Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Mol Pharmacol ; 104(1): 1-16, 2023 07.
Article in English | MEDLINE | ID: mdl-37147110

ABSTRACT

Current treatments for Alzheimer's disease (AD) help reduce symptoms for a limited time but do not treat the underlying pathology. To identify potential therapeutic targets for AD, an integrative network analysis was previously carried out using 364 human postmortem control, mild cognitive impairment, and AD brains. This analysis identified proline endopeptidase-like protein (PREPL), an understudied protein, as a downregulated protein in late-onset AD patients. In this study we investigate the role of PREPL. Analyses of data from human postmortem samples and PREPL knockdown (KD) cells suggest that PREPL expression modulates pathways associated with protein trafficking, synaptic activities, and lipid metabolism. Furthermore, PREPL KD impairs cell proliferation and modulates the structure of vesicles, levels of neuropeptide-processing enzymes, and secretion of neuropeptides. In addition, decrease in PREPL levels leads to changes in the levels of a number of synaptic proteins as well as changes in the levels of secreted amyloid beta (Aß) 42 peptide and Tau phosphorylation. Finally, we report that local decrease in PREPL levels in mouse hippocampus attenuates long-term potentiation, suggesting a role in synaptic plasticity. Together, our results indicate that PREPL affects neuronal function by modulating protein trafficking and synaptic function, an important mechanism of AD pathogenesis. SIGNIFICANCE STATEMENT: Integrative network analysis reveals proline endopeptidase-like protein (PREPL) to be downregulated in human sporadic late-onset Alzheimer's disease brains. Down regulation of PREPL leads to increases in amyloid beta secretion, Tau phosphorylation, and decreases in protein trafficking and long-term potentiation.


Subject(s)
Alzheimer Disease , Prolyl Oligopeptidases , Animals , Humans , Mice , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Disease Models, Animal , Mice, Transgenic , Multiomics , Prolyl Oligopeptidases/metabolism , Protein Transport
2.
Proc Natl Acad Sci U S A ; 117(21): 11820-11828, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32393639

ABSTRACT

Opioids, such as morphine and fentanyl, are widely used for the treatment of severe pain; however, prolonged treatment with these drugs leads to the development of tolerance and can lead to opioid use disorder. The "Opioid Epidemic" has generated a drive for a deeper understanding of the fundamental signaling mechanisms of opioid receptors. It is generally thought that the three types of opioid receptors (µ, δ, κ) are activated by endogenous peptides derived from three different precursors: Proopiomelanocortin, proenkephalin, and prodynorphin. Posttranslational processing of these precursors generates >20 peptides with opioid receptor activity, leading to a long-standing question of the significance of this repertoire of peptides. Here, we address some aspects of this question using a technical tour de force approach to systematically evaluate ligand binding and signaling properties ([35S]GTPγS binding and ß-arrestin recruitment) of 22 peptides at each of the three opioid receptors. We show that nearly all tested peptides are able to activate the three opioid receptors, and many of them exhibit agonist-directed receptor signaling (functional selectivity). Our data also challenge the dogma that shorter forms of ß-endorphin do not exhibit receptor activity; we show that they exhibit robust signaling in cultured cells and in an acute brain slice preparation. Collectively, this information lays the groundwork for improved understanding of the endogenous opioid system that will help in developing more effective treatments for pain and addiction.


Subject(s)
Opioid Peptides , Receptors, Opioid/metabolism , Signal Transduction/physiology , Animals , Cell Line, Tumor , Humans , Male , Opioid Peptides/agonists , Opioid Peptides/metabolism , Pro-Opiomelanocortin/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley
3.
Mol Pharmacol ; 2022 May 23.
Article in English | MEDLINE | ID: mdl-35605991

