Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
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.

2.
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
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.
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
5.
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
6.
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
7.
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
8.
J Med Chem ; 63(22): 13618-13637, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33170687

ABSTRACT

In this work, we studied a series of carfentanyl amide-based opioid derivatives targeting the mu opioid receptor (µOR) and the delta opioid receptor (δOR) heteromer as a credible novel target in pain management therapy. We identified a lead compound named MP135 that exhibits high G-protein activity at µ-δ heteromers compared to the homomeric δOR or µOR and low ß-arrestin2 recruitment activity at all three. Furthermore, MP135 exhibits distinct signaling profile, as compared to the previously identified agonist targeting µ-δ heteromers, CYM51010. Pharmacological characterization of MP135 supports the utility of this compound as a molecule that could be developed as an antinociceptive agent similar to morphine in rodents. In vivo characterization reveals that MP135 maintains untoward side effects such as respiratory depression and reward behavior; together, these results suggest that optimization of MP135 is necessary for the development of therapeutics that suppress the classical side effects associated with conventional clinical opioids.


Subject(s)
Fentanyl/analogs & derivatives , Receptors, Opioid, delta/agonists , Analgesics/chemical synthesis , Analgesics/pharmacology , Animals , Cell Line , Fentanyl/chemical synthesis , Fentanyl/pharmacology , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Pain Measurement/drug effects , Pain Measurement/methods , Rats , Rats, Long-Evans , Receptors, Opioid, delta/metabolism
9.
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
10.
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
11.
ACS Pharmacol Transl Sci ; 2(4): 219-229, 2019 Aug 09.
Article in English | MEDLINE | ID: mdl-31565698

ABSTRACT

Cannabinoid 1 (CB1R) and delta opioid receptors (DOR) associate to form heteromers that exhibit distinct pharmacological properties. Not much is known about CB1R-DOR heteromer location or signaling along the pain circuit in either animal models or patients with chemotherapy-induced peripheral neuropathy (CIPN). Here, we use paclitaxel to induce CIPN in mice and confirm the development of mechanical allodynia. Under these conditions, we find significant increases in CB1R-DOR heteromers in the dorsal spinal cord of mice with CIPN as well as in postmortem spinal cords from human subjects with CIPN compared to controls. Next, we investigated receptor signaling in spinal cords of mice with CIPN and found that treatment with a combination of low signaling doses of CB1R and DOR ligands leads to significant enhancement in G-protein activity that could be selectively blocked by the CB1R-DOR antibody. Consistent with this, administration of subthreshold doses of a combination of ligands (CB1R agonist, Hu-210, and DOR agonist, SNC80) leads to significant attenuation of allodynia in mice with CIPN that is not seen with the administration of individual ligands, and this could be blocked by the CB1R-DOR antibody. Together, these results imply that CB1R-DOR heteromers upregulated during CIPN-associated mechanical allodynia could serve as a potential target for treatment of neuropathic pain including CIPN.

12.
Nat Commun ; 10(1): 4112, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31511512

ABSTRACT

Many neuropsychiatric risk genes contribute to epigenetic regulation but little is known about specific chromatin-associated mechanisms governing the formation of neuronal connectivity. Here we show that transcallosal connectivity is critically dependent on C11orf46, a nuclear protein encoded in the chromosome 11p13 WAGR risk locus. C11orf46 haploinsufficiency was associated with hypoplasia of the corpus callosum. C11orf46 knockdown disrupted transcallosal projections and was rescued by wild type C11orf46 but not the C11orf46R236H mutant associated with intellectual disability. Multiple genes encoding key regulators of axonal development, including Sema6a, were hyperexpressed in C11orf46-knockdown neurons. RNA-guided epigenetic editing of Sema6a gene promoters via a dCas9-SunTag system with C11orf46 binding normalized SEMA6A expression and rescued transcallosal dysconnectivity via repressive chromatin remodeling by the SETDB1 repressor complex. Our study demonstrates that interhemispheric communication is sensitive to locus-specific remodeling of neuronal chromatin, revealing the therapeutic potential for shaping the brain's connectome via gene-targeted designer activators and repressor proteins.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Corpus Callosum/metabolism , Epigenesis, Genetic , Gene Editing , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Semaphorins/genetics , Animals , Axons/metabolism , Epigenome , Gene Expression Regulation , Genetic Predisposition to Disease , HEK293 Cells , Histone-Lysine N-Methyltransferase , Humans , Mice, Inbred C57BL , Nerve Net/metabolism , Neurites/metabolism , Phenotype , Protein Binding , Protein Methyltransferases/metabolism
13.
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
14.
15.
Sci Rep ; 7(1): 14781, 2017 11 01.
Article in English | MEDLINE | ID: mdl-29093454

ABSTRACT

Intracellular peptides generated by limited proteolysis are likely to function inside and outside cells and could represent new possibilities for drug development. Here, we used several conformational-sensitive antibodies targeting G-protein coupled receptors to screen for novel pharmacological active peptides. We find that one of these peptides, DITADDEPLT activates cannabinoid type 1 receptors. Single amino acid modifications identified a novel peptide, DIIADDEPLT (Pep19), with slightly better inverse agonist activity at cannabinoid type 1 receptors. Pep19 induced uncoupling protein 1 expression in both white adipose tissue and 3T3-L1 differentiated adipocytes; in the latter, Pep19 activates pERK1/2 and AKT signaling pathways. Uncoupling protein 1 expression induced by Pep19 in 3T3-L1 differentiated adipocytes is blocked by AM251, a cannabinoid type 1 receptors antagonist. Oral administration of Pep19 into diet-induced obese Wistar rats significantly reduces adiposity index, whole body weight, glucose, triacylglycerol, cholesterol and blood pressure, without altering heart rate; changes in the number and size of adipocytes were also observed. Pep19 has no central nervous system effects as suggested by the lack of brain c-Fos expression, cell toxicity, induction of the cannabinoid tetrad, depressive- and anxiety-like behaviors. Therefore, Pep19 has several advantages over previously identified peripherally active cannabinoid compounds, and could have clinical applications.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, White/metabolism , MAP Kinase Signaling System/drug effects , Obesity/drug therapy , Peptides , Receptor, Cannabinoid, CB1/antagonists & inhibitors , 3T3-L1 Cells , Adipocytes/pathology , Adipose Tissue, White/pathology , Animals , Brain/metabolism , Brain/pathology , Diet, High-Fat/adverse effects , Male , Mice , Obesity/chemically induced , Obesity/metabolism , Obesity/pathology , Peptides/chemistry , Peptides/pharmacology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/metabolism
16.
PLoS One ; 12(11): e0187306, 2017.
Article in English | MEDLINE | ID: mdl-29091950

ABSTRACT

The N-terminal region of G protein-coupled receptors can be efficiently targeted for the generation of receptor-selective antibodies. These antibodies are useful for the biochemical characterization of the receptors. In this study, we developed a set of criteria to select the optimal epitope and applied them to generate antibodies to the N-terminal region of 34 different G protein-coupled receptors. The antibody characterization revealed that a subset of antibodies exhibited increased recognition of the receptor following agonist treatment and this increase could be blocked by treatment with the receptor antagonist. An analysis of the epitopes showed that those antibodies that exhibit increased recognition are on average twelve residues long, have an overall net negative charge and are enriched in aspartic and glutamic acids. These antibodies are useful since they facilitate studies examining dose dependent increases in recognition of receptors in heterologous cells as well as in native tissue. Another interesting use of these antibodies is that they facilitate measuring changes in receptor recognition in brain following peripheral drug administration; for example, systemic administration of cocaine, a blocker of dopamine transporter that increases local dopamine levels at the synapse, was found to lead to increases in antibody recognition of dopamine receptors in the brain. Taken together these studies, in addition to describing novel tools to study native receptors, provide a framework for the generation of antibodies to G protein-coupled receptors that can detect ligand-induced conformational changes.


Subject(s)
Antibodies/immunology , Receptors, G-Protein-Coupled/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Epitopes/chemistry , Epitopes/immunology , HEK293 Cells , Humans , Male , Protein Conformation , Rabbits , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry
17.
Sci Signal ; 9(430): ra55, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27245612

ABSTRACT

Several neuropeptide systems in the hypothalamus, including neuropeptide Y and agouti-related protein (AgRP), control food intake. Peptides derived from proSAAS, a precursor implicated in the regulation of body weight, also control food intake. GPR171 is a heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptor (GPCR) for BigLEN (b-LEN), a peptide derived from proSAAS. To facilitate studies exploring the physiological role of GPR171, we sought to identify small-molecule ligands for this receptor by performing a virtual screen of a compound library for interaction with a homology model of GPR171. We identified MS0015203 as an agonist of GPR171 and demonstrated the selectivity of MS0015203 for GPR171 by testing the binding of this compound to 80 other membrane proteins, including family A GPCRs. Reducing the expression of GPR171 by shRNA (short hairpin RNA)-mediated knockdown blunted the cellular and tissue response to MS0015203. Peripheral injection of MS0015203 into mice increased food intake and body weight, and these responses were significantly attenuated in mice with decreased expression of GPR171 in the hypothalamus. Together, these results suggest that MS0015203 is a useful tool to probe the pharmacological and functional properties of GPR171 and that ligands targeting GPR171 may eventually lead to therapeutics for food-related disorders.


Subject(s)
Anilides/pharmacology , Eating/drug effects , Phthalic Acids/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Animals , Appetite , Body Weight , CHO Cells , Calcium/metabolism , Cell Line, Tumor , Cricetinae , Cricetulus , Feeding Behavior , Gene Expression Regulation , Ligands , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/pharmacology , Neuropeptides , Peptides/pharmacology , Protein Binding , Rats , Signal Transduction
18.
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
19.
Sci Signal ; 9(425): ra43, 2016 04 26.
Article in English | MEDLINE | ID: mdl-27117253

ABSTRACT

PEN is an abundant peptide in the brain that has been implicated in the regulation of feeding. We identified a receptor for PEN in mouse hypothalamus and Neuro2A cells. PEN bound to and activated GPR83, a G protein (heterotrimeric guanine nucleotide)-binding protein)-coupled receptor (GPCR). Reduction of GPR83 expression in mouse brain and Neuro2A cells reduced PEN binding and signaling, consistent with GPR83 functioning as the major receptor for PEN. In some brain regions, GPR83 colocalized with GPR171, a GPCR that binds the neuropeptide bigLEN, another neuropeptide that is involved in feeding and is generated from the same precursor protein as is PEN. Coexpression of these two receptors in cell lines altered the signaling properties of each receptor, suggesting a functional interaction. Our data established PEN as a neuropeptide that binds GPR83 and suggested that these two ligand-receptor systems-PEN-GPR83 and bigLEN-GPR171-may be functionally coupled in the regulation of feeding.


Subject(s)
Hypothalamus/metabolism , Neuropeptide Y/metabolism , Receptors, G-Protein-Coupled/metabolism , Adenosine Triphosphate/metabolism , Animals , Appetite Regulation/physiology , Blotting, Western , CHO Cells , Cell Membrane/metabolism , Cells, Cultured , Cricetulus , HEK293 Cells , Humans , Male , Mice , Phosphorylation , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics
20.
Front Pharmacol ; 5: 268, 2014.
Article in English | MEDLINE | ID: mdl-25520661

ABSTRACT

Over the last decade an increasing number of studies have focused on the ability of G protein-coupled receptors to form heteromers and explored how receptor heteromerization modulates the binding, signaling and trafficking properties of individual receptors. Most of these studies were carried out in heterologous cells expressing epitope tagged receptors. Very little information is available about the in vivo physiological role of G protein-coupled receptor heteromers due to a lack of tools to detect their presence in endogenous tissue. Recent advances such as the generation of mouse models expressing fluorescently labeled receptors, of TAT based peptides that can disrupt a given heteromer pair, or of heteromer-selective antibodies that recognize the heteromer in endogenous tissue have begun to elucidate the physiological and pathological roles of receptor heteromers. In this review we have focused on heteromer-selective antibodies and describe how a subtractive immunization strategy can be successfully used to generate antibodies that selectively recognize a desired heteromer pair. We also describe the uses of these antibodies to detect the presence of heteromers, to study their properties in endogenous tissues, and to monitor changes in heteromer levels under pathological conditions. Together, these findings suggest that G protein-coupled receptor heteromers represent unique targets for the development of drugs with reduced side-effects.

SELECTION OF CITATIONS
SEARCH DETAIL
...