACT

Chemokines display considerable promiscuity with multiple ligands and receptors shared in common, a phenomenon that is thought to underlie their biochemical "redundancy." Their receptors are part of a larger seven-transmembrane receptor superfamily, commonly referred to as G protein-coupled receptors, which have been demonstrated to be able to signal with different efficacies to their multiple downstream signaling pathways, a phenomenon referred to as biased agonism. Biased agonism has been primarily reported as a phenomenon of synthetic ligands, and the biologic prevalence and importance of such signaling are unclear. Here, to assess the presence of biased agonism that may underlie differential signaling by chemokines targeting the same receptor, we performed a detailed pharmacologic analysis of a set of chemokine receptors with multiple endogenous ligands using assays for G protein signaling, ß-arrestin recruitment, and receptor internalization. We found that chemokines targeting the same receptor can display marked differences in their efficacies for G protein- or ß-arrestin-mediated signaling or receptor internalization. This ligand bias correlates with changes in leukocyte migration, consistent with different mechanisms underlying the signaling downstream of these receptors induced by their ligands. These findings demonstrate that biased agonism is a common and likely evolutionarily conserved biological mechanism for generating qualitatively distinct patterns of signaling via the same receptor in response to different endogenous ligands.


Subject(s)
Receptors, Chemokine/agonists , Receptors, Chemokine/metabolism , Arrestins/metabolism , Chemokines/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Kinetics , Ligands , Models, Biological , Signal Transduction , beta-Arrestins
5.
Nat Genet ; 36(2): 183-9, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14704668

ABSTRACT

Small interfering RNA (siRNA) technology facilitates the study of loss of gene function in mammalian cells and animal models, but generating multiple siRNA vectors using oligonucleotides is slow, inefficient and costly. Here we describe a new, enzyme-mediated method for generating numerous functional siRNA constructs from any gene of interest or pool of genes. To test our restriction enzyme-generated siRNA (REGS) system, we silenced a transgene and two endogenous genes and obtained the predicted phenotypes. REGS generated on average 34 unique siRNAs per kilobase of sequence. REGS enabled us to create enzymatically a complex siRNA library (>4 x 10(5) clones) from double-stranded cDNA encompassing known and unknown genes with 96% of the clones containing inserts of the appropriate size.


Subject(s)
DNA Restriction Enzymes/metabolism , Genetic Vectors , Genomic Library , RNA, Small Interfering/metabolism , Animals , Gene Silencing/physiology , Mice , Myoblasts/metabolism
6.
FASEB J ; 21(14): 3827-34, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17942829

ABSTRACT

G-protein-coupled receptor (GPCR) signaling is involved in a wide range of physiological processes and diseases, and around one-half of currently used drugs target GPCRs. Assays for the signaling of GPCRs have suffered from drawbacks, including low signal-to-noise, temporally transient signals, and difficulty in applying a single assay to a wide range of GPCRs. We have developed a set of assays for G-protein-coupled receptor signaling based on beta-galactosidase enzyme complementation in live mammalian cells. We previously described an assay for GPCR activation by monitoring the binding of beta-arrestin to the receptor. Here we describe a second assay that monitors the internalization of GPCRs to endosomes, an event that follows receptor activation and is critical in desensitizing and resensitizing the receptor. We show that both assays display high signal-to-noise ratios with low variability and are quantitative for a wide range of GPCRs. EC50s obtained with these assays closely match results reported in the literature. Finally, we show that these assays are readily adapted to high-throughput chemical screens. Thus, these two assays for monitoring G-protein-coupled receptor activation and internalization should prove valuable in basic biological studies as well as in high-throughput screens.


Subject(s)
Genetic Complementation Test , Receptors, G-Protein-Coupled/metabolism , beta-Galactosidase/genetics , Animals , Biological Transport, Active/genetics , Cell Line , Cell Membrane/enzymology , Cell Membrane/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Humans , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/physiology , Temperature
7.
FASEB J ; 21(14): 3819-26, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17942828

ABSTRACT

G-protein coupled receptors (GPCRs) are a versatile and ubiquitous family of membrane receptors that transmit extracellular signals to mammalian cells and constitute the most important class of drug targets. Yet, sensitive and specific methods are lacking that would allow quantitative comparisons of pharmacologic properties of these receptors in physiological or pathological settings in live animals. We sought to overcome these limitations by employing low affinity, reversible beta-galactosidase complementation to quantify GPCR activation via interaction with beta-arrestin. A panel of cell lines was engineered expressing different GPCRs together with the reporter system. In vitro evaluation revealed highly sensitive, dynamic, and specific assessment of GPCR agonists and antagonists. Following implantation of the cells into mice, it was possible for the first time to monitor pharmacological GPCR activation and inhibition in their physiological context by noninvasive bioluminescence imaging in living animals. This technology has unique advantages that enable novel applications in the functional investigation of GPCR modulation in live animals in biological research and drug discovery.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Animals , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Firefly Luciferin , Humans , Luminescent Agents , Mice , Mice, Inbred BALB C , Mice, Nude , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics
8.
Elife ; 62017 05 12.
Article in English | MEDLINE | ID: mdl-28498099

ABSTRACT

Cytokine and growth-factor ligands typically signal through homo- or hetero-dimeric cell surface receptors via Janus Kinase (JAK/TYK), or Receptor Tyrosine Kinase (RTK)-mediated trans-phosphorylation. However, the number of receptor dimer pairings occurring in nature is limited to those driven by natural ligands encoded within our genome. We have engineered synthethic cytokines (synthekines) that drive formation of cytokine receptor dimer pairings that are not formed by endogenous cytokines and that are not found in nature, and which activate distinct signaling programs. We show that a wide range of non-natural cytokine receptor hetero-dimers are competent to elicit a signaling output. We engineered synthekine ligands that assembled IL-2Rß/IL-4Rα or IL-4Rα/IFNAR2 receptor heterodimers, that do not occur naturally, triggering signaling and functional responses distinct from those activated by the endogenous cytokines IL-2, IL-4, and IFN. Furthermore, hybrid synthekine ligands that dimerized a JAK/STAT cytokine receptor with a receptor tyrosine kinase (RTK) also elicited a signaling response. Synthekines represent a new family of synthetic ligands with pre-defined receptors, but 'orphan' functions, that enable the full combinatorial scope of dimeric signaling receptors encoded within the human genome to be exploited for basic research and drug discovery.


Subject(s)
Cytokines/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Receptors, Cell Surface/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Cell Line , Cytokines/genetics , Humans , Recombinant Proteins/genetics
9.
J Med Chem ; 58(10): 4220-9, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25901762

ABSTRACT

Allosteric modulators of G protein-coupled receptors (GPCRs) have a number of potential advantages compared to agonists or antagonists that bind to the orthosteric site of the receptor. These include the potential for receptor selectivity, maintenance of the temporal and spatial fidelity of signaling in vivo, the ceiling effect of the allosteric cooperativity which may prevent overdose issues, and engendering bias by differentially modulating distinct signaling pathways. Here we describe the discovery, synthesis, and molecular pharmacology of δ-opioid receptor-selective positive allosteric modulators (δ PAMs). These δ PAMs increase the affinity and/or efficacy of the orthosteric agonists leu-enkephalin, SNC80 and TAN67, as measured by receptor binding, G protein activation, ß-arrestin recruitment, adenylyl cyclase inhibition, and extracellular signal-regulated kinases (ERK) activation. As such, these compounds are useful pharmacological tools to probe the molecular pharmacology of the δ receptor and to explore the therapeutic potential of δ PAMs in diseases such as chronic pain and depression.


Subject(s)
Receptors, Opioid, delta/metabolism , Structure-Activity Relationship , Animals , Arrestins/metabolism , Benzamides/pharmacology , Binding, Competitive , CHO Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Chemistry Techniques, Synthetic , Cricetulus , Drug Discovery , Drug Evaluation, Preclinical/methods , Enkephalin, Leucine/pharmacology , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Molecular Structure , Molecular Targeted Therapy , Piperazines/pharmacology , Protein Binding , Quinolines/pharmacology , beta-Arrestins
10.
J Biomol Screen ; 18(5): 599-609, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23396314

ABSTRACT

A variety of G-protein-coupled receptor (GPCR) screening technologies have successfully partnered a number of GPCRs with their cognate ligands. GPCR-mediated ß-arrestin recruitment is now recognized as a distinct intracellular signaling pathway, and ligand-receptor interactions may show a bias toward ß-arrestin over classical GPCR signaling pathways. We hypothesized that the failure to identify native ligands for the remaining orphan GPCRs may be a consequence of biased ß-arrestin signaling. To investigate this, we assembled 10 500 candidate ligands and screened 82 GPCRs using PathHunter ß-arrestin recruitment technology. High-quality screening assays were validated by the inclusion of liganded receptors and the detection and confirmation of these established ligand-receptor pairings. We describe a candidate endogenous orphan GPCR ligand and a number of novel surrogate ligands. However, for the majority of orphan receptors studied, measurement of ß-arrestin recruitment did not lead to the identification of cognate ligands from our screening sets. ß-Arrestin recruitment represents a robust GPCR screening technology, and ligand-biased signaling is emerging as a therapeutically exploitable feature of GPCR biology. The identification of cognate ligands for the orphan GPCRs and the extent to which receptors may exist to preferentially signal through ß-arrestin in response to their native ligand remain to be determined.


Subject(s)
Arrestins/metabolism , High-Throughput Screening Assays/methods , Receptors, G-Protein-Coupled/agonists , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Drug Discovery/methods , HEK293 Cells , Humans , Ligands , Protein Binding/physiology , Receptors, G-Protein-Coupled/metabolism , Saccharomyces cerevisiae , Small Molecule Libraries/analysis , beta-Arrestins
11.
Methods Mol Biol ; 897: 171-80, 2012.
Article in English | MEDLINE | ID: mdl-22674165

ABSTRACT

More than two-thirds of all known G-protein coupled receptors are known to modulate the function of adenylate cyclase resulting in altered levels of cAMP. In turn, cAMP fluctuations transform agonist binding events into physiological changes in cell behavior. The advent of nonradioactive, homogeneous methods of measuring intracellular cAMP has enabled the rapid growth of drug discovery and research applications for these GPCR targets. In this chapter, we describe a nonradioactive, chemiluminescent cAMP detection method using enzyme fragment complementation technology to detect a wide range of GPCR modulators which is also suitable for high-throughput screening.


Subject(s)
Biological Assay/methods , Cyclic AMP/chemistry , Cyclic AMP/metabolism , Luminescent Measurements/methods , Receptors, G-Protein-Coupled/metabolism , Allosteric Regulation , Cell Line , Receptors, G-Protein-Coupled/agonists
12.
Methods Mol Biol ; 897: 181-203, 2012.
Article in English | MEDLINE | ID: mdl-22674166

ABSTRACT

The recruitment of arrestins to activated 7TMRs results in the activation of alternative signaling pathways, quenching of G-protein activation, and coupling to clathrin-mediated endocytosis. The nearly ubiquitous involvement of arrestin in 7TMR signaling has spurred the development of several methods for monitoring this interaction in mammalian cells. Nonetheless, few maintain the reproducibility and precision necessary for drug discovery applications. Enzyme fragment complementation technology (EFC) is an emerging protein-protein interaction technology based on the forced complementation of a split enzyme that has proven to be highly effective in monitoring the formation of GPCR-arrestin complexes. In these systems, the target proteins are fused to two fragments of an enzyme that show little or no spontaneous complementation. Interaction of the two proteins forces the complementation of the enzyme, resulting in an enzymatic measure of the protein interaction. This chapter discusses the utility and methods involved in using the PathHunter ß-galactosidase complementation system to monitor arrestin recruitment and the advantages of exploiting this pathway in the characterization of 7TMR function.


Subject(s)
Arrestins/metabolism , Drug Discovery/methods , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line , Humans , Protein Binding , beta-Arrestins , beta-Galactosidase/metabolism
13.
Methods Mol Biol ; 574: 249-59, 2009.
Article in English | MEDLINE | ID: mdl-19685314

ABSTRACT

Bioluminescence using the reporter enzyme firefly luciferase (Fluc) and the substrate luciferin enables non-invasive optical imaging of living animals with extremely high sensitivity. This type of analysis enables studies of gene expression, tumor growth, and cell migration over time in live animals that were previously not possible. However, a major limitation of this system is that Fluc activity is restricted to the intracellular environment, which precludes important applications of in vivo imaging such as antibody labeling, or serum protein monitoring. In order to expand the application of bioluminescence imaging to other enzymes, we characterized a sequential reporter-enzyme luminescence (SRL) technology for the in vivo detection of beta-galactosidase (beta-gal) activity. The substrate is a "caged" D-luciferin conjugate that must first be cleaved by beta-gal before it can be catalyzed by Fluc in the final, light-emitting step. Hence, luminescence is dependent on and correlates with beta-gal activity. A variety of experiments were performed in order to validate the system and explore potential new applications. We were able to visualize non-invasively over time constitutive beta-gal activity in engineered cells, as well as inducible tissue-specific beta-gal expression in transgenic mice. Since beta-gal, unlike Fluc, retains full activity outside of cells, we were able to show that antibodies conjugated to the recombinant beta-gal enzyme could be used to detect and localize endogenous cells and extracellular antigens in vivo. In addition, we developed a low-affinity beta-gal complementation system that enables inducible, reversible protein interactions to be monitored in real time in vivo, for example, sequential responses to agonists and antagonists of G-protein-coupled receptors (GPCRs). Thus, using SRL, the exquisite luminescent properties of Fluc can be combined with the advantages of another enzyme. Other substrates have been described that extend the scope to endogenous enzymes, such as cytochromes or caspases, potentially enabling additional unprecedented applications.


Subject(s)
Genes, Reporter , Luminescence , beta-Galactosidase/metabolism , Animals , Mice , Mice, Nude , beta-Galactosidase/genetics
14.
Differentiation ; 73(6): 287-93, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16138829

ABSTRACT

RNA interference constitutes a major means of eliminating mRNAs, yet how the small interfering RNAs (siRNA) within the RNA-induced silencing complex (RISC) finds its homologous target in the cell remains unknown. An attractive hypothesis is that RNA interference is linked to translation which allows RISC ready access to every translated mRNA. To test whether translation could direct siRNAs to mRNAs, chemical and biological inhibitors of translation and their effects on mRNA cleavage were tested. Our results show that mRNA degradation by siRNAs is not dependent on mRNA translation.


Subject(s)
Protein Biosynthesis/genetics , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Actins/biosynthesis , Actins/genetics , Cells, Cultured , Clotrimazole/metabolism , Humans , Iron/metabolism , Protein Biosynthesis/drug effects
15.
Nat Methods ; 2(7): 521-7, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15973423

ABSTRACT

Fundamental to eukaryotic cell signaling is the regulation of protein function by directed localization. Detection of these events has been largely qualitative owing to the limitations of existing technologies. Here we describe a method for quantitatively assessing protein translocation using proximity-induced enzyme complementation. The complementation assay for protein translocation (CAPT) is derived from beta-galactosidase and comprises one enzyme fragment, omega, which is localized to a particular subcellular region, and a small complementing peptide, alpha, which is fused to the protein of interest. The concentration of alpha in the immediate vicinity of omega correlates with the amount of enzyme activity obtained in a dose- and time-dependent manner, thus acting as a genetically encoded biosensor for local protein concentration. Using CAPT, inducible protein movement from the cytosol to the nucleus or plasma membrane was quantitatively monitored in multiwell format and in live mammalian cells by flow cytometry.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry/methods , Protein Interaction Mapping/methods , Protein Transport/physiology , Proteins/metabolism , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL