Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell ; 171(5): 1165-1175.e13, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29149605

ABSTRACT

Biased agonism has been proposed as a means to separate desirable and adverse drug responses downstream of G protein-coupled receptor (GPCR) targets. Herein, we describe structural features of a series of mu-opioid-receptor (MOR)-selective agonists that preferentially activate receptors to couple to G proteins or to recruit ßarrestin proteins. By comparing relative bias for MOR-mediated signaling in each pathway, we demonstrate a strong correlation between the respiratory suppression/antinociception therapeutic window in a series of compounds spanning a wide range of signaling bias. We find that ßarrestin-biased compounds, such as fentanyl, are more likely to induce respiratory suppression at weak analgesic doses, while G protein signaling bias broadens the therapeutic window, allowing for antinociception in the absence of respiratory suppression.


Subject(s)
Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Receptors, Opioid, mu/agonists , Animals , Fentanyl/administration & dosage , GTP-Binding Proteins/metabolism , Mice , Morphine/administration & dosage , Receptors, Opioid, mu/chemistry , Respiratory System/drug effects , Signal Transduction , beta-Arrestins/metabolism
2.
J Pain ; 18(12): 1526-1541, 2017 12.
Article in English | MEDLINE | ID: mdl-28939014

ABSTRACT

Activation of the mu-opioid receptor provides the gold standard for pain relief, but most opioids used clinically have adverse effects that have contributed to an epidemic of overdose deaths. We recently characterized mu-opioid receptor selective endomorphin (EM) analogs that provide potent antinociception with reduction or absence of a number of side effects of traditionally prescribed opioids including abuse liability, respiratory depression, motor impairment, tolerance, and inflammation. The current study explores the effectiveness of these EM analogs relative to morphine in four major pain models by intrathecal as well as intravenous administration in male Sprague Dawley rats and subcutaneous administration in male CD-1 mice. In the spared nerve injury model of neuropathic pain, mechanical allodynia and mechanical hyperalgesia were assessed with von Frey and Randall-Selitto tests, respectively. In the paw incision model of postoperative pain, von Frey testing was used to assess mechanical allodynia and thermal hyperalgesia was evaluated with Hargreaves testing. In the Complete Freund's Adjuvant model of inflammatory pain, thermal hyperalgesia was assessed using Hargreaves testing. In CD-1 mice, visceral pain was assessed with the acetic acid writhing test. In all cases, EM analogs had equal or greater potency and longer duration of action relative to morphine. The data suggest that EM analogs, particularly analog 4 (ZH853), could provide effective therapy for a diverse spectrum of pain conditions with low risk of adverse side effects compared with currently used opioids such as morphine. PERSPECTIVE: Novel EM analogs show equal or greater potency and effectiveness relative to morphine in multiple pain models. Together with substantially reduced side effects, including abuse liability, the compounds show promise for addressing the critical need for effective pain relief as well as reducing the opioid overdose epidemic.


Subject(s)
Analgesics, Opioid/pharmacology , Hyperalgesia/drug therapy , Morphine/pharmacology , Neuralgia/drug therapy , Nociceptive Pain/drug therapy , Oligopeptides/analysis , Pain, Postoperative/drug therapy , Peptides, Cyclic/pharmacology , Visceral Pain/drug therapy , Analgesics, Opioid/administration & dosage , Animals , Disease Models, Animal , Inflammation/complications , Injections, Intravenous , Injections, Spinal , Male , Mice , Nociceptive Pain/etiology , Peptides, Cyclic/administration & dosage , Rats , Rats, Sprague-Dawley
3.
Sci Signal ; 9(456): ra117, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27899527

ABSTRACT

Agonists targeting the kappa opioid receptor (KOR) have been promising therapeutic candidates because of their efficacy for treating intractable itch and relieving pain. Unlike typical opioid narcotics, KOR agonists do not produce euphoria or lead to respiratory suppression or overdose. However, they do produce dysphoria and sedation, side effects that have precluded their clinical development as therapeutics. KOR signaling can be fine-tuned to preferentially activate certain pathways over others, such that agonists can bias signaling so that the receptor signals through G proteins rather than other effectors such as ßarrestin2. We evaluated a newly developed G protein signaling-biased KOR agonist in preclinical models of pain, pruritis, sedation, dopamine regulation, and dysphoria. We found that triazole 1.1 retained the antinociceptive and antipruritic efficacies of a conventional KOR agonist, yet it did not induce sedation or reductions in dopamine release in mice, nor did it produce dysphoria as determined by intracranial self-stimulation in rats. These data demonstrated that biased agonists may be used to segregate physiological responses downstream of the receptor. Moreover, the findings suggest that biased KOR agonists may present a means to treat pain and intractable itch without the side effects of dysphoria and sedation and with reduced abuse potential.


Subject(s)
Pain/drug therapy , Pruritus/drug therapy , Receptors, Opioid, kappa/agonists , Triazoles/pharmacology , Animals , Male , Mice , Pain/metabolism , Pruritus/metabolism , Rats , Rats, Inbred F344 , Receptors, Opioid, kappa/metabolism , Triazoles/chemistry , beta-Arrestin 2/metabolism
4.
Sci Signal ; 9(454): ra111, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27902448

ABSTRACT

The striatum of the brain coordinates motor function. Dopamine-related drugs may be therapeutic to patients with striatal neurodegeneration, such as Huntington's disease (HD) and Parkinson's disease (PD), but these drugs have unwanted side effects. In addition to stimulating the release of norepinephrine, amphetamines, which are used for narcolepsy and attention-deficit/hyperactivity disorder (ADHD), trigger dopamine release in the striatum. The guanosine triphosphatase Ras homolog enriched in the striatum (Rhes) inhibits dopaminergic signaling in the striatum, is implicated in HD and L-dopa-induced dyskinesia, and has a role in striatal motor control. We found that the guanine nucleotide exchange factor RasGRP1 inhibited Rhes-mediated control of striatal motor activity in mice. RasGRP1 stabilized Rhes, increasing its synaptic accumulation in the striatum. Whereas partially Rhes-deficient (Rhes+/-) mice had an enhanced locomotor response to amphetamine, this phenotype was attenuated by coincident depletion of RasGRP1. By proteomic analysis of striatal lysates from Rhes-heterozygous mice with wild-type or partial or complete knockout of Rasgrp1, we identified a diverse set of Rhes-interacting proteins, the "Rhesactome," and determined that RasGRP1 affected the composition of the amphetamine-induced Rhesactome, which included PDE2A (phosphodiesterase 2A; a protein associated with major depressive disorder), LRRC7 (leucine-rich repeat-containing 7; a protein associated with bipolar disorder and ADHD), and DLG2 (discs large homolog 2; a protein associated with chronic pain). Thus, this Rhes network provides insight into striatal effects of amphetamine and may aid the development of strategies to treat various neurological and psychological disorders associated with the striatal dysfunction.


Subject(s)
Amphetamine/pharmacology , Behavior, Animal/drug effects , GTP-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Locomotion/drug effects , Signal Transduction/drug effects , Animals , GTP-Binding Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Mice, Mutant Strains , Rats , Signal Transduction/genetics
5.
Neuropharmacology ; 105: 215-227, 2016 06.
Article in English | MEDLINE | ID: mdl-26748051

ABSTRACT

Opioids acting at the mu opioid receptor (MOR) are the most effective analgesics, however adverse side effects severely limit their use. Of particular importance, abuse liability results in major medical, societal, and economic problems, respiratory depression is the cause of fatal overdoses, and tolerance complicates treatment and increases the risk of side effects. Motor and cognitive impairment are especially problematic for older adults. Despite the host of negative side effects, opioids such as morphine are commonly used for acute and chronic pain conditions. Separation of analgesia from unwanted effects has long been an unmet goal of opioid research. Novel MOR agonist structures may prove critical for greater success. Here we tested metabolically stable analogs of the endomorphins, endogenous opioids highly selective for the MOR. Compared to morphine, the analogs showed dramatically improved analgesia-to-side-effect ratios. At doses providing equal or greater antinociception than morphine in the rat, the analogs showed reduced a) respiratory depression, b) impairment of motor coordination, c) tolerance and hyperalgesia, d) glial p38/CGRP/P2X7 receptor signaling, and e) reward/abuse potential in both conditioned place preference and self-administration tests. Differential effects on glial activation indicate a mechanism for the relative lack of side effects by the analogs compared to morphine. The results suggest that endomorphin analogs described here could provide gold standard pain relief mediated by selective MOR activation, but with remarkably safer side effect profiles compared to opioids like morphine.


Subject(s)
Analgesics, Opioid/toxicity , Dyskinesia, Drug-Induced/physiopathology , Macrophage Activation/drug effects , Microglia/drug effects , Morphine/toxicity , Respiratory Insufficiency/chemically induced , Substance-Related Disorders/psychology , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/pharmacology , Animals , Blood-Brain Barrier/metabolism , Conditioning, Operant/drug effects , Drug Tolerance , Hyperalgesia/chemically induced , Hyperalgesia/psychology , Male , Mice , Pain Measurement/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/drug effects , Respiratory Insufficiency/physiopathology
6.
Neuropharmacology ; 99: 600-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26318102

ABSTRACT

The kappa opioid receptor (KOR) is involved in mediating pruritus; agonists targeting this receptor have been used to treat chronic intractable itch. Conversely, antagonists induce an itch response at the site of injection. As a G protein-coupled receptor (GPCR), the KOR has potential for signaling via G proteins and ßarrestins, however, it is not clear which of these pathways are involved in the KOR modulation of itch. In this study asked whether the actions of KOR in pruritus involve ßarrestins by using ßarrestin2 knockout (ßarr2-KO) mice as well as a recently described biased KOR agonist that biases receptor signaling toward G protein pathways over ßarrestin2 recruitment. We find that the KOR antagonists nor-binaltorphimine (NorBNI) and 5'-guanidinonaltrindole (5'GNTI) induce acute pruritus in C57BL/6J mice, with reduced effects in KOR-KO mice. ßArr2-KO mice display less of a response to KOR antagonist-induced itch compared to wild types, however no genotype differences are observed from chloroquine phosphate (CP)-induced itch, suggesting that the antagonists may utilize a KOR-ßarrestin2 dependent mechanism. The KOR agonist U50,488H was equally effective in both WT and ßarr2-KO mice in suppressing CP-induced itch. Furthermore, the G protein biased agonist, Isoquinolinone 2.1 was as effective as U50,488H in suppressing the itch response induced by KOR antagonist NorBNI or CP in C57BL/6J mice. Together these data suggest that the antipruritic effects of KOR agonists may not require ßarrestins.


Subject(s)
Analgesics, Opioid/pharmacology , Arrestins/metabolism , Pruritus/drug therapy , Receptors, Opioid, kappa/agonists , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Animals , Arrestins/genetics , Chloroquine/analogs & derivatives , Disease Models, Animal , Dose-Response Relationship, Drug , Guanidines , Isoquinolines/pharmacology , Male , Mice, Inbred C57BL , Mice, Knockout , Morphinans , Motor Activity/drug effects , Motor Activity/physiology , Naltrexone/analogs & derivatives , Pruritus/metabolism , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, kappa/metabolism , beta-Arrestins
7.
Pain ; 156(3): 469-482, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25599238

ABSTRACT

Repeated administration of peroxisome proliferator-activated receptor gamma (PPARγ) agonists reduces neuropathic pain-like behavior and associated changes in glial activation in the spinal cord dorsal horn. As PPARγ is a nuclear receptor, sustained changes in gene expression are widely believed to be the mechanism of pain reduction. However, we recently reported that a single intrathecal (i.t.) injection of pioglitazone, a PPARγ agonist, reduced hyperalgesia within 30 minutes, a time frame that is typically less than that required for genomic mechanisms. To determine the very rapid antihyperalgesic actions of PPARγ activation, we administered pioglitazone to rats with spared nerve injury and evaluated hyperalgesia. Pioglitazone inhibited hyperalgesia within 5 minutes of injection, consistent with a nongenomic mechanism. Systemic or i.t. administration of GW9662, a PPARγ antagonist, inhibited the antihyperalgesic actions of intraperitoneal or i.t. pioglitazone, suggesting a spinal PPARγ-dependent mechanism. To further address the contribution of nongenomic mechanisms, we blocked new protein synthesis in the spinal cord with anisomycin. When coadministered intrathecally, anisomycin did not change pioglitazone antihyperalgesia at an early 7.5-minute time point, further supporting a rapid nongenomic mechanism. At later time points, anisomycin reduced pioglitazone antihyperalgesia, suggesting delayed recruitment of genomic mechanisms. Pioglitazone reduction of spared nerve injury-induced increases in GFAP expression occurred more rapidly than expected, within 60 minutes. We are the first to show that activation of spinal PPARγ rapidly reduces neuropathic pain independent of canonical genomic activity. We conclude that acute pioglitazone inhibits neuropathic pain in part by reducing astrocyte activation and through both genomic and nongenomic PPARγ mechanisms.


Subject(s)
Astrocytes/drug effects , Hypoglycemic Agents/therapeutic use , Neuralgia/drug therapy , PPAR gamma/metabolism , Thiazolidinediones/therapeutic use , Analysis of Variance , Animal Diseases , Animals , Area Under Curve , Astrocytes/metabolism , Capsaicin/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Injections, Intraventricular , Male , Neuralgia/complications , Neuralgia/pathology , Nociception/drug effects , Oncogene Proteins v-fos/metabolism , Pain Threshold/drug effects , Pioglitazone , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Rats , Rats, Sprague-Dawley , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Dorsal Horn/pathology
8.
Stem Cells Transl Med ; 1(7): 557-65, 2012 Jul.
Article in English | MEDLINE | ID: mdl-23197860

ABSTRACT

Mesenchymal stem cells (MSCs) are very attractive candidates in cell-based strategies that target inflammatory diseases. Preclinical animal studies and many clinical trials have demonstrated that human MSCs can be safely administered and that they modify the inflammatory process in the targeted injured tissue. Our laboratory developed a novel method that optimizes the anti-inflammatory effects of MSCs. We termed the cells prepared by this method MSC2. In this study, we determined the effects of MSC2-based therapies on an inflammation-linked painful diabetic peripheral neuropathy (pDPN) mouse model. Streptozotocin-induced diabetic mice were treated with conventionally prepared MSCs, MSC2, or vehicle at three specific time points. Prior to each treatment, responses to radiant heat (Hargreaves) and mechanical stimuli (von Frey) were measured. Blood serum from each animal was collected at the end of the study to compare levels of inflammatory markers between the treatment groups. We observed that MSC2-treated mice had significant improvement in behavioral assays compared with the vehicle and MSC groups, and moreover these responses did not differ from the observations seen in the healthy wild-type control group. Mice treated with conventional MSCs showed significant improvement in the radiant heat assay, but not in the von Frey test. Additionally, mice treated with MSC2 had decreased serum levels in many proinflammatory cytokines compared with the values measured in the MSC- or vehicle-treated groups. These findings indicate that MSC2-based therapy is a new anti-inflammatory treatment to consider in the management of pDPN.


Subject(s)
Cytokines/blood , Diabetes Mellitus, Experimental/therapy , Diabetic Neuropathies/therapy , Inflammation Mediators/blood , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Animals , Biomarkers/blood , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/pathology , Diabetic Neuropathies/blood , Diabetic Neuropathies/pathology , Humans , Inflammation/blood , Inflammation/pathology , Inflammation/therapy , Male , Mesenchymal Stem Cells/pathology , Mice , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...