Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
FASEB J ; 38(8): e23603, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38648368

ABSTRACT

Recent evidence suggests that chronic exposure to opioid analgesics such as morphine disrupts the intestinal epithelial layer and causes intestinal dysbiosis. Depleting gut bacteria can preclude the development of tolerance to opioid-induced antinociception, suggesting an important role of the gut-brain axis in mediating opioid effects. The mechanism underlying opioid-induced dysbiosis, however, remains unclear. Host-produced antimicrobial peptides (AMPs) are critical for the integrity of the intestinal epithelial barrier as they prevent the pathogenesis of the enteric microbiota. Here, we report that chronic morphine or fentanyl exposure reduces the antimicrobial activity in the ileum, resulting in changes in the composition of bacteria. Fecal samples from morphine-treated mice had increased levels of Akkermansia muciniphila with a shift in the abundance ratio of Firmicutes and Bacteroidetes. Fecal microbial transplant (FMT) from morphine-naïve mice or oral supplementation with butyrate restored (a) the antimicrobial activity, (b) the expression of the antimicrobial peptide, Reg3γ, (c) prevented the increase in intestinal permeability and (d) prevented the development of antinociceptive tolerance in morphine-dependent mice. Improved epithelial barrier function with FMT or butyrate prevented the enrichment of the mucin-degrading A. muciniphila in morphine-dependent mice. These data implicate impairment of the antimicrobial activity of the intestinal epithelium as a mechanism by which opioids disrupt the microbiota-gut-brain axis.


Subject(s)
Analgesics, Opioid , Dysbiosis , Fentanyl , Gastrointestinal Microbiome , Intestinal Mucosa , Mice, Inbred C57BL , Morphine , Animals , Morphine/pharmacology , Mice , Dysbiosis/chemically induced , Dysbiosis/microbiology , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Male , Fentanyl/pharmacology , Analgesics, Opioid/pharmacology , Brain-Gut Axis/drug effects , Fecal Microbiota Transplantation , Pancreatitis-Associated Proteins/metabolism , Akkermansia/drug effects , Antimicrobial Peptides/pharmacology , Bacteroidetes/drug effects
2.
Bioorg Chem ; 148: 107489, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38797065

ABSTRACT

The number of opioid-related overdose deaths and individuals that have suffered from opioid use disorders have significantly increased over the last 30 years. FDA approved maintenance therapies to treat opioid use disorder may successfully curb drug craving and prevent relapse but harbor adverse effects that reduce patient compliance. This has created a need for new chemical entities with improved patient experience. Previously our group reported a novel lead compound, NAT, a mu-opioid receptor antagonist that potently antagonized the antinociception of morphine and showed significant blood-brain barrier permeability. However, NAT belongs to thiophene containing compounds which are known structural alerts for potential oxidative metabolism. To overcome this, 15 NAT derivatives with various substituents at the 5'-position of the thiophene ring were designed and their structure-activity relationships were studied. These derivatives were characterized for their binding affinity, selectivity, and functional activity at the mu opioid receptor and assessed for their ability to antagonize the antinociceptive effects of morphine in vivo. Compound 12 showed retention of the basic pharmacological attributes of NAT while improving the withdrawal effects that were experienced in opioid-dependent mice. Further studies will be conducted to fully characterize compound 12 to examine whether it would serve as a new lead for opioid use disorder treatment and management.


Subject(s)
Receptors, Opioid, mu , Animals , Structure-Activity Relationship , Mice , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/antagonists & inhibitors , Humans , Molecular Structure , Thiophenes/chemistry , Thiophenes/pharmacology , Thiophenes/chemical synthesis , Thiophenes/therapeutic use , Male , Dose-Response Relationship, Drug , Analgesics, Opioid/pharmacology , Analgesics, Opioid/chemistry , Narcotic Antagonists/pharmacology , Narcotic Antagonists/chemistry , Morphine/pharmacology
3.
Physiology (Bethesda) ; 36(5): 315-323, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34431418

ABSTRACT

Bidirectional interactions of the gut epithelium with commensal bacteria are critical for maintaining homeostasis within the gut. Chronic opioid exposure perturbs gut homeostasis through a multitude of neuro-immune-epithelial mechanisms, resulting in the development of analgesic tolerance, a major underpinning of the current opioid crisis. Differences in molecular mechanisms of opioid tolerance between the enteric and central pain pathways pose a significant challenge for managing chronic pain without untoward gastrointestinal effects.


Subject(s)
Gastrointestinal Microbiome , Opioid Epidemic , Analgesics, Opioid/adverse effects , Drug Tolerance , Humans , Intestinal Mucosa
4.
J Pharmacol Exp Ther ; 370(3): 504-513, 2019 09.
Article in English | MEDLINE | ID: mdl-31248978

ABSTRACT

Opioids are highly effective analgesics, however, their therapeutic use is limited by adverse effects that include respiratory depression, dependence, and tolerance. Inflammation has been implicated as a significant driver for the development of tolerance to opioids. Recent studies show that chronic morphine in mice results in gut microbial dysbiosis and inflammation in the colon. In the present study, we examined whether colonic inflammation results in tolerance to the antinociceptive effects of morphine. Colonic inflammation was induced in mice by intrarectal administration of 2,4,6-trinitro-benzene sulfonic acid. The development of antinociceptive tolerance was determined by warm-water tail-immersion assay in mice implanted with 25-, 50-, or 75-mg morphine pellet. Colonic inflammation significantly enhanced the rate at which tolerance developed in each cohort of chronic morphine-treated mice. At the lowest dose of morphine pellet (25 mg), antinociceptive tolerance only developed in the presence of colonic inflammation, whereas in 50- and 75-mg pelleted mice, tolerance developed faster in the inflamed animals than in the noninflamed mice. The enhanced antinociceptive tolerance was attenuated with daily administration of peripheral opioid receptor antagonist, 6ß-N-heterocyclic-substituted naltrexamine derivative [17-cyclopropylmethyl-3,14ß-dihydroxy-4,5α-epoxy-6ß-[(4'pyridyl)acetamido]morphinan (NAP)], irrespective of colonic inflammation. Collectively, these findings show that the rate of tolerance to morphine antinociception is exaggerated in the presence of colonic inflammation, and tolerance is prevented by a peripheral µ-opioid receptor antagonist. These studies suggest a peripheral component to the development of antinociceptive tolerance to opioids. Furthermore, peripherally selective opioid antagonists may be useful adjuncts in opioid-based pain management. SIGNIFICANCE STATEMENT: This study supports the notion that inflammation influences the development of antinociceptive tolerance to chronic morphine exposure. We found that, in the presence of colonic inflammation, the rate of development of tolerance to the antinociceptive effects of morphine increased. We also found that treatment with a peripheral opioid receptor antagonist prevented morphine antinociceptive tolerance. Increasing opioid intake during an inflammatory state would result in decreased analgesia and enhanced analgesic tolerance, which puts patients with inflammatory bowel diseases, inflammatory joint diseases, and sickle cell anemia at risk for heavy opioid use.


Subject(s)
Analgesics/pharmacology , Colitis/drug therapy , Colitis/metabolism , Drug Tolerance , Morphine/pharmacology , Receptors, Opioid/metabolism , Analgesics/therapeutic use , Animals , Colitis/pathology , Colon/drug effects , Colon/metabolism , Colon/pathology , Disease Models, Animal , Male , Mice , Morphine/therapeutic use
5.
Biomed Chromatogr ; 33(4): e4465, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30548295

ABSTRACT

Carbenoxolone is a derivative of glycyrrhetinic acid found in the root of Glycyrrhiza glabra, colloquially known as licorice. It has been used as a treatment for peptic and oral ulcers. In recent years, carbenoxolone has been utilized in basic research for its ability to block gap junctional communication. Better understanding the distribution of carbenoxolone after systemic administration can lead to a better understanding of its potential sites of action. Presented is an ultra high-performance liquid chromatography tandem mass spectrometer (UHPLC-MS/MS) method for the identification and quantification of carbenoxolone in mouse blood and brain tissue. Twenty mice were injected intraperitoneally with 25 mg/kg carbenoxolone and brain tissue and blood were collected for analysis. Blood concentrations (mean ± SD) at 15, 30, 60 and 120 min were determined to be (n = 5) 5394 ± 778, 2636 ± 836, 1564 ± 541 and 846 ± 252 ng/mL, respectively. Brain concentrations (mean ± SD) at 15, 30, 60 and 120 mins were determined to be (n = 5) 171 ± 62, 102 ± 35, 55 ± 10 and 27 ± 9 ng/g, respectively. The analysis of these specimens at the four different time points resulted in blood and brain half-lives in mice of ~43 and 41 min, respectively. The UHPLC-MS/MS method was determined to be sensitive and robust for quantification of carbenoxolone.


Subject(s)
Brain Chemistry/physiology , Carbenoxolone/analysis , Chromatography, High Pressure Liquid/methods , Tandem Mass Spectrometry/methods , Animals , Carbenoxolone/administration & dosage , Carbenoxolone/chemistry , Carbenoxolone/pharmacokinetics , Drug Stability , Injections, Intraperitoneal , Limit of Detection , Linear Models , Male , Mice , Reproducibility of Results
6.
Mol Pharmacol ; 93(5): 417-426, 2018 05.
Article in English | MEDLINE | ID: mdl-29467238

ABSTRACT

Oxycodone is a semisynthetic opioid compound that is widely prescribed, used, and abused today, and has a well-established role in shaping the current opioid epidemic. Previously, we have shown that tolerance develops to the antinociceptive and respiratory depressive effects of oxycodone in mice, and that a moderate dose of acute ethanol or a protein kinase C (PKC) inhibitor reversed that tolerance. To investigate further if tolerance was occurring through neuronal mechanisms, our aims for this study were to assess the effects of acute and prolonged oxycodone in isolated dorsal root ganglia (DRG) neurons and to determine if this tolerance was reversed by either ethanol or a PKC inhibitor. We found that an acute exposure to 3 µM oxycodone reduced neuronal excitability, as measured by increased threshold potentials and reduced action potential amplitude, without eliciting measurable changes in resting membrane potential. Exposure to 10 µM oxycodone for 18-24 hours prevented oxycodone's effect on neuronal excitability, indicative of tolerance development. The development of opioid tolerance was mitigated in DRG neurons from ß-arrestin 2 knockout mice. Oxycodone tolerance was reversed in isolated DRG neurons by the acute application of either ethanol (20 mM) or the PKC inhibitor, bisindolylmaleimide XI hydrochloride (Bis XI), when a challenge of 3 µM oxycodone significantly reduced neuronal excitability following prolonged exposure. Through these studies, we concluded that oxycodone acutely reduced neuronal excitability, tolerance developed to this effect, and reversal of that tolerance occurred at the level of a single neuron, suggesting that reversal of oxycodone tolerance by either ethanol or Bis XI involves cellular mechanisms.


Subject(s)
Drug Tolerance , Ethanol/pharmacology , Ganglia, Spinal/drug effects , Neurons/drug effects , Oxycodone/pharmacology , Action Potentials/drug effects , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacology , Animals , Cells, Cultured , Ganglia, Spinal/cytology , Indoles/pharmacology , Male , Maleimides/pharmacology , Membrane Potentials/drug effects , Mice, Inbred C57BL , Mice, Knockout , Neurons/physiology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , beta-Arrestin 2/genetics
7.
FASEB J ; 31(6): 2649-2660, 2017 06.
Article in English | MEDLINE | ID: mdl-28280004

ABSTRACT

Morphine is one of the most widely used drugs for the treatment of pain. However, side effects, including persistent constipation and antinociceptive tolerance, limit its clinical efficacy. Prolonged morphine treatment results in a "leaky" gut, predisposing to colonic inflammation that is facilitated by microbial dysbiosis and associated bacterial translocation. In this study, we examined the role of enteric glia in mediating this secondary inflammatory response to prolonged treatment with morphine. We found that purinergic P2X receptor activity was significantly enhanced in enteric glia that were isolated from mice with long-term morphine treatment (in vivo) but not upon direct exposure of glia to morphine (in vitro). LPS, a major bacterial product, also increased ATP-induced currents, as well as expression of P2X4, P2X7, IL6, IL-1ß mRNA in enteric glia. LPS increased connexin43 (Cx43) expression and enhanced ATP release from enteric glia cells. LPS-induced P2X currents and proinflammatory cytokine mRNA expression were blocked by the Cx43 blockers Gap26 and carbenoxolone. Likewise, colonic inflammation related to prolonged exposure to morphine was significantly attenuated by carbenoxolone (25 mg/kg). Carbenoxolone also prevented gut wall disruption and significantly reduced morphine-induced constipation. These findings imply that enteric glia activation is a significant modulator of morphine-related inflammation and constipation.-Bhave, S., Gade, A., Kang, M., Hauser, K. F., Dewey, W. L., Akbarali, H. I. Connexin-purinergic signaling in enteric glia mediates the prolonged effect of morphine on constipation.


Subject(s)
Connexin 43/metabolism , Constipation/chemically induced , Morphine/pharmacology , Neuroglia/physiology , Receptors, Purinergic P2X/metabolism , Signal Transduction/drug effects , Adenosine Triphosphate , Analgesics, Opioid/pharmacology , Animals , Electrophysiological Phenomena , Gene Expression Regulation , Intestines/drug effects , Intestines/physiology , Lipopolysaccharides/toxicity , Male , Membrane Potentials , Mice , RNA, Messenger , Receptors, Purinergic P2X/genetics
8.
Brain Behav Immun ; 69: 124-138, 2018 03.
Article in English | MEDLINE | ID: mdl-29146238

ABSTRACT

The HIV-1 regulatory protein, trans-activator of transcription (Tat), interacts with opioids to potentiate neuroinflammation and neurodegeneration within the CNS. These effects may involve the C-C chemokine receptor type 5 (CCR5); however, the behavioral contribution of CCR5 on Tat/opioid interactions is not known. Using a transgenic murine model that expresses HIV-1 Tat protein in a GFAP-regulated, doxycycline-inducible manner, we assessed morphine tolerance, dependence, and reward. To assess the influence of CCR5 on these effects, mice were pretreated with oral vehicle or the CCR5 antagonist, maraviroc, prior to morphine administration. We found that HIV-1 Tat expression significantly attenuated the antinociceptive potency of acute morphine (2-64 mg/kg, i.p.) in non-tolerant mice. Consistent with this, Tat attenuated withdrawal symptoms among morphine-tolerant mice. Pretreatment with maraviroc blocked the effects of Tat, reinstating morphine potency in non-tolerant mice and restoring withdrawal symptomology in morphine-tolerant mice. Twenty-four hours following morphine administration, HIV-1 Tat significantly potentiated (∼3.5-fold) morphine-conditioned place preference and maraviroc further potentiated these effects (∼5.7-fold). Maraviroc exerted no measurable behavioral effects on its own. Protein array analyses revealed only minor changes to cytokine profiles when morphine was administered acutely or repeatedly; however, 24 h post morphine administration, the expression of several cytokines was greatly increased, including endogenous CCR5 chemokine ligands (CCL3, CCL4, and CCL5), as well as CCL2. Tat further elevated levels of several cytokines and maraviroc pretreatment attenuated these effects. These data demonstrate that CCR5 mediates key aspects of HIV-1 Tat-induced alterations in the antinociceptive potency and rewarding properties of opioids.


Subject(s)
Analgesics, Opioid/pharmacology , Drug Tolerance/physiology , Inflammation/metabolism , Morphine/pharmacology , Receptors, CCR5/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Animals , CCR5 Receptor Antagonists/pharmacology , Caudate Nucleus/metabolism , Conditioning, Operant/drug effects , Cytokines/metabolism , Inflammation/chemically induced , Inflammation/genetics , Male , Maraviroc/pharmacology , Mice , Mice, Transgenic , Motor Activity/drug effects , Reward , tat Gene Products, Human Immunodeficiency Virus/genetics
9.
J Cell Mol Med ; 21(2): 364-374, 2017 02.
Article in English | MEDLINE | ID: mdl-27629819

ABSTRACT

Statins, 3-hydroxyl-3-methylglutaryl coenzyme A reductase inhibitors, are the first-line medications prescribed for the prevention and treatment of coronary artery diseases. The efficacy of statins has been attributed not only to their systemic cholesterol-lowering actions but also to their pleiotropic effects that are unrelated to cholesterol reduction. These pleiotropic effects have been increasingly recognized as essential in statins therapy. This study was designed to investigate the pleiotropic actions of simvastatin, one of the most commonly prescribed statins, on macrophage cholesterol homeostasis with a focus on lysosomal free cholesterol egression. With simultaneous nile red and filipin staining, analysis of confocal/multi-photon imaging demonstrated that simvastatin markedly attenuated unesterified (free) cholesterol buildup in macrophages loaded with oxidized low-density lipoprotein but had little effect in reducing the sizes of cholesteryl ester-containing lipid droplets; the reduction in free cholesterol was mainly attributed to decreases in lysosome-compartmentalized cholesterol. Functionally, the egression of free cholesterol from lysosomes attenuated pro-inflammatory cytokine secretion. It was determined that the reduction of lysosomal free cholesterol buildup by simvastatin was due to the up-regulation of Niemann-Pick C1 (NPC1), a lysosomal residing cholesterol transporter. Moreover, the enhanced enzymatic production of 7-hydroxycholesterol by cytochrome P450 7A1 and the subsequent activation of liver X receptor α underscored the up-regulation of NPC1. These findings reveal a novel pleiotropic effect of simvastatin in affecting lysosomal cholesterol efflux in macrophages and the associated significance in the treatment of atherosclerosis.


Subject(s)
Cholesterol 7-alpha-Hydroxylase/metabolism , Cholesterol/metabolism , Lipoproteins, LDL/pharmacology , Liver X Receptors/metabolism , Lysosomes/metabolism , Macrophages/metabolism , Proteins/metabolism , Simvastatin/pharmacology , Animals , Biological Transport/drug effects , Cytokines/metabolism , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins , Lysosomes/drug effects , Macrophages/drug effects , Mice, Inbred C57BL , Niemann-Pick C1 Protein , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects
10.
J Pharmacol Exp Ther ; 362(1): 45-52, 2017 07.
Article in English | MEDLINE | ID: mdl-28442580

ABSTRACT

This study compared the development of tolerance to two orally bioavailable prescription opioids, oxycodone and hydrocodone, to that of morphine, and the reversal of this tolerance by ethanol. Oxycodone (s.c.) was significantly more potent in the mouse tail-withdrawal assay than either morphine or hydrocodone. Oxycodone was also significantly more potent in this assay than hydrocodone when administered orally. Tolerance was seen following chronic subcutaneous administration of each of the three drugs and by the chronic administration of oral oxycodone, but not following the chronic oral administration of hydrocodone. Ethanol (1 g/kg i.p.) significantly reversed the tolerance to the subcutaneous administration of each of the three opioids that developed when given 30 minutes prior to challenge doses. It took twice as much ethanol, when given orally, to reverse the tolerance to oxycodone. We investigated whether the observed tolerance to oxycodone and its reversal by ethanol were due to biodispositional changes or reflected a true neuronal tolerance. As expected, a relationship between brain oxycodone concentrations and activity in the tail-immersion test existed following administration of acute oral oxycodone. Following chronic treatment, brain oxycodone concentrations were significantly lower than acute concentrations. Oral ethanol (2 g/kg) reversed the tolerance to chronic oxycodone, but did not alter brain concentrations of either acute or chronic oxycodone. These studies show that there is a metabolic component of tolerance to oxycodone; however, the reversal of that tolerance by ethanol is not due to an alteration of the biodisposition of oxycodone, but rather is neuronal in nature.


Subject(s)
Analgesics, Opioid/pharmacology , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Hydrocodone/pharmacology , Oxycodone/pharmacology , Analgesics, Opioid/pharmacokinetics , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Drug Tolerance , Hydrocodone/pharmacokinetics , Male , Mice , Oxycodone/pharmacokinetics , Pain/psychology , Pain Measurement/drug effects
11.
J Pharmacol Exp Ther ; 361(1): 51-59, 2017 04.
Article in English | MEDLINE | ID: mdl-28130265

ABSTRACT

Respiratory depression is the major cause of death in opioid overdose. We have previously shown that prolonged treatment of mice with morphine induces profound tolerance to the respiratory-depressant effects of the drug (Hill et al., 2016). The aim of the present study was to investigate whether tolerance to opioid-induced respiratory depression is mediated by protein kinase C (PKC) and/or c-Jun N-terminal kinase (JNK). We found that although mice treated for up to 6 days with morphine developed tolerance, as measured by the reduced responsiveness to an acute challenge dose of morphine, administration of the brain-penetrant PKC inhibitors tamoxifen and calphostin C restored the ability of acute morphine to produce respiratory depression in morphine-treated mice. Importantly, reversal of opioid tolerance was dependent on the nature of the opioid ligand used to induce tolerance, as these PKC inhibitors did not reverse tolerance induced by prolonged treatment of mice with methadone nor did they reverse the protection to acute morphine-induced respiratory depression afforded by prolonged treatment with buprenorphine. We found no evidence for the involvement of JNK in morphine-induced tolerance to respiratory depression. These results indicate that PKC represents a major mechanism underlying morphine tolerance, that the mechanism of opioid tolerance to respiratory depression is ligand-dependent, and that coadministration of drugs with PKC-inhibitory activity and morphine (as well as heroin, largely metabolized to morphine in the body) may render individuals more susceptible to overdose death by reversing tolerance to the effects of morphine.


Subject(s)
Analgesics, Opioid/toxicity , JNK Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Respiratory Insufficiency/enzymology , Tamoxifen/therapeutic use , Analgesics, Opioid/administration & dosage , Animals , Brain/drug effects , Brain/metabolism , Drug Tolerance/physiology , Infusion Pumps, Implantable , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Male , Mice , Protein Kinase C/antagonists & inhibitors , Random Allocation , Respiratory Insufficiency/chemically induced , Tamoxifen/pharmacology , Treatment Outcome
12.
J Cell Mol Med ; 20(6): 1001-13, 2016 06.
Article in English | MEDLINE | ID: mdl-26818887

ABSTRACT

The disruption in transportation of oxLDL-derived cholesterol and the subsequent lipid accumulation in macrophages are the hallmark events in atherogenesis. Our recent studies demonstrated that lysosomal Ca(2+) messenger of nicotinic acid adenine dinucleotide phosphate (NAADP), an enzymatic product of CD38 ADP-ribosylcyclase (CD38), promoted lipid endocytic trafficking in human fibroblast cells. The current studies are designed to examine the functional role of CD38/NAADP pathway in the regulation of lysosomal cholesterol efflux in atherosclerosis. Oil red O staining showed that oxLDL concentration-dependently increased lipid buildup in bone marrow-derived macrophages from both wild type and CD38(-/-) , but to a significant higher extent with CD38 gene deletion. Bodipy 493/503 fluorescence staining found that the deposited lipid in macrophages was mainly enclosed in lysosomal organelles and largely enhanced with the blockade of CD38/NAADP pathway. Filipin staining and direct measurement of lysosome fraction further revealed that the free cholesterol constituted a major portion of the total cholesterol segregated in lysosomes. Moreover, in situ assay disclosed that both lysosomal lumen acidity and the acid lipase activity were reduced upon cholesterol buildup in lysosomes. In CD38(-/-) mice, treatment with Western diet (12 weeks) produced atherosclerotic damage in coronary artery with striking lysosomal cholesterol sequestration in macrophages. These data provide the first experimental evidence that the proper function of CD38/NAADP pathway plays an essential role in promoting free cholesterol efflux from lysosomes and that a defection of this signalling leads to lysosomal cholesterol accumulation in macrophages and results in coronary atherosclerosis in CD38(-/-) mice.


Subject(s)
ADP-ribosyl Cyclase 1/deficiency , ADP-ribosyl Cyclase 1/metabolism , Atherosclerosis/metabolism , Cholesterol/metabolism , Lysosomes/metabolism , Macrophages/metabolism , ADP-ribosyl Cyclase 1/genetics , Acids/metabolism , Animals , Atherosclerosis/pathology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Aggregation/drug effects , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Coronary Vessels/pathology , Genotype , Lipid Metabolism/drug effects , Lipoproteins, LDL/pharmacology , Lysosomes/drug effects , Lysosomes/ultrastructure , Macrophages/drug effects , Macrophages/ultrastructure , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , NADP/analogs & derivatives , NADP/metabolism , Reproducibility of Results , Signal Transduction/drug effects , Sterol Esterase/metabolism
13.
J Pharmacol Exp Ther ; 357(3): 520-8, 2016 06.
Article in English | MEDLINE | ID: mdl-27068812

ABSTRACT

Opioid-induced constipation is a major side effect that persists with long-term opioid use. Previous studies demonstrated that nicotine-induced contractions are enhanced after long-term morphine exposure in guinea pig ileum. In the present study, we examined whether the increased sensitivity to nicotine could be observed in single enteric neurons after long-term morphine exposure, determined the subunits in mouse enteric neurons, and examined the effect of nicotine in reversing opioid-induced constipation. Nicotine (0.03-1 mM) dose-dependently induced inward currents from a holding potential of -60 mV in isolated single enteric neurons from the mouse ileum. The amplitude of the currents, but not the potency to nicotine, was significantly increased in neurons receiving long-term (16-24 h) but not short-term (10 min) exposure to morphine. Quantitative mRNA analysis showed that nicotinic acetylcholine receptor (nAChR) subunit expression in the mouse ileum was α3 ≥ ß2 > ß4 > α5 > α4 > ß3 > α6. Nicotine-induced currents were obtained in neurons from α7, ß2, α5, and α6 knockout mice. The currents were, however, inhibited by mecamylamine (10 µM) and the α3ß4 blocker α-conotoxin AuIB (3 µM), suggesting that nicotine-induced currents were mediated by the α3ß4 subtype of nAChRs on enteric neurons. Conversely, NS3861, a partial agonist at α3ß4 nAChR, enhanced fecal pellet expulsion in a dose-dependent manner in mice that received long-term, but not short-term, morphine treatment. Overall, our findings suggest that the efficacy of nAChR agonists on enteric neurons is enhanced after long-term morphine exposure, and activation of the α3ß4 subtype of nAChR reverses chronic, but not acute, morphine-induced constipation.


Subject(s)
Constipation/chemically induced , Enteric Nervous System/cytology , Morphine/adverse effects , Neurons/cytology , Neurons/drug effects , Receptors, Nicotinic/metabolism , Adenosine Triphosphate/pharmacology , Animals , Constipation/metabolism , Constipation/pathology , Constipation/physiopathology , Drug Partial Agonism , Drug Synergism , Electrophysiological Phenomena/drug effects , Enteric Nervous System/pathology , Gastrointestinal Motility/drug effects , Gene Expression Regulation/drug effects , Intestine, Small/innervation , Male , Mice , Neurons/metabolism , Neurons/pathology , Nicotine/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Nicotinic/genetics , Time Factors
14.
J Pharmacol Exp Ther ; 356(1): 96-105, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26542403

ABSTRACT

Despite considerable evidence that chronic opiate use selectively affects the pathophysiologic consequences of human immunodeficiency virus type 1 (HIV-1) infection in the nervous system, few studies have examined whether neuro-acquired immune deficiency syndrome (neuroAIDS) might intrinsically alter the pharmacologic responses to chronic opiate exposure. This is an important matter because HIV-1 and opiate abuse are interrelated epidemics, and HIV-1 patients are often prescribed opiates as a treatment of HIV-1-related neuropathic pain. Tolerance and physical dependence are inevitable consequences of frequent and repeated administration of morphine. In the present study, mice expressing HIV-1 Tat in a doxycycline (DOX)-inducible manner [Tat(+)], their Tat(-) controls, and control C57BL/6 mice were chronically exposed to placebo or 75-mg morphine pellets to explore the effects of Tat induction on morphine tolerance and dependence. Antinociceptive tolerance and locomotor activity tolerance were assessed using tail-flick and locomotor activity assays, respectively, and physical dependence was measured with the platform-jumping assay and recording of other withdrawal signs. We found that Tat(+) mice treated with DOX [Tat(+)/DOX] developed an increased tolerance in the tail-flick assay compared with control Tat(-)/DOX and/or C57/DOX mice. Equivalent tolerance was developed in all mice when assessed by locomotor activity. Further, Tat(+)/DOX mice expressed reduced levels of physical dependence to chronic morphine exposure after a 1-mg/kg naloxone challenge compared with control Tat(-)/DOX and/or C57/DOX mice. Assuming the results seen in Tat transgenic mice can be generalized to neuroAIDS, our findings suggest that HIV-1-infected individuals may display heightened analgesic tolerance to similar doses of opiates compared with uninfected individuals and show fewer symptoms of physical dependence.


Subject(s)
Drug Tolerance/genetics , Morphine Dependence/genetics , Morphine Dependence/psychology , Morphine/pharmacology , Narcotics/pharmacology , tat Gene Products, Human Immunodeficiency Virus/genetics , Animals , Body Weight/drug effects , Body Weight/genetics , Doxycycline/pharmacology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Neuralgia/etiology , Pain Measurement/drug effects , Reaction Time/drug effects , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/psychology
15.
J Pharmacol Exp Ther ; 357(1): 145-56, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26791602

ABSTRACT

Serious clinical liabilities associated with the prescription of opiates for pain control include constipation, respiratory depression, pruritus, tolerance, abuse, and addiction. A recognized strategy to circumvent these side effects is to combine opioids with other antinociceptive agents. The combination of opiates with the primary active constituent of cannabis (Δ(9)-tetrahydrocannabinol) produces enhanced antinociceptive actions, suggesting that cannabinoid receptor agonists can be opioid sparing. Here, we tested whether elevating the endogenous cannabinoid 2-arachidonoylglycerol through the inhibition of its primary hydrolytic enzyme monoacylglycerol lipase (MAGL), will produce opioid-sparing effects in the mouse chronic constriction injury (CCI) of the sciatic nerve model of neuropathic pain. The dose-response relationships of i.p. administration of morphine and the selective MAGL inhibitor 2,5-dioxopyrrolidin-1-yl 4-(bis(4-chlorophenyl)methyl)piperazine-1-carboxylate (MJN110) were tested alone and in combination at equieffective doses for reversal of CCI-induced mechanical allodynia and thermal hyperalgesia. The respective ED50 doses (95% confidence interval) of morphine and MJN110 were 2.4 (1.9-3.0) mg/kg and 0.43 (0.23-0.79) mg/kg. Isobolographic analysis of these drugs in combination revealed synergistic antiallodynic effects. Acute antinociceptive effects of the combination of morphine and MJN110 required µ-opioid, CB1, and CB2 receptors. This combination did not reduce gastric motility or produce subjective cannabimimetic effects in the drug discrimination assay. Importantly, combinations of MJN110 and morphine given repeatedly (i.e., twice a day for 6 days) continued to produce antiallodynic effects with no evidence of tolerance. Taken together, these findings suggest that MAGL inhibition produces opiate-sparing events with diminished tolerance, constipation, and cannabimimetic side effects.


Subject(s)
Analgesics, Opioid/therapeutic use , Carbamates/pharmacology , Enzyme Inhibitors/pharmacology , Monoacylglycerol Lipases/antagonists & inhibitors , Neuralgia/drug therapy , Succinimides/pharmacology , Animals , Arachidonic Acids/metabolism , Behavior, Animal/drug effects , Constriction, Pathologic/complications , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Endocannabinoids/metabolism , Glycerides/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Male , Mice , Mice, Inbred C57BL , Morphine/administration & dosage , Morphine/therapeutic use , Neuralgia/chemically induced , Neuralgia/psychology , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB2/drug effects , Receptors, Opioid, mu/drug effects
16.
J Neurosci ; 34(43): 14243-51, 2014 Oct 22.
Article in English | MEDLINE | ID: mdl-25339738

ABSTRACT

The gastrointestinal (GI) tract presents a major site of immune modulation by HIV, resulting in significant morbidity. Most GI processes affected during HIV infection are regulated by the enteric nervous system. HIV has been identified in GI histologic specimens in up to 40% of patients, and the presence of viral proteins, including the trans-activator of transcription (Tat), has been reported in the gut indicating that HIV itself may be an indirect gut pathogen. Little is known of how Tat affects the enteric nervous system. Here we investigated the effects of the Tat protein on enteric neuronal excitability, proinflammatory cytokine release, and its overall effect on GI motility. Direct application of Tat (100 nm) increased the number of action potentials and reduced the threshold for action potential initiation in isolated myenteric neurons. This effect persisted in neurons pretreated with Tat for 3 d (19 of 20) and in neurons isolated from Tat(+) (Tat-expressing) transgenic mice. Tat increased sodium channel isoforms Nav1.7 and Nav1.8 levels. This increase was accompanied by an increase in sodium current density and a leftward shift in the sodium channel activation voltage. RANTES, IL-6, and IL-1ß, but not TNF-α, were enhanced by Tat. Intestinal transit and cecal water content were also significantly higher in Tat(+) transgenic mice than Tat(-) littermates (controls). Together, these findings show that Tat has a direct and persistent effect on enteric neuronal excitability, and together with its effect on proinflammatory cytokines, regulates gut motility, thereby contributing to GI dysmotilities reported in HIV patients.


Subject(s)
Enteric Nervous System/pathology , Gastrointestinal Motility/physiology , HIV-1 , Ileum/pathology , tat Gene Products, Human Immunodeficiency Virus/toxicity , Action Potentials/drug effects , Action Potentials/physiology , Animals , Enteric Nervous System/drug effects , Enteric Nervous System/metabolism , Female , Gastrointestinal Motility/drug effects , Humans , Ileum/drug effects , Ileum/metabolism , Inflammation Mediators/metabolism , Male , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Organ Culture Techniques , Rats, Sprague-Dawley
17.
Bioorg Med Chem ; 23(8): 1701-15, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25783191

ABSTRACT

A series of 17-cyclopropylmethyl-3,14ß-dihydroxy-4,5α-epoxy-6α-(isoquinoline-3'-carboxamido)morphinan (NAQ) analogues were synthesized and pharmacologically characterized to study their structure-activity relationship at the mu opioid receptor (MOR). The competition binding assay showed two-atom spacer and aromatic side chain were optimal for MOR selectivity. Meanwhile, substitutions at the 1'- and/or 4'-position of the isoquinoline ring retained or improved MOR selectivity over the kappa opioid receptor while still possessing above 20-fold MOR selectivity over the delta opioid receptor. In contrast, substitutions at the 6'- and/or 7'-position of the isoquinoline ring reduced MOR selectivity as well as MOR efficacy. Among this series of ligands, compound 11 acted as an antagonist when challenged with morphine in warm-water tail immersion assay and produced less significant withdrawal symptoms compared to naltrexone in morphine-pelleted mice. Compound 11 also antagonized the intracellular Ca(2+) increase induced by DAMGO. Molecular dynamics simulation studies of 11 in three opioid receptors indicated orientation of the 6'-nitro group varied significantly in the different 'address' domains of the receptors and played a crucial role in the observed binding affinities and selectivity. Collectively, the current findings provide valuable insights for future development of NAQ-based MOR selective ligands.


Subject(s)
Isoquinolines/chemistry , Isoquinolines/pharmacology , Morphinans/chemistry , Morphinans/pharmacology , Narcotic Antagonists/chemistry , Narcotic Antagonists/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Animals , CHO Cells , Cricetulus , Drug Design , Humans , Isoquinolines/therapeutic use , Ligands , Mice , Molecular Dynamics Simulation , Morphinans/therapeutic use , Morphine Dependence/drug therapy , Morphine Dependence/metabolism , Narcotic Antagonists/therapeutic use , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
18.
J Med Chem ; 67(11): 9552-9574, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38814086

ABSTRACT

Despite the availability of numerous pain medications, the current array of Food and Drug Administration-approved options falls short in adequately addressing pain states for numerous patients and consequently worsens the opioid crisis. Thus, it is imperative for basic research to develop novel and nonaddictive pain medications. Toward addressing this clinical goal, nalfurafine (NLF) was chosen as a lead and its structure-activity relationship (SAR) systematically studied through design, syntheses, and in vivo characterization of 24 analogues. Two analogues, 21 and 23, showed longer durations of action than NLF in a warm-water tail immersion assay, produced in vivo effects primarily mediated by KOR and DOR, penetrated the blood-brain barrier, and did not function as reinforcers. Additionally, 21 produced fewer sedative effects than NLF. Taken together, these results aid the understanding of NLF SAR and provide insights for future endeavors in developing novel nonaddictive therapeutics to treat pain.


Subject(s)
Morphinans , Spiro Compounds , Structure-Activity Relationship , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Spiro Compounds/chemical synthesis , Animals , Morphinans/pharmacology , Morphinans/chemistry , Morphinans/chemical synthesis , Morphinans/therapeutic use , Mice , Male , Humans , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/metabolism , Pain Management/methods , Pain/drug therapy , Analgesics/pharmacology , Analgesics/chemistry , Analgesics/chemical synthesis , Analgesics/therapeutic use
19.
J Med Chem ; 67(1): 603-619, 2024 01 11.
Article in English | MEDLINE | ID: mdl-38156970

ABSTRACT

While there are approved therapeutics to treat opioid overdoses, the need for treatments to reverse overdoses due to ultrapotent fentanyls remains unmet. This may be due in part to an adrenergic mechanism of fentanyls in addition to their stereotypical mu-opioid receptor (MOR) effects. Herein, we report our efforts to further understanding of the functions these distinct mechanisms impart. Employing the known MOR neutral antagonist phenylfentanil as a lead, 17 analogues were designed based on the concept of isosteric replacement. To probe mechanisms of action, these analogues were pharmacologically evaluated in vitro and in vivo, while in silico modeling studies were also conducted on phenylfentanil. While it did not indicate MOR involvement in vivo, phenylfentanil yielded respiratory minute volumes similar to those caused by fentanyl. Taken together with molecular modeling studies, these results indicated that respiratory effects of fentanyls may also correlate to inhibition of both α1A- and α1B-adrenergic receptors.


Subject(s)
Adrenergic Agents , Fentanyl , Fentanyl/pharmacology , Receptors, Opioid, mu , Narcotic Antagonists , Analgesics, Opioid/pharmacology
20.
Mol Pharmacol ; 84(2): 252-60, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23716621

ABSTRACT

Consumption of ethanol is a considerable risk factor for death in heroin overdose. We sought to determine whether a mildly intoxicating concentration of ethanol could alter morphine tolerance at the cellular level. In rat locus coeruleus (LC) neurons, tolerance to morphine was reversed by acute exposure of the brain slice to ethanol (20 mM). Tolerance to the opioid peptide [d-Ala(2),N-MePhe(4),Gly-ol]-enkephalin was not reversed by ethanol. Previous studies in LC neurons have revealed a role for protein kinase C (PKC)α in µ-opioid receptor (MOPr) desensitization by morphine and in the induction and maintenance of morphine tolerance, but we have been unable to demonstrate that 20 mM ethanol produces significant inhibition of PKCα. The ability of ethanol to reverse cellular tolerance to morphine in LC neurons was absent in the presence of the phosphatase inhibitor okadaic acid, indicating that dephosphorylation is involved. In human embryonic kidney 293 cells expressing the MOPr, ethanol reduced the level of MOPr phosphorylation induced by morphine. Ethanol reversal of tolerance did not appear to result from a direct effect on MOPr since acute exposure to ethanol (20 mM) did not modify the affinity of binding of morphine to the MOPr or the efficacy of morphine for G-protein activation as measured by guanosine 5'-O-(3-[(35)S]thio)triphosphate binding. Similarly, ethanol did not affect MOPr trafficking. We conclude that acute exposure to ethanol enhances the effects of morphine by reversing the processes underlying morphine cellular tolerance.


Subject(s)
Ethanol/pharmacology , Locus Coeruleus/drug effects , Morphine/pharmacology , Neurons/drug effects , Animals , Brain/drug effects , Brain/metabolism , Cell Line , Drug Interactions , Drug Tolerance , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Locus Coeruleus/metabolism , Male , Neurons/metabolism , Opioid Peptides/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation/drug effects , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/metabolism , Rats , Rats, Wistar , Receptors, Opioid, mu/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL