Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.270
Filter
Add more filters

Publication year range
1.
Mol Psychiatry ; 29(3): 624-632, 2024 03.
Article in English | MEDLINE | ID: mdl-38145984

ABSTRACT

(R,S)-methadone ((R,S)-MTD) is a µ-opioid receptor (MOR) agonist comprised of (R)-MTD and (S)-MTD enantiomers. (S)-MTD is being developed as an antidepressant and is considered an N-methyl-D-aspartate receptor (NMDAR) antagonist. We compared the pharmacology of (R)-MTD and (S)-MTD and found they bind to MORs, but not NMDARs, and induce full analgesia. Unlike (R)-MTD, (S)-MTD was a weak reinforcer that failed to affect extracellular dopamine or induce locomotor stimulation. Furthermore, (S)-MTD antagonized motor and dopamine releasing effects of (R)-MTD. (S)-MTD acted as a partial agonist at MOR, with complete loss of efficacy at the MOR-galanin Gal1 receptor (Gal1R) heteromer, a key mediator of the dopaminergic effects of opioids. In sum, we report novel and unique pharmacodynamic properties of (S)-MTD that are relevant to its potential mechanism of action and therapeutic use. One-sentence summary: (S)-MTD, like (R)-MTD, binds to and activates MORs in vitro, but (S)-MTD antagonizes the MOR-Gal1R heteromer, decreasing its abuse liability.


Subject(s)
Analgesics, Opioid , Methadone , Receptors, Opioid, mu , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/drug effects , Animals , Methadone/pharmacology , Male , Analgesics, Opioid/pharmacology , Humans , Mice , Dopamine/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Ligands , Stereoisomerism
2.
Am J Gastroenterol ; 119(10): 1970-1978, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38870087

ABSTRACT

Opioid receptors are found throughout the gastrointestinal tract, including the large intestine. Many patients treated with opioids experience opioid-induced constipation (OIC). Laxatives are not effective in most patients, and in those who do initially respond, the efficacy of laxatives generally diminishes over time. In addition, OIC does not spontaneously resolve for most patients. However, complications of opioids extend far beyond simply slowing gastrointestinal transit. Opioid use can affect intestinal permeability through a variety of mechanisms. Toll-like receptors are a crucial component of innate immunity and are tightly regulated within the gut epithelium. Pathologic µ-opioid receptor (MOR) and toll-like receptor signaling, resulting from chronic opioid exposure, disrupts intestinal permeability leading to potentially harmful bacterial translocation, elevated levels of bacterial toxins, immune activation, and increased cytokine production. Peripherally active MOR antagonists, including methylnaltrexone, are effective at treating OIC. Benefits extend beyond simply blocking the MOR; these agents also act to ameliorate opioid-induced disrupted intestinal permeability. In this review, we briefly describe the physiology of the gastrointestinal epithelial border and discuss the impact of opioids on gastrointestinal function. Finally, we consider the use of peripherally active MOR antagonists to treat disrupted intestinal permeability resulting from opioid use and discuss the potential for improved morbidity and mortality in patients treated with methylnaltrexone for opioid-induced bowel disorders.


Subject(s)
Analgesics, Opioid , Intestinal Mucosa , Narcotic Antagonists , Permeability , Receptors, Opioid, mu , Humans , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/pharmacology , Permeability/drug effects , Narcotic Antagonists/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Opioid-Induced Constipation/drug therapy , Naltrexone/pharmacology , Naltrexone/analogs & derivatives , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Quaternary Ammonium Compounds/pharmacology , Quaternary Ammonium Compounds/therapeutic use , Intestinal Barrier Function
3.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Article in English | MEDLINE | ID: mdl-33846240

ABSTRACT

Positive allosteric modulators (PAMs) of the mu-opioid receptor (MOR) have been hypothesized as potentially safer analgesics than traditional opioid drugs. This is based on the idea that PAMs will promote the action of endogenous opioid peptides while preserving their temporal and spatial release patterns and so have an improved therapeutic index. However, this hypothesis has never been tested. Here, we show that a mu-PAM, BMS-986122, enhances the ability of the endogenous opioid Methionine-enkephalin (Met-Enk) to stimulate G protein activity in mouse brain homogenates without activity on its own and to enhance G protein activation to a greater extent than ß-arrestin recruitment in Chinese hamster ovary (CHO) cells expressing human mu-opioid receptors. Moreover, BMS-986122 increases the potency of Met-Enk to inhibit GABA release in the periaqueductal gray, an important site for antinociception. We describe in vivo experiments demonstrating that the mu-PAM produces antinociception in mouse models of acute noxious heat pain as well as inflammatory pain. These effects are blocked by MOR antagonists and are consistent with the hypothesis that in vivo mu-PAMs enhance the activity of endogenous opioid peptides. Because BMS-986122 does not bind to the orthosteric site and has no inherent agonist action at endogenously expressed levels of MOR, it produces a reduced level of morphine-like side effects of constipation, reward as measured by conditioned place preference, and respiratory depression. These data provide a rationale for the further exploration of the action and safety of mu-PAMs as an innovative approach to pain management.


Subject(s)
Allosteric Regulation/physiology , Pain/drug therapy , Receptors, Opioid, mu/metabolism , Allosteric Regulation/drug effects , Analgesia/methods , Analgesics , Analgesics, Opioid/pharmacology , Animals , CHO Cells , Cricetulus , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Morphine , Narcotic Antagonists , Pain Management/methods , Proof of Concept Study , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/drug effects
4.
J Neurochem ; 160(4): 454-468, 2022 02.
Article in English | MEDLINE | ID: mdl-34919270

ABSTRACT

There is increasing evidence for a daily rhythm of µ-opioid receptor (MOR) efficacy and the development of alcohol dependence. Previous studies show that ß-arrestin 2 (bArr2) has an impact on alcohol intake, at least partially mediated via modulation of MOR signaling, which in turn mediates the alcohol rewarding effects. Considering the interplay of circadian rhythms on MOR and alcohol dependence, we aimed to investigate bArr2 in alcohol dependence at different time points of the day/light cycle on the level of bArr2 mRNA (in situ hybridization), MOR availability (receptor autoradiography), and MOR signaling (Damgo-stimulated G-protein coupling) in the nucleus accumbens of alcohol-dependent and non-dependent Wistar rats. Using a microarray data set we found that bArr2, but not bArr1, shows a diurnal transcription pattern in the accumbens of naïve rats with higher expression levels during the active cycle. In 3-week abstinent rats, bArr2 is up-regulated in the accumbens at the beginning of the active cycle (ZT15), whereas no differences were found at the beginning of the inactive cycle (ZT3) compared with controls. This effect was accompanied by a specific down-regulation of MOR binding in the active cycle. Additionally, we detect a higher receptor coupling during the inactive cycle compared with the active cycle in alcohol-dependent animals. Together, we report daily rhythmicity for bArr2 expression linked to an inverse pattern of MOR, suggesting an involvement for bArr2 on circadian regulation of G-protein coupled receptors in alcohol dependence. The presented data may have implications for the development of novel bArr2-related treatment targets for alcoholism.


Subject(s)
Alcoholism/genetics , Circadian Rhythm/genetics , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/genetics , beta-Arrestin 2/genetics , Alcoholism/drug therapy , Animals , Down-Regulation , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Male , Microarray Analysis , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Reward
5.
Am J Physiol Gastrointest Liver Physiol ; 322(1): G66-G78, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34755545

ABSTRACT

Allosteric modulators (AMs) are molecules that can fine-tune signaling by G protein-coupled receptors (GPCRs). Although they are a promising therapeutic approach for treating a range of disorders, allosteric modulation of GPCRs in the context of the enteric nervous system (ENS) and digestive dysfunction remains largely unexplored. This study examined allosteric modulation of the delta opioid receptor (DOR) in the ENS and assessed the suitability of DOR AMs for the treatment of irritable bowel syndrome (IBS) symptoms using mouse models. The effects of the positive allosteric modulator (PAM) of DOR, BMS-986187, on neurogenic contractions of the mouse colon and on DOR internalization in enteric neurons were quantified. The ability of BMS-986187 to influence colonic motility was assessed both in vitro and in vivo. BMS-986187 displayed DOR-selective PAM-agonist activity and orthosteric agonist probe dependence in the mouse colon. BMS-986187 augmented the inhibitory effects of DOR agonists on neurogenic contractions and enhanced reflex-evoked DOR internalization in myenteric neurons. BMS-986187 significantly increased DOR endocytosis in myenteric neurons in response to the weakly internalizing agonist ARM390. BMS-986187 reduced the generation of complex motor patterns in the isolated intact colon. BMS-986187 reduced fecal output and diarrhea onset in the novel environment stress and castor oil models of IBS symptoms, respectively. DOR PAMs enhance DOR-mediated signaling in the ENS and have potential benefit for the treatment of dysmotility. This study provides proof of concept to support the use of GPCR AMs for the treatment of gastrointestinal motility disorders.NEW & NOTEWORTHY This study assesses the use of positive allosteric modulation as a pharmacological approach to enhance opioid receptor signaling in the enteric nervous system. We demonstrate that selective modulation of endogenous delta opioid receptor signaling can suppress colonic motility without causing constipation. We propose that allosteric modulation of opioid receptor signaling may be a therapeutic strategy to normalize gastrointestinal motility in conditions such as irritable bowel syndrome.


Subject(s)
Enteric Nervous System/drug effects , Gastrointestinal Motility/drug effects , Receptors, Opioid, delta/drug effects , Xanthones/pharmacology , Analgesics, Opioid/pharmacology , Benzamides/pharmacology , Colon/drug effects , Enteric Nervous System/physiopathology , Gastrointestinal Motility/physiology , Humans , Receptors, Opioid/drug effects , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/drug effects , Signal Transduction/drug effects
6.
Int J Obes (Lond) ; 46(2): 400-407, 2022 02.
Article in English | MEDLINE | ID: mdl-34728775

ABSTRACT

BACKGROUND: Obesity is a pressing public health concern worldwide. Novel pharmacological means are urgently needed to combat the increase of obesity and accompanying type 2 diabetes (T2D). Although fully established obesity is associated with neuromolecular alterations and insulin resistance in the brain, potential obesity-promoting mechanisms in the central nervous system have remained elusive. In this triple-tracer positron emission tomography study, we investigated whether brain insulin signaling, µ-opioid receptors (MORs) and cannabinoid CB1 receptors (CB1Rs) are associated with risk for developing obesity. METHODS: Subjects were 41 young non-obese males with variable obesity risk profiles. Obesity risk was assessed by subjects' physical exercise habits, body mass index and familial risk factors, including parental obesity and T2D. Brain glucose uptake was quantified with [18F]FDG during hyperinsulinemic euglycemic clamp, MORs were quantified with [11C]carfentanil and CB1Rs with [18F]FMPEP-d2. RESULTS: Subjects with higher obesity risk had globally increased insulin-stimulated brain glucose uptake (19 high-risk subjects versus 19 low-risk subjects), and familial obesity risk factors were associated with increased brain glucose uptake (38 subjects) but decreased availability of MORs (41 subjects) and CB1Rs (36 subjects). CONCLUSIONS: These results suggest that the hereditary mechanisms promoting obesity may be partly mediated via insulin, opioid and endocannabinoid messaging systems in the brain.


Subject(s)
Cerebrum/metabolism , Glucose Intolerance/etiology , Obesity/diagnosis , Receptor, Cannabinoid, CB1/drug effects , Receptors, Opioid, mu/drug effects , Adult , Body Mass Index , Cerebrum/physiopathology , Female , Finland/epidemiology , Glucose Intolerance/epidemiology , Glucose Intolerance/metabolism , Humans , Linear Models , Male , Obesity/epidemiology , Obesity/metabolism , Positron-Emission Tomography/methods , Positron-Emission Tomography/statistics & numerical data , Receptor, Cannabinoid, CB1/metabolism , Receptors, Opioid, mu/metabolism , Risk Factors
7.
Addict Biol ; 27(1): e13086, 2022 01.
Article in English | MEDLINE | ID: mdl-34382313

ABSTRACT

Repeated morphine exposure has been shown to induce neuronal plasticity in reward-related areas of the brain. miR-132, a CREB-induced and activation-dependent microRNA, has been suggested to be involved in the neuronal plasticity by increasing neuronal dendritic branches and spinogenesis. However, it is still unclear whether miR-132 is related to morphine dependence. Here, we investigate whether miR-132 is involved in morphine dependence and whether it is related to the structural plasticity of the dentate gyrus (DG) neurons. Sprague-Dawley rats are treated with increasing doses of morphine injection for six consecutive days to develop morphine dependence. Our results show that dendritic branching and spinogenesis of the DG neurons of morphine dependent rats are increased. Morphine treatment (24 h) promotes the differentiation of N2a cells stably expressing µ-opioid receptor by up-regulating miR-132 expression. Moreover, inhibiting miR-132 3p (but not 5p) of the DG neurons can reverse the structural plasticity and disrupt the formation of morphine dependence in rats. These findings indicate that miR-132 in the DG neurons is involved in morphine dependence via modifying the neuronal plasticity.


Subject(s)
Dentate Gyrus/drug effects , MicroRNAs/metabolism , Morphine Dependence/physiopathology , Neuronal Plasticity/drug effects , Animals , Dose-Response Relationship, Drug , Male , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/drug effects
8.
J Neurosci ; 40(31): 5894-5907, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32601247

ABSTRACT

The orbitofrontal cortex (OFC) plays a critical role in evaluating outcomes in a changing environment. Administering opioids to the OFC can alter the hedonic reaction to food rewards and increase their consumption in a subregion-specific manner. However, it is unknown how mu-opioid signaling influences synaptic transmission in the OFC. Thus, we investigated the cellular actions of mu-opioids within distinct subregions of the OFC. Using in vitro patch-clamp electrophysiology in brain slices containing the OFC, we found that the mu-opioid agonist DAMGO produced a concentration-dependent inhibition of GABAergic synaptic transmission onto medial OFC (mOFC), but not lateral OFC (lOFC) neurons. This effect was mediated by presynaptic mu-opioid receptor activation of local parvalbumin (PV+)-expressing interneurons. The DAMGO-induced suppression of inhibition was long lasting and not reversed on washout of DAMGO or by application of the mu-opioid receptor antagonist CTAP, suggesting an inhibitory long-term depression (LTD) induced by an exogenous mu-opioid. We show that LTD at inhibitory synapses is dependent on downstream cAMP/protein kinase A (PKA) signaling, which differs between the mOFC and lOFC. Finally, we demonstrate that endogenous opioid release triggered via moderate physiological stimulation can induce LTD. Together, these results suggest that presynaptic mu-opioid stimulation of local PV+ interneurons induces a long-lasting suppression of GABAergic synaptic transmission, which depends on subregional differences in mu-opioid receptor coupling to the downstream cAMP/PKA intracellular cascade. These findings provide mechanistic insight into the opposing functional effects produced by mu-opioids within the OFC.SIGNIFICANCE STATEMENT Considering that both the orbitofrontal cortex (OFC) and the opioid system regulate reward, motivation, and food intake, understanding the role of opioid signaling within the OFC is fundamental for a mechanistic understanding of the sequelae for several psychiatric disorders. This study makes several novel observations. First, mu-opioids induce a long-lasting suppression of inhibitory synaptic transmission onto OFC pyramidal neurons in a regionally selective manner. Second, mu-opioids recruit parvalbumin inputs to suppress inhibitory synaptic transmission in the mOFC. Third, the regional selectivity of mu-opioid action of endogenous opioids is due to the efficacy of mu-opioid receptor coupling to the downstream cAMP/PKA intracellular cascades. These experiments are the first to reveal a cellular mechanism of opioid action within the OFC.


Subject(s)
Analgesics, Opioid/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Frontal Lobe/drug effects , Pyramidal Cells/drug effects , Receptors, Opioid, mu/drug effects , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid , Animals , Cyclic AMP-Dependent Protein Kinases , Endorphins/metabolism , In Vitro Techniques , Interneurons/drug effects , Long-Term Synaptic Depression/drug effects , Male , Mice , Mice, Inbred C57BL , Parvalbumins , Patch-Clamp Techniques , Signal Transduction/drug effects
9.
Bioorg Med Chem Lett ; 41: 127953, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33766769

ABSTRACT

In the present work, we reported the application of a nitrogen-walk approach on developing a series of novel opioid ligands containing an azaindole moiety at the C6-position of the epoxymorphinan skeleton. In vitro study results showed that introducing a nitrogen atom around the indole moiety not only retained excellent binding affinity, but also led to significant functional switch at the mu opioid receptor (MOR). Further computational investigations provided corroborative evidence and plausible explanations of the results of the in vitro studies. Overall, our current work implemented a series of novel MOR ligands with high binding affinity and considerably low efficacy, which may shed light on rational design of low efficacy MOR ligands for opioid use disorder therapeutics.


Subject(s)
Naltrexone/analogs & derivatives , Nitrogen/chemistry , Receptors, Opioid, mu/drug effects , Binding Sites , Humans , Ligands , Models, Molecular , Molecular Docking Simulation , Naltrexone/chemical synthesis , Naltrexone/pharmacology , Opioid-Related Disorders/drug therapy , Protein Conformation
10.
J Nat Prod ; 84(4): 1034-1043, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33635670

ABSTRACT

Ten indole and oxindole alkaloids (1-10) were isolated from the freshly collected leaves of Malaysian Mitragyna speciosa (Kratom). The chemical structures of these compounds were established on the basis of extensive 1D and 2D NMR and HRMS data analysis. The spectroscopic data of mitragynine oxindole B (4) are reported herein for the first time. The spatial configuration of mitragynine oxindole B (4) was confirmed by single-crystal X-ray diffraction. Simultaneous quantification of the isolated alkaloids in the M. speciosa leaf specimens collected from different locations in the northern region of Peninsular Malaysia was also performed using UPLC-MS/MS. The oxindole alkaloids (1-4) and the indole alkaloid (10) were assessed for binding affinity at opioid receptors. Corynoxine (1) showed high binding affinity to µ-opioid receptors with a Ki value of 16.4 nM. Further, corynoxine (1) was 1.8-fold more potent than morphine in rats subjected to a nociceptive hot plate assay. These findings have important implications for evaluating the combined effects of the minor oxindole alkaloids in the overall therapeutic activity of M. speciosa.


Subject(s)
Analgesics/pharmacology , Mitragyna/chemistry , Oxindoles/pharmacology , Receptors, Opioid, mu/drug effects , Animals , Female , Humans , Indoles , Malaysia , Male , Molecular Structure , Plant Leaves/chemistry , Rats , Rats, Sprague-Dawley , Secologanin Tryptamine Alkaloids/pharmacology , Spiro Compounds
11.
Biol Pharm Bull ; 44(11): 1746-1751, 2021.
Article in English | MEDLINE | ID: mdl-34719650

ABSTRACT

Tramadol is a weak opioid that produces analgesic effect via both the µ-opioid receptor (MOR) and non-opioid targets. Constipation is the most common opioid-related side effect in patients with cancer and non-cancer pain. However, the contribution of MOR to tramadol-induced constipation is unclear. Therefore, we used naldemedine, a peripherally acting MOR antagonist, and MOR-knockout mice to investigate the involvement of peripheral MOR in tramadol-induced constipation using a small intestinal transit model. A single dose of tramadol (3-100 mg/kg, per os (p.o.)) inhibited small intestinal transit dose-dependently in rats. Naldemedine (0.01-10 mg/kg, p.o.) blocked the inhibition of small intestinal transit induced by tramadol (30 mg/kg, p.o.) in rats. The transition rate increased dose-dependently over the range of naldemedine 0.01-0.3 mg/kg, and complete recovery was observed at 0.3-10 m/kg. Additionally, tramadol (30 and 100 mg/kg, subcutaneously (s.c.)) inhibited small intestinal transit in wild-type mice but not in MOR-knockout mice. These results suggest that peripheral MOR participates in tramadol-induced constipation.


Subject(s)
Analgesics, Opioid/adverse effects , Opioid-Induced Constipation/etiology , Receptors, Opioid, mu/drug effects , Tramadol/adverse effects , Analgesics, Opioid/blood , Analgesics, Opioid/pharmacokinetics , Animals , Intestine, Small/drug effects , Male , Naltrexone/adverse effects , Naltrexone/analogs & derivatives , Naltrexone/blood , Naltrexone/pharmacokinetics , Nociception/drug effects , Opioid-Induced Constipation/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Opioid, mu/metabolism , Tramadol/blood , Tramadol/pharmacokinetics
12.
J Clin Pharm Ther ; 46(6): 1501-1504, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34111307

ABSTRACT

WHAT IS KNOWN AND OBJECTIVE: Food and Drug Administration (FDA) risk evaluation and mitigation strategies (REMs) encourage emergency responders, paramedics, law enforcement agents, and even laypeople to be trained in the administration of naloxone with the intent of rescuing individuals from a known or suspected opioid overdose. COMMENT: Although naloxone is generally safe and effective at reversing respiratory depression caused by a conventional opioid such as morphine or heroin by competing with the opioid and displacing it from the µ-opioid receptor, questions increasingly are arising as to whether naloxone can adequately reverse opioid overdoses that may involve the potent opioids fentanyl and its analogues (F/FAs). In other words, as more and more opioid overdoses involve F/FAs, can naloxone keep up? WHAT IS NEW AND CONCLUSION: As a competitive antagonist at µ-opioid receptors, naloxone is often a life-saving agent in cases of overdose caused by conventional opioids, but it may not be versatile or powerful enough to combat the rising tide of overdoses due to fentanyl and its illicit analogues, or in cases of overdose involving combinations of opioids and non-opioids.


Subject(s)
Fentanyl/toxicity , Naloxone/therapeutic use , Narcotic Antagonists/therapeutic use , Opiate Overdose/drug therapy , Diaphragm/drug effects , Dose-Response Relationship, Drug , Fentanyl/pharmacology , Heroin/toxicity , Humans , Laryngismus/chemically induced , Muscle Rigidity/chemically induced , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid, mu/drug effects , Thoracic Wall/drug effects
13.
Neural Plast ; 2021: 6682275, 2021.
Article in English | MEDLINE | ID: mdl-33688340

ABSTRACT

Chronic pain is considered an economic burden on society as it often results in disability, job loss, and early retirement. Opioids are the most common analgesics prescribed for the management of moderate to severe pain. However, chronic exposure to these drugs can result in opioid tolerance and opioid-induced hyperalgesia. On pain modulation strategies, exploiting the multitarget drugs with the ability of the superadditive or synergistic interactions attracts more attention. In the present report, we have reviewed the analgesic effects of different dopamine receptors, particularly D1 and D2 receptors, in different regions of the central nervous system, including the spinal cord, striatum, nucleus accumbens (NAc), and periaqueductal gray (PAG). According to the evidence, these regions are not only involved in pain modulation but also express a high density of DA receptors. The findings can be categorized as follows: (1) D2-like receptors may exert a higher analgesic potency, but D1-like receptors act in different manners across several mechanisms in the mentioned regions; (2) in the spinal cord and striatum, antinociception of DA is mainly mediated by D2-like receptors, while in the NAc and PAG, both D1- and D2-like receptors are involved as analgesic targets; and (3) D2-like receptor agonists can act as adjuvants of µ-opioid receptor agonists to potentiate analgesic effects and provide a better approach to pain relief.


Subject(s)
Pain/drug therapy , Pain/physiopathology , Periaqueductal Gray/physiopathology , Receptors, Dopamine D2/agonists , Analgesics/pharmacology , Animals , Drug Tolerance/physiology , Humans , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiopathology , Pain Measurement/methods , Periaqueductal Gray/metabolism , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/metabolism
14.
Molecules ; 26(20)2021 Oct 13.
Article in English | MEDLINE | ID: mdl-34684749

ABSTRACT

The current protocols for neuropathic pain management include µ-opioid receptor (MOR) analgesics alongside other drugs; however, there is debate on the effectiveness of opioids. Nevertheless, dose escalation is required to maintain their analgesia, which, in turn, contributes to a further increase in opioid side effects. Finding novel approaches to effectively control chronic pain, particularly neuropathic pain, is a great challenge clinically. Literature data related to pain transmission reveal that angiotensin and its receptors (the AT1R, AT2R, and MAS receptors) could affect the nociception both in the periphery and CNS. The MOR and angiotensin receptors or drugs interacting with these receptors have been independently investigated in relation to analgesia. However, the interaction between the MOR and angiotensin receptors has not been excessively studied in chronic pain, particularly neuropathy. This review aims to shed light on existing literature information in relation to the analgesic action of AT1R and AT2R or MASR ligands in neuropathic pain conditions. Finally, based on literature data, we can hypothesize that combining MOR agonists with AT1R or AT2R antagonists might improve analgesia.


Subject(s)
Chronic Pain/drug therapy , Receptors, Angiotensin/drug effects , Receptors, Opioid, mu/drug effects , Analgesics/pharmacology , Analgesics, Opioid/pharmacology , Animals , Humans , Neuralgia/drug therapy , Nociception/drug effects , Pain Management/methods , Proto-Oncogene Mas , Receptors, Angiotensin/metabolism , Receptors, Opioid/agonists , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism
15.
Molecules ; 26(14)2021 Jul 08.
Article in English | MEDLINE | ID: mdl-34299443

ABSTRACT

Although persistent pain is estimated to affect about 20% of the adult population, current treatments have poor results. Polypharmacology, which is the administration of more than one drug targeting on two or more different sites of action, represents a prominent therapeutic approach for the clinical management of persistent pain. Thus, in the drug discovery process the "one-molecule-multiple targets" strategy nowadays is highly recognized. Indeed, multitarget ligands displaying a better antinociceptive activity with fewer side effects, combined with favorable pharmacokinetic and pharmacodynamic characteristics, have already been shown. Multitarget ligands possessing non-opioid/opioid and opioid/opioid mechanisms of action are considered as potential drug candidates for the management of various pain conditions. In particular, dual-target MOPr (mu opioid peptide receptor)/DOPr (delta opioid peptide receptor) ligands exhibit an improved antinociceptive profile associated with a reduced tolerance-inducing capability. The benzomorphan-based compounds LP1 and LP2 belong to this class of dual-target MOPr/DOPr ligands. In the present manuscript, the structure-activity relationships and the pharmacological fingerprint of LP1 and LP2 compounds as suitable drug candidates for persistent pain relief is described.


Subject(s)
Benzomorphans/pharmacology , Pain/drug therapy , Receptors, Opioid, delta/drug effects , Receptors, Opioid, mu/drug effects , Analgesics/pharmacology , Analgesics, Opioid/pharmacology , Animals , Benzomorphans/chemistry , Drug Delivery Systems , Drug Discovery , Humans , Ligands , Pain/physiopathology , Pain Management/methods , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
16.
Anesthesiology ; 133(3): 628-644, 2020 09.
Article in English | MEDLINE | ID: mdl-32568844

ABSTRACT

BACKGROUND: Increased descending pain facilitation accounts for opioid-induced hyperalgesia, but the underlying mechanisms remain elusive. Given the role of µ-opioid receptors in opioid-induced hyperalgesia in animals, the authors hypothesized that the dorsal reticular nucleus, a medullary pain facilitatory area, is involved in opioid-induced hyperalgesia through altered µ-opioid receptor signaling. METHODS: The authors used male Wistar rats (n = 5 to 8 per group), chronically infused with morphine, to evaluate in the dorsal reticular nucleus the expressions of the µ-opioid receptor and phosphorylated cAMP response element-binding, a downstream marker of excitatory µ-opioid receptor signaling. The authors used pharmacologic and gene-mediated approaches. Nociceptive behaviors were evaluated by the von Frey and hot-plates tests. RESULTS: Lidocaine fully reversed mechanical and thermal hypersensitivity induced by chronic morphine. Morphine-infusion increased µ-opioid receptor, without concomitant messenger RNA changes, and phosphorylated cAMP response element-binding levels at the dorsal reticular nucleus. µ-opioid receptor knockdown in morphine-infused animals attenuated the decrease of mechanical thresholds and heat-evoked withdrawal latencies compared with the control vector (von Frey [mean ± SD]: -17 ± 8% vs. -40 ± 9.0%; P < 0.001; hot-plate: -10 ± 5% vs. -32 ± 10%; P = 0.001). µ-opioid receptor knockdown in control animals induced the opposite (von Frey: -31 ± 8% vs. -17 ± 8%; P = 0.053; hotplate: -24 ± 6% vs. -3 ± 10%; P = 0.001). The µ-opioid receptor agonist (D-ALA2,N-ME-PHE4,GLY5-OL)-enkephalin acetate (DAMGO) decreased mechanical thresholds and did not affect heat-evoked withdrawal latencies in morphine-infused animals. In control animals, DAMGO increased both mechanical thresholds and heat-evoked withdrawal latencies. Ultra-low-dose naloxone, which prevents the excitatory signaling of the µ-opioid receptor, administered alone, attenuated mechanical and thermal hypersensitivities, and coadministered with DAMGO, restored DAMGO analgesic effects and decreased phosphorylated cAMP response element-binding levels. CONCLUSIONS: Chronic morphine shifted µ-opioid receptor signaling from inhibitory to excitatory at the dorsal reticular nucleus, likely enhancing descending facilitation during opioid-induced hyperalgesia in the rat.


Subject(s)
Analgesics, Opioid/pharmacology , Hyperalgesia/chemically induced , Medulla Oblongata/drug effects , Morphine/pharmacology , Receptors, Opioid, mu/drug effects , Animals , Disease Models, Animal , Male , Rats , Rats, Wistar , Signal Transduction/drug effects
17.
Addict Biol ; 25(5): e12806, 2020 09.
Article in English | MEDLINE | ID: mdl-31267641

ABSTRACT

Opioid use among pregnant women is a growing public health concern in the United States. Infants exposed to opioids in utero are at risk of exhibiting neonatal opioid withdrawal syndrome (NOWS). The biological mechanisms underlying short and long-term consequences of in utero opioid exposure and NOWS are unknown. A potential genetic factor is a single-nucleotide polymorphism (SNP) in the mu-opioid receptor gene (OPRM1 A118G). Opioid exposed infants with the G-allele spend less time in hospitals after birth. To determine whether this SNP modulates the neurobehavioral effects of neonatal opioid exposure and withdrawal, we used mice possessing the equivalent Oprm1 SNP (A112G). Pups were treated chronically with saline or morphine from postnatal days (PNDs) 1 to 14, a developmental period equivalent to the third trimester of a human pregnancy and a sensitive period for opioid exposure in rodents. Morphine treatment produced significant developmental delays regardless of genotype and increased total ultrasonic vocalizations in males during spontaneous withdrawal. Animals were aged and tested for anxiety and drug response during adolescence and adulthood, respectively. AA morphine-treated animals showed reduced activity in the marble burying task compared with saline controls; however, this effect was absent in AG and GG animals. As adults, AA males exposed to morphine from PNDs 1 to 14 exhibited enhanced development of locomotor sensitization to morphine, whereas females showed reduced locomotor sensitization. These data suggest the involvement of the Oprm1 SNP for certain outcomes of neonatal opioid exposure and highlight the importance of considering sex and genetic variability for the prognosis of NOWS.


Subject(s)
Analgesics, Opioid/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Opioid-Related Disorders/genetics , Polymorphism, Single Nucleotide/genetics , Prenatal Exposure Delayed Effects/genetics , Receptors, Opioid, mu/genetics , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Polymorphism, Single Nucleotide/drug effects , Pregnancy , Receptors, Opioid, mu/drug effects
18.
Addict Biol ; 25(3): e12765, 2020 05.
Article in English | MEDLINE | ID: mdl-31074060

ABSTRACT

The purpose of this study was to explore the effects of cannabidiol (CBD) on binge drinking and evaluate potential gender-related differences. To this aim, male and female C57BL/6J mice (n = 60 per sex) were exposed to the drinking in the dark (DID) model for 4 weeks (DID-1 to DID-4). Dose-response effects of CBD on the ethanol intake were tested by acute (day-4 of DID-3) or repeated administration (day-1 to 4 of DID-4) (experiment 1: CBD 15, 30, and 60 mg/kg, i.p.; experiment 2: CBD 90 mg/kg, i.p.). Finally, we analyzed the relative gene expression of tyrosine hydroxylase (TH) and µ-opioid receptor (OPRM1) and cannabinoid CB1 receptor (CB1 r) in the ventral tegmental area (VTA) and in the nucleus accumbens (NAc), respectively, by real-time quantitative PCR. Females exhibited higher ethanol intake during each DID session. Interestingly, females also showed higher expression of TH and OPRM1, without any difference in CB1 r. Only the acute administration of CBD at the highest dose (90 mg/kg) reduced significantly ethanol consumption in both sexes. Chronic CBD administration (30, 60 and 90 mg/kg) reduced ethanol intake in males, whereas in females a significant reduction was only achieved with the highest dose (90 mg/kg). Repeated administration with CBD (60 mg/kg) significantly reduced TH and OPRM1 in males. In addition, CBD (30 and 60 mg/kg) significantly reduced CB1 r in males. No effect was observed in females. Taken together, these findings suggest that CBD may be of interest for treating binge-drinking patterns and that gender-related difference may affect the treatment outcome.


Subject(s)
Alcohol Drinking , Behavior, Animal/drug effects , Binge Drinking , Cannabidiol/pharmacology , Central Nervous System Depressants/administration & dosage , Ethanol/administration & dosage , Animals , Female , Male , Mice , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Random Allocation , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/genetics , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/genetics , Self Administration , Sex Factors , Tyrosine 3-Monooxygenase/drug effects , Tyrosine 3-Monooxygenase/genetics
19.
Proc Natl Acad Sci U S A ; 114(20): 5247-5252, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28461466

ABSTRACT

To provide new preclinical evidence toward improving the efficacy of oxytocin (OT) in treating social dysfunction, we tested the benefit of administering OT under simultaneously induced opioid antagonism during dyadic gaze interactions in monkeys. OT coadministered with a µ-opioid receptor antagonist, naloxone, invoked a supralinear enhancement of prolonged and selective social attention, producing a stronger effect than the summed effects of each administered separately. These effects were consistently observed when averaging over entire sessions, as well as specifically following events of particular social importance, including mutual eye contact and mutual reward receipt. Furthermore, attention to various facial regions was differentially modulated depending on social context. Using the Allen Institute's transcriptional atlas, we further established the colocalization of µ-opioid and κ-opioid receptor genes and OT genes at the OT-releasing sites in the human brain. These data across monkeys and humans support a regulatory relationship between the OT and opioid systems and suggest that administering OT under opioid antagonism may boost the therapeutic efficacy of OT for enhancing social cognition.


Subject(s)
Fixation, Ocular/drug effects , Oxytocin/metabolism , Oxytocin/pharmacology , Analgesics, Opioid/antagonists & inhibitors , Animals , Attention/drug effects , Behavior, Animal/drug effects , Female , Macaca mulatta/physiology , Male , Naloxone/metabolism , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid, kappa , Receptors, Opioid, mu/drug effects , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism , Social Behavior
20.
Int J Mol Sci ; 21(13)2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32630190

ABSTRACT

Opioid analgesics such as morphine have indispensable roles in analgesia. However, morphine use can elicit side effects such as respiratory depression and constipation. It has been reported that G protein-biased agonists as substitutes for classic opioid agonists can alleviate (or even eliminate) these side effects. The compounds PZM21 and TRV130 could be such alternatives. Nevertheless, there are controversies regarding the efficacy and G protein-biased ability of PZM21. To demonstrate a rationale for the reduced biasing agonism of PZM21 compared with that of TRV130 at the molecular level, we undertook a long-term molecular dynamics simulation of the µ-opioid receptor (MOR) upon the binding of three ligands: morphine, TRV130, and PZM21. We found that the delayed movement of the W2936.48 (Ballesteros-Weinstein numbering) side chain was a factor determining the dose-dependent agonism of PZM21. Differences in conformational changes of W3187.35, Y3267.43, and Y3367.53 in PZM21 and TRV130 explained the observed differences in bias between these ligands. The extent of water movements across the receptor channel was correlated with analgesic effects. Taken together, these data suggest that the observed differences in conformational changes of the studied MOR-ligand complexes point to the low-potency and lower bias effects of PZM21 compared with the other two ligands, and they lay the foundation for the development of G protein-biased agonists.


Subject(s)
Receptors, Opioid, mu/drug effects , Thiophenes/chemistry , Thiophenes/pharmacology , Urea/analogs & derivatives , Analgesia/methods , Analgesics, Opioid/adverse effects , Animals , Dose-Response Relationship, Drug , GTP-Binding Proteins/metabolism , Humans , Ligands , Molecular Dynamics Simulation , Morphine/metabolism , Morphine/pharmacology , Pain/chemically induced , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/ultrastructure , Spiro Compounds/metabolism , Spiro Compounds/pharmacology , Thiophenes/metabolism , Urea/chemistry , Urea/metabolism , Urea/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL