Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Compr Physiol ; 12(4): 4103-4118, 2022 08 29.
Article in English | MEDLINE | ID: mdl-36036567

ABSTRACT

Serotonin is often referred to as a "happy hormone" as it maintains good mood, well-being, and happiness. It is involved in communication between nerve cells and plays a role in sleeping and digestion. However, too much serotonin can have pathogenic effects and serotonin synthesis is elevated in pulmonary artery endothelial cells from patients with pulmonary arterial hypertension (PAH). PAH is characterized by elevated pulmonary pressures, right ventricular failure, inflammation, and pulmonary vascular remodeling; serotonin has been shown to be associated with these pathologies. The rate-limiting enzyme in the synthesis of serotonin in the periphery of the body is tryptophan hydroxylase 1 (TPH1). TPH1 expression and serotonin synthesis are elevated in pulmonary artery endothelial cells in patients with PAH. The serotonin synthesized in the pulmonary arterial endothelium can act on the adjacent pulmonary arterial smooth muscle cells (PASMCs), adventitial macrophages, and fibroblasts, in a paracrine fashion. In humans, serotonin enters PASMCs cells via the serotonin transporter (SERT) and it can cooperate with the 5-HT1B receptor on the plasma membrane; this activates both contractile and proliferative signaling pathways. The "serotonin hypothesis of pulmonary hypertension" arose when serotonin was associated with PAH induced by diet pills such as fenfluramine, aminorex, and chlorphentermine; these act as indirect serotonergic agonists causing the release of serotonin from platelets and cells through the SERT. Here the role of serotonin in PAH is reviewed. Targeting serotonin synthesis or signaling is a promising novel alternative approach which may lead to novel therapies for PAH. © 2022 American Physiological Society. Compr Physiol 12: 1-16, 2022.


Subject(s)
Hypertension, Pulmonary , Cell Proliferation , Endothelial Cells/metabolism , Humans , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pulmonary Artery , Serotonin/metabolism , Serotonin/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/pharmacology
2.
Sci Signal ; 15(726): eabg5203, 2022 03 22.
Article in English | MEDLINE | ID: mdl-35316095

ABSTRACT

G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors and signal through the proximal effectors, G proteins and ß-arrestins, to influence nearly every biological process. The G protein and ß-arrestin signaling pathways have largely been considered separable; however, direct interactions between Gα proteins and ß-arrestins have been described that appear to be part of a distinct GPCR signaling pathway. Within these complexes, Gαi/o, but not other Gα protein subtypes, directly interacts with ß-arrestin, regardless of the canonical Gα protein that is coupled to the GPCR. Here, we report that the endogenous biased chemokine agonists of CXCR3 (CXCL9, CXCL10, and CXCL11), together with two small-molecule biased agonists, differentially formed Gαi:ß-arrestin complexes. Formation of the Gαi:ß-arrestin complexes did not correlate well with either G protein activation or ß-arrestin recruitment. ß-arrestin biosensors demonstrated that ligands that promoted Gαi:ß-arrestin complex formation generated similar ß-arrestin conformations. We also found that Gαi:ß-arrestin complexes did not couple to the mitogen-activated protein kinase ERK, as is observed with other receptors such as the V2 vasopressin receptor, but did couple with the clathrin adaptor protein AP-2, which suggests context-dependent signaling by these complexes. These findings reinforce the notion that Gαi:ß-arrestin complex formation is a distinct GPCR signaling pathway and enhance our understanding of the spectrum of biased agonism.


Subject(s)
GTP-Binding Proteins , Receptors, G-Protein-Coupled , GTP-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , beta-Arrestin 1/genetics , beta-Arrestin 1/metabolism , beta-Arrestins/metabolism
3.
eNeuro ; 9(5)2022.
Article in English | MEDLINE | ID: mdl-36635920

ABSTRACT

The protease caspase-3 is a key mediator of apoptotic programmed cell death. But weak or transient caspase activity can contribute to neuronal differentiation, axonal pathfinding, and synaptic long-term depression. Despite the importance of sublethal, or nonapoptotic, caspase activity in neurodevelopment and neural plasticity, there has been no simple method for mapping and quantifying nonapoptotic caspase activity (NACA) in rodent brains. We therefore generated a transgenic mouse expressing a highly sensitive and specific fluorescent reporter of caspase activity, with peak signal localized to the nucleus. As a proof of concept, we first obtained evidence that NACA influences neurophysiology in an amygdalar circuit. Then focusing on the amygdala, we were able to quantify a sex-specific persistent elevation in caspase activity in females after restraint stress. This simple in vivo caspase activity reporter will facilitate systems-level studies of apoptotic and nonapoptotic phenomena in behavioral and pathologic models.


Subject(s)
Apoptosis , Brain , Male , Female , Mice , Animals , Apoptosis/physiology , Mice, Transgenic , Neuronal Plasticity , Caspase 9
4.
FEBS J ; 288(8): 2550-2561, 2021 04.
Article in English | MEDLINE | ID: mdl-33539669

ABSTRACT

G protein-coupled receptors (GPCRs) canonically couple to specific Gα protein subtypes and ß-arrestin adaptor proteins to initiate cellular signaling events. G protein-mediated signaling and ß-arrestin-mediated signaling have broadly been considered separable. However, noncanonical interactions between G proteins and GPCRs are now appreciated that do not result in nucleotide exchange and classical G protein signaling. New findings also demonstrate direct interactions between G proteins and ß-arrestins that are required for certain signaling and physiological events. Further adding to the intrigue of these newly appreciated G protein:ß-arrestin complexes, only the Gαi subtype family members, and not Gαs, Gαq/11, or Gα12/13 subtypes, appear to form direct interactions with ß-arrestin. Here, we review the recent discovery and initial characterization of G protein:ß-arrestin complexes and describe how these complexes provide mechanistic insight into seemingly disparate observations. G protein:ß-arrestin complexes build upon other observations of noncanonical G protein and ß-arrestin signaling events to add an additional dimension to our understanding of GPCR signaling.


Subject(s)
GTP-Binding Proteins/genetics , Protein Conformation , Receptors, G-Protein-Coupled/genetics , beta-Arrestins/genetics , HEK293 Cells , Humans , Phosphorylation/genetics , Protein Binding/genetics , Signal Transduction/genetics
5.
Science ; 371(6534)2021 03 12.
Article in English | MEDLINE | ID: mdl-33479120

ABSTRACT

Heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) are common drug targets and canonically couple to specific Gα protein subtypes and ß-arrestin adaptor proteins. G protein-mediated signaling and ß-arrestin-mediated signaling have been considered separable. We show here that GPCRs promote a direct interaction between Gαi protein subtype family members and ß-arrestins regardless of their canonical Gα protein subtype coupling. Gαi:ß-arrestin complexes bound extracellular signal-regulated kinase (ERK), and their disruption impaired both ERK activation and cell migration, which is consistent with ß-arrestins requiring a functional interaction with Gαi for certain signaling events. These results introduce a GPCR signaling mechanism distinct from canonical G protein activation in which GPCRs cause the formation of Gαi:ß-arrestin signaling complexes.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Receptors, G-Protein-Coupled/metabolism , beta-Arrestins/metabolism , Bioluminescence Resonance Energy Transfer Techniques , Cell Movement , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Signal Transduction
6.
J Clin Transl Sci ; 5(1): e10, 2020 Jun 11.
Article in English | MEDLINE | ID: mdl-34192045

ABSTRACT

INTRODUCTION: The COVID-19 pandemic has created a high demand on personal protective equipment, including disposable N95 masks. Given the need for mask reuse, we tested the feasibility of vaporized hydrogen peroxide (VHP), ultraviolet light (UV), and ethanol decontamination strategies on N95 mask integrity and the ability to remove the infectious potential of SARS-CoV-2. METHODS: Disposable N95 masks, including medical grade (1860, 1870+) and industrial grade (8511) masks, were treated by VHP, UV, and ethanol decontamination. Mask degradation was tested using a quantitative respirator fit testing. Pooled clinical samples of SARS-CoV-2 were applied to mask samples, treated, and then either sent immediately for real-time reverse transcriptase-polymerase chain reaction (RT-PCR) or incubated with Vero E6 cells to assess for virucidal effect. RESULTS: Both ethanol and UV decontamination showed functional degradation to different degrees while VHP treatment showed no significant change after two treatments. We also report a single SARS-CoV-2 virucidal experiment using Vero E6 cell infection in which only ethanol treatment eliminated detectable SARS-CoV-2 RNA. CONCLUSIONS: We hope our data will guide further research for evidenced-based decisions for disposable N95 mask reuse and help protect caregivers from SARS-CoV-2 and other pathogens.

7.
J Biol Chem ; 293(16): 6161-6171, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29487132

ABSTRACT

The dopamine D2 receptor (D2R) is a G protein-coupled receptor (GPCR) that is critical for many central nervous system functions. The D2R carries out these functions by signaling through two transducers: G proteins and ß-arrestins (ßarrs). Selectively engaging either the G protein or ßarr pathway may be a way to improve drugs targeting GPCRs. The current model of GPCR signal transduction posits a chain of events where G protein activation ultimately leads to ßarr recruitment. GPCR kinases (GRKs), which are regulated by G proteins and whose kinase action facilitates ßarr recruitment, bridge these pathways. Therefore, ßarr recruitment appears to be intimately tied to G protein activation via GRKs. Here we sought to understand how GRK2 action at the D2R would be disrupted when G protein activation is eliminated and the effect of this on ßarr recruitment. We used two recently developed biased D2R mutants that can preferentially interact either with G proteins or ßarrs as well as a ßarr-biased D2R ligand, UNC9994. With these functionally selective tools, we investigated the mechanism whereby the ßarr-preferring D2R achieves ßarr pathway activation in the complete absence of G protein activation. We describe how direct, G protein-independent recruitment of GRK2 drives interactions at the ßarr-preferring D2R and also contributes to ßarr recruitment at the WT D2R. Additionally, we found an additive interaction between the ßarr-preferring D2R mutant and UNC9994. These results reveal that the D2R can directly recruit GRK2 without G protein activation and that this mechanism may have relevance to achieving ßarr-biased signaling.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , GTP-Binding Proteins/metabolism , Receptors, Dopamine D2/metabolism , Dopamine Agonists/pharmacology , Energy Transfer , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 3/antagonists & inhibitors , HEK293 Cells , Humans , Models, Theoretical , Mutation , Pertussis Toxin/metabolism , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/pharmacology , Receptors, Dopamine D2/genetics , Signal Transduction , beta-Arrestins/metabolism
8.
Synapse ; 72(1)2018 01.
Article in English | MEDLINE | ID: mdl-28941296

ABSTRACT

The "brain-gut" peptide ghrelin, which mediates food-seeking behaviors, is recognized as a very strong endogenous modulator of dopamine (DA) signaling. Ghrelin binds the G protein-coupled receptor GHSR1a, and administration of ghrelin increases the rewarding properties of psychostimulants while ghrelin receptor antagonists decrease them. In addition, the GHSR1a signals through ßarrestin-2 to regulate actin/stress fiber rearrangement, suggesting ßarrestin-2 participation in the regulation of actin-mediated synaptic plasticity for addictive substances like cocaine. The effects of ghrelin receptor ligands on reward strongly suggest that modulation of ghrelin signaling could provide an effective strategy to ameliorate undesirable behaviors arising from addiction. To investigate this possibility, we tested the effects of ghrelin receptor antagonism in a cocaine behavioral sensitization paradigm using DA neuron-specific ßarrestin-2 KO mice. Our results show that these mice sensitize to cocaine as well as wild-type littermates. The ßarrestin-2 KO mice, however, no longer respond to the locomotor attenuating effects of the GHSR1a antagonist YIL781. The data presented here suggest that the separate stages of addictive behavior differ in their requirements for ßarrestin-2 and show that pharmacological inhibition of ßarrestin-2 function through GHSR1a antagonism is not equivalent to the loss of ßarrestin-2 function achieved by genetic ablation. These data support targeting GHSR1a signaling in addiction therapy but indicate that using signaling biased compounds that modulate ßarrestin-2 activity differentially from G protein activity may be required.


Subject(s)
Cocaine-Related Disorders/metabolism , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Motor Activity/drug effects , Receptors, Ghrelin/antagonists & inhibitors , beta-Arrestin 2/metabolism , Animals , Cell Line, Tumor , Central Nervous System Agents , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Female , Ghrelin/metabolism , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Piperidines/pharmacology , Quinazolinones/pharmacology , Receptors, Ghrelin/metabolism , beta-Arrestin 2/genetics
9.
Neuropsychopharmacology ; 43(5): 1164-1173, 2018 04.
Article in English | MEDLINE | ID: mdl-29068002

ABSTRACT

The dopamine D2 receptor (D2R), like many G-protein-coupled receptors, signals through G-protein- and ß-arrestin-dependent pathways. Preferential activation of one of these pathways is termed functional selectivity or biased signaling and is a promising therapeutic strategy. Though biased signaling through D2Rs has been demonstrated, acquiring the mechanistic details of biased D2R/G-protein and D2R/ß-arrestin signaling in vivo has been challenging because of the lack of techniques that specifically target these interactions in discrete cell populations. To address this question, we employed a cell type-specific viral expression approach to restore D2R variants that preferentially engage either G-protein or ß-arrestin signaling in 'indirect pathway' medium spiny neurons (iMSNs), because of their central role in dopamine circuitry. We found that the effect of haloperidol antagonism on D2R metabolic signaling events is largely mediated by acute blockade of D2R/G-protein signaling. We show that a D2R-driven behavior, nestlet shredding, is similarly driven by D2R/G-protein signaling. On the other hand, D2R-driven locomotion and rearing require coordinated D2R/G-protein and D2R/ß-arrestin signaling. The acute locomotor response to amphetamine and cocaine similarly depend on both G-protein and ß-arrestin D2R signaling. Surprisingly, another psychotropic drug, phencyclidine, displayed a selective D2R/ß-arrestin potentiation of locomotion. These findings highlight how D2R mostly relies upon balanced G-protein and ß-arrestin signaling in iMSNs. However, the response to haloperidol and phencyclidine indicates that normal D2R signaling homeostasis can be dramatically altered, indicating that targeting a specific D2R signal transduction pathway could allow for more precise modulation of dopamine circuit function.


Subject(s)
Dopamine/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Receptors, Dopamine D2/genetics , beta-Arrestins/metabolism , Amphetamine/pharmacology , Animals , Behavior, Animal/physiology , Cocaine/pharmacology , Female , Genetic Vectors , Haloperidol/pharmacology , Male , Mice , Mice, Knockout , Mice, Transgenic , Phencyclidine , Signal Transduction/drug effects , Signal Transduction/physiology
10.
Mol Pharmacol ; 92(2): 136-150, 2017 08.
Article in English | MEDLINE | ID: mdl-28559424

ABSTRACT

Biased agonism, the ability of different ligands for the same receptor to selectively activate some signaling pathways while blocking others, is now an established paradigm for G protein-coupled receptor signaling. One group of receptors in which endogenous bias is critical is the chemokine system, consisting of over 50 ligands and 20 receptors that bind one another with significant promiscuity. We have previously demonstrated that ligands for the same receptor can cause biased signaling responses. The goal of this study was to identify mechanisms that could underlie biased signaling between different receptor splice variants. The C-X-C motif chemokine receptor 3 (CXCR3) has two splice variants, CXCR3A and CXCR3B, which differ by 51 amino acids at its N-terminus. Consistent with an earlier study, we found that C-X-C motif chemokine ligands 4, 9, 10, and 11 all activated G αi at CXCR3A, while at CXCR3B these ligands demonstrated no measurable G αi or G αs activity. ß-arrestin (ßarr) was recruited at a reduced level to CXCR3B relative to CXCR3A, which was also associated with differences in ßarr2 conformation. ßarr2 recruitment to CXCR3A was attenuated by both G protein receptor kinase (GRK) 2/3 and GRK5/6 knockdown, while only GRK2/3 knockdown blunted recruitment to CXCR3B. Extracellular regulated kinase 1/2 phosphorylation downstream from CXCR3A and CXCR3B was increased and decreased, respectively, by ßarr1/2 knockout. The splice variants also differentially activated transcriptional reporters. These findings demonstrate that differential splicing of CXCR3 results in biased responses associated with distinct patterns of ßarr conformation and recruitment. Differential splicing may serve as a common mechanism for generating biased signaling and provides insights into how chemokine receptor signaling can be modulated post-transcriptionally.


Subject(s)
Receptors, CXCR3/metabolism , Signal Transduction/physiology , beta-Arrestins/metabolism , Amino Acid Sequence , HEK293 Cells , Humans , Luciferases, Renilla/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, CXCR3/genetics , beta-Arrestins/genetics
11.
J Biol Chem ; 292(17): 7208-7222, 2017 04 28.
Article in English | MEDLINE | ID: mdl-28275053

ABSTRACT

The leucine-rich G protein-coupled receptor-5 (LGR5) is expressed in adult tissue stem cells of many epithelia, and its overexpression is negatively correlated with cancer prognosis. LGR5 potentiates WNT/ß-catenin signaling through its unique constitutive internalization property that clears negative regulators of the WNT-receptor complex from the membrane. However, both the mechanism and physiological relevance of LGR5 internalization are unclear. Therefore, a natural product library was screened to discover LGR5 internalization inhibitors and gain mechanistic insight into LGR5 internalization. The plant lignan justicidin B blocked the constitutive internalization of LGR5. Justicidin B is structurally similar to more potent vacuolar-type H+-ATPase inhibitors, which all inhibited LGR5 internalization by blocking clathrin-mediated endocytosis. We then tested the physiological relevance of LGR5 internalization blockade in vivo A LGR5-rainbow (LBOW) mouse line was engineered to express three different LGR5 isoforms along with unique fluorescent protein lineage reporters in the same mouse. In this manner, the effects of each isoform on cell fate can be simultaneously assessed through simple fluorescent imaging for each lineage reporter. LBOW mice express three different forms of LGR5, a wild-type form that constitutively internalizes and two mutant forms whose internalization properties have been compromised by genetic perturbations within the carboxyl-terminal tail. LBOW was activated in the intestinal epithelium, and a year-long lineage-tracing course revealed that genetic blockade of LGR5 internalization diminished cell fitness. Together these data provide proof-of-concept genetic evidence that blocking the clathrin-mediated endocytosis of LGR5 could be used to pharmacologically control cell behavior.


Subject(s)
Clathrin/chemistry , Endocytosis , Leucine/chemistry , Receptors, G-Protein-Coupled/chemistry , Adenosine Triphosphatases/chemistry , Animals , Cell Line, Tumor , Cell Lineage , Cell Proliferation , Dioxolanes/chemistry , Epithelium/metabolism , Female , Homeostasis , Humans , Lignans/chemistry , Mice , Mice, Inbred C57BL , Protein Isoforms , Rats , Stem Cells/cytology , Stochastic Processes , Wnt Signaling Pathway
12.
Proc Natl Acad Sci U S A ; 113(50): E8178-E8186, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27911814

ABSTRACT

The current dopamine (DA) hypothesis of schizophrenia postulates striatal hyperdopaminergia and cortical hypodopaminergia. Although partial agonists at DA D2 receptors (D2Rs), like aripiprazole, were developed to simultaneously target both phenomena, they do not effectively improve cortical dysfunction. In this study, we investigate the potential for newly developed ß-arrestin2 (ßarr2)-biased D2R partial agonists to simultaneously target hyper- and hypodopaminergia. Using neuron-specific ßarr2-KO mice, we show that the antipsychotic-like effects of a ßarr2-biased D2R ligand are driven through both striatal antagonism and cortical agonism of D2R-ßarr2 signaling. Furthermore, ßarr2-biased D2R agonism enhances firing of cortical fast-spiking interneurons. This enhanced cortical agonism of the biased ligand can be attributed to a lack of G-protein signaling and elevated expression of ßarr2 and G protein-coupled receptor (GPCR) kinase 2 in the cortex versus the striatum. Therefore, we propose that ßarr2-biased D2R ligands that exert region-selective actions could provide a path to develop more effective antipsychotic therapies.


Subject(s)
Antipsychotic Agents/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/metabolism , beta-Arrestin 2/metabolism , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Dopamine D2 Receptor Antagonists/pharmacology , Female , G-Protein-Coupled Receptor Kinase 2/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Interneurons/metabolism , Ligands , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Phencyclidine/toxicity , Signal Transduction/drug effects
13.
BMC Biol ; 13: 107, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26678094

ABSTRACT

BACKGROUND: Membrane proteins regulate a diversity of physiological processes and are the most successful class of targets in drug discovery. However, the number of targets adequately explored in chemical space and the limited resources available for screening are significant problems shared by drug-discovery centers and small laboratories. Therefore, a low-cost and universally applicable screen for membrane protein trafficking was developed. RESULTS: This high-throughput screen (HTS), termed IRFAP-HTS, utilizes the recently described MarsCy1-fluorogen activating protein and the near-infrared and membrane impermeant fluorogen SCi1. The cell surface expression of MarsCy1 epitope-tagged receptors can be visualized by simple addition of SCi1. User-friendly, rapid, and quantitative detection occurs on a standard infrared western-blotting scanner. The reliability and robustness of IRFAP-HTS was validated by confirming human vasopressin-2 receptor and dopamine receptor-2 trafficking in response to agonist or antagonist. The IRFAP-HTS screen was deployed against the leucine-rich G protein-coupled receptor-5 (Lgr5). Lgr5 is expressed in stem cells, modulates Wnt/ß-catenin signaling, and is therefore a promising drug target. However, small molecule modulators have yet to be reported. The constitutive internalization of Lgr5 appears to be one primary mode through which its function is regulated. Therefore, IRFAP-HTS was utilized to screen 11,258 FDA-approved and drug-like small molecules for those that antagonize Lgr5 internalization. Glucocorticoids were found to potently increase Lgr5 expression at the plasma membrane. CONCLUSION: The IRFAP-HTS platform provides a versatile solution for screening more targets with fewer resources. Using only a standard western-blotting scanner, we were able to screen 5,000 compounds per hour in a robust and quantitative assay. Multi-purposing standardly available laboratory equipment eliminates the need for idiosyncratic and more expensive high-content imaging systems. The modular and user-friendly IRFAP-HTS is a significant departure from current screening platforms. Small laboratories will have unprecedented access to a robust and reliable screening platform and will no longer be limited by the esoteric nature of assay development, data acquisition, and post-screening analysis. The discovery of glucocorticoids as modulators for Lgr5 trafficking confirms that IRFAP-HTS can accelerate drug-discovery and drug-repurposing for even the most obscure targets.


Subject(s)
Drug Discovery/methods , High-Throughput Screening Assays/methods , Membrane Proteins/metabolism , Drug Discovery/economics , HEK293 Cells , High-Throughput Screening Assays/economics , Humans , Protein Transport , Reproducibility of Results
14.
PLoS One ; 10(10): e0141637, 2015.
Article in English | MEDLINE | ID: mdl-26516769

ABSTRACT

Functional selectivity (or biased agonism) is a property exhibited by some G protein-coupled receptor (GPCR) ligands, which results in the modulation of a subset of a receptor's signaling capabilities and more precise control over complex biological processes. The dopamine D2 receptor (D2R) exhibits pleiotropic responses to the biogenic amine dopamine (DA) to mediate complex central nervous system functions through activation of G proteins and ß-arrestins. D2R is a prominent therapeutic target for psychological and neurological disorders in which DA biology is dysregulated and targeting D2R with functionally selective drugs could provide a means by which pharmacotherapies could be developed. However, factors that determine GPCR functional selectivity in vivo may be multiple with receptors, ligands and transducers contributing to the process. We have recently described a mutagenesis approach to engineer biased D2R mutants in which G protein-dependent ([Gprot]D2R) and ß-arrestin-dependent signaling ([ßarr]D2R) were successfully separated (Peterson, et al. PNAS, 2015). Here, permutations of these mutants were used to identify critical determinants of the D2R signaling complex that impart signaling bias in response to the natural or synthetic ligands. Critical residues identified in generating [Gprot]D2R and [ßarr]D2R conferred control of partial agonism at G protein and/or ß-arrestin activity. Another set of mutations that result in G protein bias was identified that demonstrated that full agonists can impart unique activation patterns, and provided further credence to the concept of ligand texture. Finally, the contributions and interplay between different transducers indicated that G proteins are not aberrantly activated, and that receptor kinase and ß-arrestin activities are inextricably linked. These data provide a thorough elucidation of the feasibility and malleability of D2R functional selectivity and point to means by which novel in vivo therapies could be modeled.


Subject(s)
Arrestins/metabolism , GTP-Binding Proteins/metabolism , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Arrestins/chemistry , Binding Sites , Dopamine/metabolism , GTP-Binding Proteins/chemistry , HEK293 Cells , Humans , Ligands , Models, Molecular , Mutagenesis , Receptors, Dopamine D2/chemistry , Signal Transduction , beta-Arrestins
15.
Proc Natl Acad Sci U S A ; 112(22): 7097-102, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-25964346

ABSTRACT

The neuromodulator dopamine signals through the dopamine D2 receptor (D2R) to modulate central nervous system functions through diverse signal transduction pathways. D2R is a prominent target for drug treatments in disorders where dopamine function is aberrant, such as schizophrenia. D2R signals through distinct G-protein and ß-arrestin pathways, and drugs that are functionally selective for these pathways could have improved therapeutic potential. How D2R signals through the two pathways is still not well defined, and efforts to elucidate these pathways have been hampered by the lack of adequate tools for assessing the contribution of each pathway independently. To address this, Evolutionary Trace was used to produce D2R mutants with strongly biased signal transduction for either the G-protein or ß-arrestin interactions. These mutants were used to resolve the role of G proteins and ß-arrestins in D2R signaling assays. The results show that D2R interactions with the two downstream effectors are dissociable and that G-protein signaling accounts for D2R canonical MAP kinase signaling cascade activation, whereas ß-arrestin only activates elements of this cascade under certain conditions. Nevertheless, when expressed in mice in GABAergic medium spiny neurons of the striatum, the ß-arrestin-biased D2R caused a significant potentiation of amphetamine-induced locomotion, whereas the G protein-biased D2R had minimal effects. The mutant receptors generated here provide a molecular tool set that should enable a better definition of the individual roles of G-protein and ß-arrestin signaling pathways in D2R pharmacology, neurobiology, and associated pathologies.


Subject(s)
Arrestins/metabolism , GTP-Binding Proteins/metabolism , MAP Kinase Signaling System/physiology , Models, Molecular , Receptors, Dopamine D2/metabolism , Animals , Arrestins/chemistry , Corpus Striatum/cytology , Crystallography , GTP-Binding Proteins/chemistry , HEK293 Cells , Humans , MAP Kinase Signaling System/genetics , Mice , Mutagenesis , Neurons/metabolism , Protein Conformation , Receptors, Dopamine D2/chemistry , Receptors, Dopamine D2/genetics , Regression Analysis , beta-Arrestins
16.
Mol Pharmacol ; 84(3): 459-75, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23839567

ABSTRACT

Positive allosteric modulators (PAMs) of α7 nicotinic acetylcholine receptors can enhance ion channel currents and downstream effects of α7 stimulation. We investigated the approach of using noncompetitive antagonists to regulate α7 receptor function, potentially distinguishing effects requiring ion channel currents from signaling induced by nonconducting states. Three small readily reversible antagonists, (1S,2R,4R)-N,2,3,3-tetramethylbicyclo[2.2.1]heptan-2-amine (mecamylamine), N-(2.6-dimethylphenylcarbamoylmethyl)triethylammonium bromide (QX-314), and 2-(dimethylamino)ethyl 4-(butylamino)benzoate (tetracaine), as well as three large slowly reversible antagonists, bis-(2,2,6,6-tetramethyl-4-piperidinyl) sebacate (BTMPS), 2,2,6,6-tetramethylpiperidin-4-yl heptanoate (TMPH), and 1,2,4,5-tetra-{5-[1-(3-benzyl)pyridinium]pent-1-yl}benzene tetrabromide (tkP3BzPB), were investigated for their effectiveness and voltage dependence in the inhibition of responses evoked by acetylcholine alone or augmented by the α7-selective PAM N-(5-chloro-2,4-dimethoxyphenyl)-N'-(5-methyl-3-isoxazolyl)-urea (PNU-120596). Analyses of the small antagonists on PNU-120596-potentiated single-channel bursts indicated that each agent had a distinct mechanism of inhibition and only that of QX-314 was consistent with simple open channel block. In addition to decreasing channel open times and burst durations, mecamylamine and tetracaine induced unique subconductance states. To determine whether channel-blocking activity alone would be sufficient to prevent cell death, the antagonists were tested for their ability to protect α7-expressing cells from cytotoxic effects of the α7 agonist choline in combination with PNU-120596. Only tetracaine and tkP3BzPB, the two agents that had effects least consistent with simple ion channel block, were fully cytoprotective at concentrations that gave submaximal inhibition of macroscopic currents in oocytes. Further analyses indicated that toxicity produced by PNU-120596 and choline was calcium independent and likely an apoptotic event. Our results are consistent with the hypothesis that PAMs may modulate conformational states important for both channel activity and ion channel-independent signaling.


Subject(s)
Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/physiology , Allosteric Regulation , Animals , Calcium/metabolism , Cell Line , Choline/pharmacology , Cytotoxins/pharmacology , Female , Humans , Isoxazoles/pharmacology , Neurons/drug effects , Neurons/physiology , Oocytes/drug effects , Oocytes/physiology , Patch-Clamp Techniques , Phenylurea Compounds/pharmacology , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...