ABSTRACT

PEN is an abundant neuropeptide that activates GPR83, a G protein-coupled receptor that is considered a novel therapeutic target due to its roles in regulation of feeding, reward, and anxiety-related behaviors. The major form of PEN in the brain is 22 residues in length. Previous studies have identified shorter forms of PEN in mouse brain and neuroendocrine cells; these shorter forms were named PEN18, PEN19 and PEN20, with the number reflecting the length of the peptide. The C-terminal five residues of PEN20 are identical to the C-terminus of a procholecystokinin (proCCK)-derived peptide, named proCCK56-62, that is present in mouse brain. ProCCK56-62 is highly conserved across species although it has no homology to the bioactive cholecystokinin domain. ProCCK56-62 and a longer form, proCCK56-63 were tested for their ability to engage GPR83. Both peptides bind GPR83 with high affinity, activate second messenger pathways, and induce ligand-mediated receptor endocytosis. Interestingly, the shorter PEN peptides, ProCC56-62, and ProCCK56-63 differentially activate signal transduction pathways. Whereas PEN22 and PEN20 facilitate receptor coupling to Gai, PEN18, PEN19 and ProCCK peptides facilitate coupling to Gas. Furthermore, the ProCCK peptides exhibit dose dependent Ga subtype selectivity in that they faciliate coupling to Gas at low concentrations and Gai at high concentrations. These data demonstrate that peptides derived from two distinct peptide precursors can differentially activate GPR83, and that GPR83 exhibits Ga subtype preference depending on the nature and concentration of the peptide. These results are consistent with the emerging idea that endogenous neuropeptides function as biased ligands. Significance Statement We found that peptides derived from proCCK bind and activate GPR83, a G protein-coupled receptor that is known to bind peptides derived from proSAAS. Different forms of the proCCK- and proSAAS-derived peptides show biased agonism, activating Gas or Gai depending on the length of the peptide and/or its concentration.

4.
Ann Neurol ; 88(6): 1237-1243, 2020 12.
Article in English | MEDLINE | ID: mdl-32833276

ABSTRACT

A 10-year-old girl presented with ileus, urinary retention, dry mouth, lack of tears, fixed dilated pupils, and diffuse anhidrosis 7 days after a febrile illness. We hypothesized that her syndrome was due to autoimmunity against muscarinic acetylcholine receptors, blocking their activation. Using an indirect enzyme-linked immunosorbent assay for all 5 muscarinic receptors (M1 -M5 ), we identified in the patient's serum antibodies that selectively bound to M3 receptors. In vitro functional studies confirmed that these autoantibodies selectively blocked M3 receptor activation. Thus, autoantibodies against M3 acetylcholine receptors cause acute postganglionic cholinergic dysautonomia. ANN NEUROL 2020;88:1237-1243.


Subject(s)
Autoantibodies/immunology , Primary Dysautonomias/immunology , Receptor, Muscarinic M3/immunology , Autoantibodies/blood , Child , Female , Humans , Receptor, Muscarinic M3/antagonists & inhibitors
5.
Cell Mol Neurobiol ; 41(5): 1103-1118, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33389463

ABSTRACT

Activation of µ, δ, and κ opioid receptors by endogenous opioid peptides leads to the regulation of many emotional and physiological responses. The three major endogenous opioid peptides, ß-endorphin, enkephalins, and dynorphins result from the processing of three main precursors: proopiomelanocortin, proenkephalin, and prodynorphin. Using a knockout approach, we sought to determine whether the absence of endogenous opioid peptides would affect the expression or activity of opioid receptors in mice lacking either proenkephalin, ß-endorphin, or both. Since gene knockout can lead to changes in the levels of peptides generated from related precursors by compensatory mechanisms, we directly measured the levels of Leu-enkephalin and dynorphin-derived peptides in the brain of animals lacking proenkephalin, ß-endorphin, or both. We find that whereas the levels of dynorphin-derived peptides were relatively unaltered, the levels of Leu-enkephalin were substantially decreased compared to wild-type mice suggesting that preproenkephalin is the major source of Leu-enkephalin. This data also suggests that the lack of ß-endorphin and/or proenkephalin does not lead to a compensatory change in prodynorphin processing. Next, we examined the effect of loss of the endogenous peptides on the regulation of opioid receptor levels and activity in specific regions of the brain. We also compared the receptor levels and activity in males and females and show that the lack of ß-endorphin and/or proenkephalin leads to differential modulation of the three opioid receptors in a region- and gender-specific manner. These results suggest that endogenous opioid peptides are important modulators of the expression and activity of opioid receptors in the brain.


Subject(s)
Analgesics, Opioid/metabolism , Brain/metabolism , Opioid Peptides/metabolism , Receptors, Opioid/agonists , Receptors, Opioid/metabolism , Analgesics, Opioid/pharmacology , Animals , Brain/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Opioid Peptides/pharmacology
6.
Mol Pharmacol ; 95(1): 11-19, 2019 01.
Article in English | MEDLINE | ID: mdl-30348895

ABSTRACT

Signaling by classic analgesics, such as morphine, is governed primarily by the relative abundance of opioid receptors at the cell surface, and this is regulated by receptor delivery to, and retrieval from, the plasma membrane. Although retrieval mechanisms, such as receptor endocytosis, have been extensively investigated, fewer studies have explored mechanisms of receptor maturation and delivery to the plasma membrane. A previous study implicated receptor transporter proteins (RTPs) in the latter process. Since not much is known about regulation of RTP expression, we initiated studies examining the effect of chronic morphine administration on the levels of RTPs in the brain. Among the four RTPs, we detected selective and region-specific changes in RTP4 expression; RTP4 mRNA is significantly upregulated in the hypothalamus compared with other brain regions. We examined whether increased RTP4 expression impacted receptor protein levels and found a significant increase in the abundance of mu opioid receptors (MOPrs) but not other related G protein-coupled receptors (GPCRs, such as delta opioid, CB1 cannabinoid, or D2 dopamine receptors) in hypothalamic membranes from animals chronically treated with morphine. Next, we used a cell culture system to show that RTP4 expression is necessary and sufficient for regulating opioid receptor abundance at the cell surface. Interestingly, selective MOPr-mediated increase in RTP4 expression leads to increases in cell surface levels of MOPr-delta opioid receptor heteromers, and this increase is significantly attenuated by RTP4 small interfering RNA. Together, these results suggest that RTP4 expression is regulated by chronic morphine administration, and this, in turn, regulates opioid receptor cell surface levels and function.


Subject(s)
Molecular Chaperones/metabolism , Morphine/pharmacology , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/pharmacology , Animals , Cell Line, Tumor , Endocytosis/drug effects , Male , Mice , Mice, Inbred C57BL , Narcotic Antagonists/pharmacology
7.
Proc Natl Acad Sci U S A ; 113(21): 6041-6, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27162327

ABSTRACT

Among the opioid receptors, the κ-opioid receptor (κOR) has been gaining considerable attention as a potential therapeutic target for the treatment of complex CNS disorders including depression, visceral pain, and cocaine addiction. With an interest in discovering novel ligands targeting κOR, we searched natural products for unusual scaffolds and identified collybolide (Colly), a nonnitrogenous sesquiterpene from the mushroom Collybia maculata. This compound has a furyl-δ-lactone core similar to that of Salvinorin A (Sal A), another natural product from the plant Salvia divinorum Characterization of the molecular pharmacological properties reveals that Colly, like Sal A, is a highly potent and selective κOR agonist. However, the two compounds differ in certain signaling and behavioral properties. Colly exhibits 10- to 50-fold higher potency in activating the mitogen-activated protein kinase pathway compared with Sal A. Taken with the fact that the two compounds are equipotent for inhibiting adenylyl cyclase activity, these results suggest that Colly behaves as a biased agonist of κOR. Behavioral studies also support the biased agonistic activity of Colly in that it exhibits ∼10-fold higher potency in blocking non-histamine-mediated itch compared with Sal A, and this difference is not seen in pain attenuation by these two compounds. These results represent a rare example of functional selectivity by two natural products that act on the same receptor. The biased agonistic activity, along with an easily modifiable structure compared with Sal A, makes Colly an ideal candidate for the development of novel therapeutics targeting κOR with reduced side effects.


Subject(s)
Agaricales/chemistry , Antipruritics/pharmacology , Diterpenes, Clerodane/pharmacology , MAP Kinase Signaling System/drug effects , Receptors, Opioid, kappa/agonists , Second Messenger Systems/drug effects , Animals , Antipruritics/chemistry , Diterpenes, Clerodane/chemistry , HEK293 Cells , Humans , Mice , Mice, Knockout , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/metabolism
8.
Proc Natl Acad Sci U S A ; 110(40): 16211-6, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24043826

ABSTRACT

Multiple peptide systems, including neuropeptide Y, leptin, ghrelin, and others, are involved with the control of food intake and body weight. The peptide LENSSPQAPARRLLPP (BigLEN) has been proposed to act through an unknown receptor to regulate body weight. In the present study, we used a combination of ligand-binding and receptor-activity assays to characterize a Gαi/o protein-coupled receptor activated by BigLEN in the mouse hypothalamus and Neuro2A cells. We then selected orphan G protein-coupled receptors expressed in the hypothalamus and Neuro2A cells and tested each for activation by BigLEN. G protein-coupled receptor 171 (GPR171) is activated by BigLEN, but not by the C terminally truncated peptide LittleLEN. The four C-terminal amino acids of BigLEN are sufficient to bind and activate GPR171. Overexpression of GPR171 leads to an increase, and knockdown leads to a decrease, in binding and signaling by BigLEN and the C-terminal peptide. In the hypothalamus GPR171 expression complements the expression of BigLEN, and its level and activity are elevated in mice lacking BigLEN. In mice, shRNA-mediated knockdown of hypothalamic GPR171 leads to a decrease in BigLEN signaling and results in changes in food intake and metabolism. The combination of GPR171 shRNA together with neutralization of BigLEN peptide by antibody absorption nearly eliminates acute feeding in food-deprived mice. Taken together, these results demonstrate that GPR171 is the BigLEN receptor and that the BigLEN-GPR171 system plays an important role in regulating responses associated with feeding and metabolism in mice.


Subject(s)
Body Weight/physiology , Feeding Behavior/physiology , Neuropeptides/metabolism , Receptors, G-Protein-Coupled/metabolism , Analysis of Variance , Animals , Blotting, Western , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Immunohistochemistry , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Phosphorylation , Real-Time Polymerase Chain Reaction
9.
Proc Natl Acad Sci U S A ; 110(29): 12072-7, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23818586

ABSTRACT

G protein-coupled receptors play a pivotal role in many physiological signaling pathways. Mounting evidence suggests that G protein-coupled receptors, including opioid receptors, form dimers, and dimerization is necessary for receptor maturation, signaling, and trafficking. However, the physiological role of dimerization in vivo has not been well-explored because of the lack of tools to study these dimers in endogenous systems. To address this problem, we previously generated antibodies to µ-δ opioid receptor (µOR-δOR) dimers and used them to study the pharmacology and signaling by this heteromer. We also showed that the heteromer exhibits restricted distribution in the brain and that its abundance is increased in response to chronic morphine administration. Thus, the µOR-δOR heteromer represents a potentially unique target for the development of therapeutics to treat pain. Here, we report the identification of compounds targeting µOR-δOR heteromers through high-throughput screening of a small-molecule library. These compounds exhibit activity in µOR-δOR cells but not µOR or δOR cells alone. Among them, CYM51010 was found to be a µOR-δOR-biased ligand, because its activity is blocked by the µOR-δOR heteromer antibody. Notably, systemic administration of CYM51010 induced antinociceptive activity similar to morphine, and chronic administration of CYM51010 resulted in lesser antinociceptive tolerance compared with morphine. Taken together, these results suggest that CYM51010, a µOR-δOR-biased ligand, could serve as a scaffold for the development of a unique type (heteromer-biased) of drug that is more potent and without the severe side effects associated with conventional clinical opioids.


Subject(s)
Analgesics/pharmacology , Brain/metabolism , Piperidines/pharmacology , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Analgesics/metabolism , Analysis of Variance , Animals , Antibodies, Monoclonal/metabolism , Cell Line , Dimerization , Drug Tolerance/physiology , High-Throughput Screening Assays , Male , Mice , Mice, Inbred C57BL , Piperidines/metabolism , Radioligand Assay , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Small Molecule Libraries
10.
J Biol Chem ; 289(28): 19613-26, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24847082

ABSTRACT

Most neuroendocrine peptides are generated in the secretory compartment by proteolysis of the precursors at classical cleavage sites consisting of basic residues by well studied endopeptidases belonging to the subtilisin superfamily. In contrast, a subset of bioactive peptides is generated by processing at non-classical cleavage sites that do not contain basic residues. Neither the peptidases responsible for non-classical cleavages nor the compartment involved in such processing has been well established. Members of the endothelin-converting enzyme (ECE) family are considered good candidate enzymes because they exhibit functional properties that are consistent with such a role. In this study we have explored a role for ECE2 in endocytic processing of δ opioid peptides and its effect on modulating δ opioid receptor function by using selective inhibitors of ECE2 that we had identified previously by homology modeling and virtual screening of a library of small molecules. We found that agonist treatment led to intracellular co-localization of ECE2 with δ opioid receptors. Furthermore, selective inhibitors of ECE2 and reagents that increase the pH of the acidic compartment impaired receptor recycling by protecting the endocytosed peptide from degradation. This, in turn, led to a substantial decrease in surface receptor signaling. Finally, we showed that treatment of primary neurons with the ECE2 inhibitor during recycling led to increased intracellular co-localization of the receptors and ECE2, which in turn led to decreased receptor recycling and signaling by the surface receptors. Together, these results support a role for differential modulation of opioid receptor signaling by post-endocytic processing of peptide agonists by ECE2.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Endocytosis/physiology , Metalloendopeptidases/metabolism , Neurons/metabolism , Peptides/metabolism , Proteolysis , Receptors, Opioid, delta/metabolism , Signal Transduction/physiology , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/genetics , CHO Cells , Cricetinae , Cricetulus , Endothelin-Converting Enzymes , Hydrogen-Ion Concentration , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/genetics , Mice , Mice, Knockout , Neurons/cytology , Peptides/genetics , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/genetics
11.
EMBO J ; 30(12): 2350-63, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21540834

ABSTRACT

The mechanism of G protein-coupled receptor (GPCR) signal integration is controversial. While GPCR assembly into hetero-oligomers facilitates signal integration of different receptor types, cross-talk between Gαi- and Gαq-coupled receptors is often thought to be oligomerization independent. In this study, we examined the mechanism of signal integration between the Gαi-coupled type I cannabinoid receptor (CB(1)R) and the Gαq-coupled AT1R. We find that these two receptors functionally interact, resulting in the potentiation of AT1R signalling and coupling of AT1R to multiple G proteins. Importantly, using several methods, that is, co-immunoprecipitation and resonance energy transfer assays, as well as receptor- and heteromer-selective antibodies, we show that AT1R and CB(1)R form receptor heteromers. We examined the physiological relevance of this interaction in hepatic stellate cells from ethanol-administered rats in which CB(1)R is upregulated. We found a significant upregulation of AT1R-CB(1)R heteromers and enhancement of angiotensin II-mediated signalling, as compared with cells from control animals. Moreover, blocking CB(1)R activity prevented angiotensin II-mediated mitogenic signalling and profibrogenic gene expression. These results provide a molecular basis for the pivotal role of heteromer-dependent signal integration in pathology.


Subject(s)
Angiotensin II/chemistry , Angiotensin II/metabolism , Receptor, Angiotensin, Type 1/chemistry , Receptor, Angiotensin, Type 1/metabolism , Receptor, Cannabinoid, CB1/chemistry , Receptor, Cannabinoid, CB1/metabolism , Angiotensin II/genetics , Animals , Cell Communication/genetics , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/chemistry , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , HEK293 Cells , Humans , Ligands , MAP Kinase Signaling System/genetics , Mice , Mice, Inbred BALB C , Rats , Receptor Cross-Talk/physiology , Receptor, Angiotensin, Type 1/genetics , Receptor, Cannabinoid, CB1/genetics
12.
Nat Neurosci ; 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39009835

ABSTRACT

Neuropeptides are ubiquitous in the nervous system. Research into neuropeptides has been limited by a lack of experimental tools that allow for the precise dissection of their complex and diverse dynamics in a circuit-specific manner. Opioid peptides modulate pain, reward and aversion and as such have high clinical relevance. To illuminate the spatiotemporal dynamics of endogenous opioid signaling in the brain, we developed a class of genetically encoded fluorescence sensors based on kappa, delta and mu opioid receptors: κLight, δLight and µLight, respectively. We characterized the pharmacological profiles of these sensors in mammalian cells and in dissociated neurons. We used κLight to identify electrical stimulation parameters that trigger endogenous opioid release and the spatiotemporal scale of dynorphin volume transmission in brain slices. Using in vivo fiber photometry in mice, we demonstrated the utility of these sensors in detecting optogenetically driven opioid release and observed differential opioid release dynamics in response to fearful and rewarding conditions.

13.
Mol Pharmacol ; 81(2): 264-72, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22072818

ABSTRACT

The peripheral δ opioid receptor (DOR) is an attractive target for analgesic drug development. There is evidence that DOR can form heteromers with the κ-opioid receptor (KOR). As drug targets, heteromeric receptors offer an additional level of selectivity and, because of allosteric interactions between protomers, functionality. Here we report that selective KOR antagonists differentially altered the potency and/or efficacy of DOR agonists in primary cultures of adult rat peripheral sensory neurons and in a rat behavioral model of thermal allodynia. In vitro, the KOR antagonist nor-binaltorphimine (nor-BNI) enhanced the potency of [D-Pen(2,5)]-enkephalin (DPDPE), decreased the potency of [D-Ala(2),D-Leu(5)]-enkephalin (DADLE), and decreased the potency and efficacy of 4-[(R)-[(2S,5R)-4-allyl-2,5-dimethylpiperazin-1-yl](3-methoxyphenyl)methyl]-N,N-diethylbenzamide (SNC80) to inhibit prostaglandin E(2) (PGE(2))-stimulated adenylyl cyclase activity. In vivo, nor-BNI enhanced the effect of DPDPE and decreased the effect of SNC80 to inhibit PGE(2)-stimulated thermal allodynia. In contrast to nor-BNI, the KOR antagonist 5'-guanidinonaltrindole (5'-GNTI) reduced the response of DPDPE both in cultured neurons and in vivo. Evidence for DOR-KOR heteromers in peripheral sensory neurons included coimmunoprecipitation of DOR with KOR, a DOR-KOR heteromer selective antibody augmented the antinociceptive effect of DPDPE in vivo, and the DOR-KOR heteromer agonist 6'-GNTI inhibited adenylyl cyclase activity in vitro as well as PGE(2)-stimulated thermal allodynia in vivo. Taken together, these data suggest that DOR-KOR heteromers exist in rat primary sensory neurons and that KOR antagonists can act as modulators of DOR agonist responses most likely through allosteric interactions between the protomers of the DOR-KOR heteromer.


Subject(s)
Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Sensory Receptor Cells/chemistry , Allosteric Regulation , Animals , Cells, Cultured , Drug Design , Hyperalgesia/etiology , Ligands , Protein Multimerization , Rats , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, kappa/agonists
14.
Pain ; 163(7): 1414-1423, 2022 07 01.
Article in English | MEDLINE | ID: mdl-34724682

ABSTRACT

ABSTRACT: Diabetic neuropathy, often associated with diabetes mellitus, is a painful condition with no known effective treatment except glycemic control. Studies with neuropathic pain models report alterations in cannabinoid and opioid receptor expression levels; receptors whose activation induces analgesia. We examined whether interactions between CB1R and opioid receptors could be targeted for the treatment of diabetic neuropathy. For this, we generated antibodies that selectively recognize native CB1R-MOR and CB1R-DOR heteromers using a subtractive immunization strategy. We assessed the levels of CB1R, MOR, DOR, and interacting complexes using a model of streptozotocin-induced diabetic neuropathy and detected increased levels of CB1R, MOR, DOR, and CB1R-MOR complexes compared with those in controls. An examination of G-protein signaling revealed that activity induced by the MOR, but not the DOR agonist, was potentiated by low nanomolar doses of CB1R ligands, including antagonists, suggesting an allosteric modulation of MOR signaling by CB1R ligands within CB1R-MOR complexes. Because the peptide endocannabinoid, hemopressin, caused a significant potentiation of MOR activity, we examined its effect on mechanical allodynia and found that it blocked allodynia in wild-type mice and mice with diabetic neuropathy lacking DOR (but have CB1R-MOR complexes). However, hemopressin does not alter the levels of CB1R-MOR complexes in diabetic mice lacking DOR but increases the levels of CB1R-DOR complexes in diabetic mice lacking MOR. Together, these results suggest the involvement of CB1R-MOR and CB1R-DOR complexes in diabetic neuropathy and that hemopressin could be developed as a potential therapeutic for the treatment of this painful condition.


Subject(s)
Cannabinoids , Diabetes Mellitus, Experimental , Diabetic Neuropathies , Neuralgia , Animals , Diabetes Mellitus, Experimental/complications , Diabetic Neuropathies/drug therapy , Disease Models, Animal , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Ligands , Mice , Neuralgia/drug therapy , Receptors, Opioid , Receptors, Opioid, mu/metabolism
15.
J Neurochem ; 119(2): 294-302, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21692801

ABSTRACT

Ketamine, a clinically relevant drug, has been shown to enhance opioid-induced analgesia and prevent hyperalgesia. However, the molecular mechanisms involved are not clearly understood. As previous studies found that activation of opioid receptors leads to the phosphorylation of mitogen-activated protein kinases, we investigated whether ketamine could modulate µ-opioid receptor (µOR)-mediated ERK1/2 phosphorylation. We find that acute treatment with ketamine enhances (∼2- to 3-fold) the levels of opioid-induced ERK1/2 phosphorylation in recombinant as well as cells endogenously expressing µOR. Interestingly, we find that in the absence of ketamine ERK1/2 signaling is desensitized 10 min after opioid exposure whereas in its presence significant levels (∼3-fold over basal) are detected. In addition, ketamine increases the rate of resensitization of opioid-mediated ERK1/2 signaling (15 min in its presence vs. 30 min in its absence). These results suggest that ketamine increases the effectiveness of opiate-induced signaling by affecting multiple mechanisms. In addition, these effects are observed in heterologous cells expressing µOR suggesting a non-NMDA receptor-mediated action of ketamine. Together this could, in part, account for the observed effects of ketamine on the enhancement of the analgesic effects of opiates as well as in the duration of opiate-induced analgesia.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Receptors, Opioid, mu/drug effects , Signal Transduction/drug effects , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/physiology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Kinetics , Mitogen-Activated Protein Kinases/physiology , Neurons/drug effects , Phosphorylation , Receptors, Opioid, mu/agonists
16.
Proc Natl Acad Sci U S A ; 105(41): 16045-50, 2008 Oct 14.
Article in English | MEDLINE | ID: mdl-18836069

ABSTRACT

Mu opioid receptors are G protein-coupled receptors that mediate the pain-relieving effects of clinically used analgesics, such as morphine. Accumulating evidence shows that mu-delta opioid heterodimers have a pharmacologic profile distinct from those of the mu or delta homodimers. Because the heterodimers exhibit distinct signaling properties, the protein and mechanism regulating their levels have significant effects on morphine-mediated physiology. We report the characterization of RTP4, a Golgi chaperone, as a regulator of the levels of heterodimers at the cell surface. We show that the association with RTP4 protects mu-delta receptors from ubiquitination and degradation. This leads to increases in surface heterodimer levels, thereby affecting signaling. Thus, the oligomeric organization of opioid receptors is controlled by RTP4, and this governs their membrane targeting and functional activity. This work is the first report of the identification of a chaperone involved in the regulation of the biogenesis of a family A GPCR heterodimer. The identification of such factors as RTP4 controlling dimerization will provide insight into the regulation of heterodimers in vivo. This has implications in the modulation of pharmacology of their endogenous ligands, and in the development of drugs with specific therapeutic effects.


Subject(s)
Membrane Transport Proteins/physiology , Molecular Chaperones/physiology , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Animals , Cell Line , Dimerization , Humans , Mice , Ubiquitination
17.
Elife ; 102021 04 28.
Article in English | MEDLINE | ID: mdl-33908346

ABSTRACT

Many signal transduction systems have an apparent redundancy built into them, where multiple physiological agonists activate the same receptors. Whether this is true redundancy, or whether this provides an as-yet unrecognized specificity in downstream signaling, is not well understood. We address this question using the kappa opioid receptor (KOR), a physiologically relevant G protein-coupled receptor (GPCR) that is activated by multiple members of the Dynorphin family of opioid peptides. We show that two related peptides, Dynorphin A and Dynorphin B, bind and activate KOR to similar extents in mammalian neuroendocrine cells and rat striatal neurons, but localize KOR to distinct intracellular compartments and drive different post-endocytic fates of the receptor. Strikingly, localization of KOR to the degradative pathway by Dynorphin A induces sustained KOR signaling from these compartments. Our results suggest that seemingly redundant endogenous peptides can fine-tune signaling by regulating the spatiotemporal profile of KOR signaling.


Subject(s)
Dynorphins/metabolism , Receptors, Opioid, kappa/metabolism , Animals , Corpus Striatum/cytology , Corpus Striatum/metabolism , Neuroendocrine Cells/metabolism , Neurons/metabolism , PC12 Cells , Rats , Receptors, Opioid, kappa/genetics , Signal Transduction
18.
Commun Biol ; 4(1): 238, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33619305

ABSTRACT

Antibodies represent powerful tools to examine signal transduction pathways. Here, we present a strategy integrating multiple state-of-the-art methods to produce, validate, and utilize antibodies. Focusing on understudied synaptic proteins, we generated 137 recombinant antibodies. We used yeast display antibody libraries from the B cells of immunized rabbits, followed by FACS sorting under stringent conditions to identify high affinity antibodies. The antibodies were validated by high-throughput functional screening, and genome editing. Next, we explored the temporal dynamics of signaling in single cells. A subset of antibodies targeting opioid receptors were used to examine the effect of treatment with opiates that have played central roles in the worsening of the 'opioid epidemic.' We show that morphine and fentanyl exhibit differential temporal dynamics of receptor phosphorylation. In summary, high-throughput approaches can lead to the identification of antibody-based tools required for an in-depth understanding of the temporal dynamics of opioid signaling.


Subject(s)
Antibodies/pharmacology , Antibody Specificity , High-Throughput Screening Assays , Protein Kinase C/antagonists & inhibitors , Receptors, Opioid, mu/antagonists & inhibitors , Synapses/drug effects , Analgesics, Opioid/pharmacology , Animals , Antibodies/immunology , Cell Line, Tumor , Enzyme Activation , Fentanyl/pharmacology , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Morphine/pharmacology , Phosphorylation , Protein Kinase C/immunology , Protein Kinase C/metabolism , Rabbits , Receptors, Opioid, mu/immunology , Receptors, Opioid, mu/metabolism , Signal Transduction , Synapses/immunology , Synapses/metabolism , Time Factors
19.
FASEB J ; 23(9): 3020-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19380512

ABSTRACT

Hemopressin (Hp), a 9-residue alpha-hemoglobin-derived peptide, was previously reported to function as a CB(1) cannabinoid receptor antagonist (1) . In this study, we report that mass spectrometry (MS) data from peptidomics analyses of mouse brain extracts identified N-terminally extended forms of Hp containing either three (RVD-Hpalpha) or two (VD-Hpalpha) additional amino acids, as well as a beta-hemoglobin-derived peptide with sequence similarity to that of hemopressin (VD-Hpbeta). Characterization of the alpha-hemoglobin-derived peptides using binding and functional assays shows that in contrast to Hp, which functions as a CB(1) cannabinoid receptor antagonist, both RVD-Hpalpha and VD-Hpalpha function as agonists. Studies examining the increase in the phosphorylation of ERK1/2 levels or release of intracellular Ca(2+) indicate that these peptides activate a signal transduction pathway distinct from that activated by the endocannabinoid, 2-arachidonoylglycerol, or the classic CB(1) agonist, Hu-210. This finding suggests an additional mode of regulation of endogenous cannabinoid receptor activity. Taken together, these results suggest that the CB(1) receptor is involved in the integration of signals from both lipid- and peptide-derived signaling molecules.


Subject(s)
Cannabinoid Receptor Agonists , Peptides/pharmacology , Brain , Cell Line , Hemoglobins/chemistry , Humans , Lipids , Mitogen-Activated Protein Kinase 3/metabolism , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptides/chemical synthesis , Phosphorylation , Signal Transduction
20.
J Cardiovasc Pharmacol ; 56(5): 548-59, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20729750

ABSTRACT

Chemokines are small secreted proteins with chemoattractant properties that play a key role in inflammation, metastasis, and embryonic development. We previously demonstrated a nonchemotactic role for one such chemokine pair, stromal cell-derived factor-1α and its G-protein coupled receptor, CXCR4. Stromal cell-derived factor-1/CXCR4 are expressed on cardiac myocytes and have direct consequences on cardiac myocyte physiology by inhibiting contractility in response to the nonselective ß-adrenergic receptor (ßAR) agonist, isoproterenol. As a result of the importance of ß-adrenergic signaling in heart failure pathophysiology, we investigated the underlying mechanism involved in CXCR4 modulation of ßAR signaling. Our studies demonstrate activation of CXCR4 by stromal cell-derived factor-1 leads to a decrease in ßAR-induced PKA activity as assessed by cAMP accumulation and PKA-dependent phosphorylation of phospholamban, an inhibitor of SERCA2a. We determined CXCR4 regulation of ßAR downstream targets is ß2AR-dependent. We demonstrated a physical interaction between CXCR4 and ß2AR as determined by coimmunoprecipitation, confocal microscopy, and BRET techniques. The CXCR4-ß2AR interaction leads to G-protein signal modulation and suggests the interaction is a novel mechanism for regulating cardiac myocyte contractility. Chemokines are physiologically and developmentally relevant to myocardial biology and represent a novel receptor class of cardiac modulators. The CXCR4-ß2AR complex could represent a hitherto unknown target for therapeutic intervention.


Subject(s)
Myocytes, Cardiac/metabolism , Receptors, Adrenergic, beta-2/physiology , Receptors, CXCR4/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , In Vitro Techniques , Isoproterenol/pharmacology , Phosphorylation , Rats , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL