Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Int J Mol Sci ; 24(9)2023 Apr 27.
Article in English | MEDLINE | ID: mdl-37175678

ABSTRACT

Despite the large arsenal of analgesic medications, neuropathic pain (NP) management is not solved yet. Angiotensin II receptor type 1 (AT1) has been identified as a potential target in NP therapy. Here, we investigate the antiallodynic effect of AT1 blockers telmisartan and losartan, and particularly their combination with morphine on rat mononeuropathic pain following acute or chronic oral administration. The impact of telmisartan on morphine analgesic tolerance was also assessed using the rat tail-flick assay. Morphine potency and efficacy in spinal cord samples of treated neuropathic animals were assessed by [35S]GTPγS-binding assay. Finally, the glutamate content of the cerebrospinal fluid (CSF) was measured by capillary electrophoresis. Oral telmisartan or losartan in higher doses showed an acute antiallodynic effect. In the chronic treatment study, the combination of subanalgesic doses of telmisartan and morphine ameliorated allodynia and resulted in a leftward shift in the dose-response curve of morphine in the [35S]GTPγS binding assay and increased CSF glutamate content. Telmisartan delayed morphine analgesic-tolerance development. Our study has identified a promising combination therapy composed of telmisartan and morphine for NP and opioid tolerance. Since telmisartan is an inhibitor of AT1 and activator of PPAR-γ, future studies are needed to analyze the effect of each component.


Subject(s)
Analgesics, Opioid , Neuralgia , Rats , Animals , Analgesics, Opioid/therapeutic use , Telmisartan/pharmacology , Telmisartan/therapeutic use , Losartan/therapeutic use , Guanosine 5'-O-(3-Thiotriphosphate) , Drug Tolerance , Analgesics/pharmacology , Analgesics/therapeutic use , Morphine/pharmacology , Morphine/therapeutic use , Neuralgia/drug therapy , Glutamates/therapeutic use
2.
Int J Mol Sci ; 22(5)2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33804568

ABSTRACT

The limited effect of current medications on neuropathic pain (NP) has initiated large efforts to develop effective treatments. Animal studies showed that glycine transporter (GlyT) inhibitors are promising analgesics in NP, though concerns regarding adverse effects were raised. We aimed to study NFPS and Org-25543, GlyT-1 and GlyT-2 inhibitors, respectively and their combination in rat mononeuropathic pain evoked by partial sciatic nerve ligation. Cerebrospinal fluid (CSF) glycine content was also determined by capillary electrophoresis. Subcutaneous (s.c.) 4 mg/kg NFPS or Org-25543 showed analgesia following acute administration (30-60 min). Small doses of each compound failed to produce antiallodynia up to 180 min after the acute administration. However, NFPS (1 mg/kg) produced antiallodynia after four days of treatment. Co-treatment with subanalgesic doses of NFPS (1 mg/kg) and Org-25543 (2 mg/kg) produced analgesia at 60 min and thereafter meanwhile increased significantly the CSF glycine content. This combination alleviated NP without affecting motor function. Test compounds failed to activate G-proteins in spinal cord. To the best of our knowledge for the first time we demonstrated augmented analgesia by combining GlyT-1 and 2 inhibitors. Increased CSF glycine content supports involvement of glycinergic system. Combining selective GlyT inhibitors or developing non-selective GlyT inhibitors might have therapeutic value in NP.


Subject(s)
Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Glycine/cerebrospinal fluid , Hyperalgesia/prevention & control , Neuralgia/drug therapy , Sarcosine/analogs & derivatives , Animals , Hyperalgesia/metabolism , Hyperalgesia/pathology , Male , Motor Activity , Neuralgia/metabolism , Neuralgia/pathology , Rats , Rats, Wistar , Sarcosine/pharmacology , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology
3.
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
4.
Molecules ; 25(11)2020 May 26.
Article in English | MEDLINE | ID: mdl-32466522

ABSTRACT

There is growing evidence on the role of peripheral µ-opioid receptors (MORs) in analgesia and analgesic tolerance. Opioid analgesics are the mainstay in the management of moderate to severe pain, and their efficacy in the alleviation of pain is well recognized. Unfortunately, chronic treatment with opioid analgesics induces central analgesic tolerance, thus limiting their clinical usefulness. Numerous molecular mechanisms, including receptor desensitization, G-protein decoupling, ß-arrestin recruitment, and alterations in the expression of peripheral MORs and microbiota have been postulated to contribute to the development of opioid analgesic tolerance. However, these studies are largely focused on central opioid analgesia and tolerance. Accumulated literature supports that peripheral MORs mediate analgesia, but controversial results on the development of peripheral opioid receptors-mediated analgesic tolerance are reported. In this review, we offer evidence on the consequence of the activation of peripheral MORs in analgesia and analgesic tolerance, as well as approaches that enhance analgesic efficacy and decrease the development of tolerance to opioids at the peripheral sites. We have also addressed the advantages and drawbacks of the activation of peripheral MORs on the sensory neurons and gut (leading to dysbiosis) on the development of central and peripheral analgesic tolerance.


Subject(s)
Analgesia , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/therapeutic use , Animals , Humans , Pain/drug therapy , Pain/metabolism , Pain Management/methods
5.
Molecules ; 25(6)2020 Mar 17.
Article in English | MEDLINE | ID: mdl-32192229

ABSTRACT

The present work represents the in vitro (potency, affinity, efficacy) and in vivo (antinociception, constipation) opioid pharmacology of the novel compound 14-methoxycodeine-6-O-sulfate (14-OMeC6SU), compared to the reference compounds codeine-6-O-sulfate (C6SU), codeine and morphine. Based on in vitro tests (mouse and rat vas deferens, receptor binding and [35S]GTPγS activation assays), 14-OMeC6SU has µ-opioid receptor-mediated activity, displaying higher affinity, potency and efficacy than the parent compounds. In rats, 14-OMeC6SU showed stronger antinociceptive effect in the tail-flick assay than codeine and was equipotent to morphine, whereas C6SU was less efficacious after subcutaneous (s.c.) administration. Following intracerebroventricular injection, 14-OMeC6SU was more potent than morphine. In the Complete Freund's Adjuvant-induced inflammatory hyperalgesia, 14-OMeC6SU and C6SU in s.c. doses up to 6.1 and 13.2 µmol/kg, respectively, showed peripheral antihyperalgesic effect, because co-administered naloxone methiodide, a peripherally acting opioid receptor antagonist antagonized the measured antihyperalgesia. In addition, s.c. C6SU showed less pronounced inhibitory effect on the gastrointestinal transit than 14-OMeC6SU, codeine and morphine. This study provides first evidence that 14-OMeC6SU is more effective than codeine or C6SU in vitro and in vivo. Furthermore, despite C6SU peripheral antihyperalgesic effects with less gastrointestinal side effects the superiority of 14-OMeC6SU was obvious throughout the present study.


Subject(s)
Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/pharmacology , Codeine/chemical synthesis , Codeine/pharmacology , Analgesics, Opioid/chemistry , Analgesics, Opioid/therapeutic use , Animals , Binding, Competitive , Codeine/chemistry , Codeine/therapeutic use , Freund's Adjuvant , Gastrointestinal Transit/drug effects , Inflammation/drug therapy , Injections, Intraventricular , Male , Mice , Naloxone/pharmacology , Naloxone/therapeutic use , Nociception/drug effects , Pain/drug therapy , Rats, Wistar , Receptors, Opioid, mu/metabolism
6.
Neurochem Res ; 43(6): 1250-1257, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29725918

ABSTRACT

Opioid analgesics devoid of central side effects are unmet medical need in the treatment of acute pain (e.g. post-operative pain). Recently, we have reported on 14-O-methylmorphine-6-O-sulfate (14-O-MeM6SU), a novel opioid agonist of high efficacy producing peripheral antinociception in subchronic inflammatory pain in certain doses. The present study focused on the antinociceptive effect of 14-O-MeM6SU compared to morphine in formalin test of an early/acute (Phase I) and late/tonic (Phase II) pain phases. Subcutaneous 14-O-MeM6SU (253-1012 nmol/kg) and morphine (3884-31075 nmol/kg) dose dependently reduced the pain behaviors of both phases. Co-administered naloxone methiodide (NAL-M), a peripherally acting opioid antagonist, abolished the antinociceptive effect of 506 nmol/kg 14-O-MeM6SU. On the other hand, the effects of 14-O-MeM6SU (1012 nmol/kg) and morphine (15538 nmol/kg) were only partially affected by NAL-M, indicating the contribution of CNS to antinociception. Locally injected test compounds into formalin treated paws caused antinociception in both phases. Locally effective doses of test compounds were also injected into contralateral paws. Morphine showed effects in both phases, 14-O-MeM6SU in certain doses failed to produce antinociception in either phase. A NAL-M reversible systemic dose of 14-O-MeM6SU and the lowest systemic effective dose of morphine were evaluated for their sedative effects following isoflurane-induced sleeping (righting reflex). In contrast to morphine, 14-O-MeM6SU in certain antinociceptive doses showed no impact on sleeping time. These data highlight that high efficacy opioids of limited CNS penetration in certain doses mitigate somatic and inflammatory pain by targeting MOR at the periphery.


Subject(s)
Acute Pain/drug therapy , Analgesics, Opioid/administration & dosage , Analgesics/administration & dosage , Codeine/analogs & derivatives , Pain Measurement/drug effects , Acute Pain/metabolism , Acute Pain/psychology , Analgesics/chemistry , Analgesics, Opioid/chemistry , Animals , Codeine/administration & dosage , Codeine/chemistry , Dose-Response Relationship, Drug , Inflammation/drug therapy , Inflammation/metabolism , Injections, Subcutaneous , Male , Pain Measurement/methods , Rats , Rats, Wistar
7.
J Pharmacol Exp Ther ; 359(1): 171-81, 2016 10.
Article in English | MEDLINE | ID: mdl-27435180

ABSTRACT

Growing data support peripheral opioid antinociceptive effects, particularly in inflammatory pain models. Here, we examined the antinociceptive effects of subcutaneously administered, recently synthesized 14-O-methylmorphine-6-O-sulfate (14-O-MeM6SU) compared with morphine-6-O-sulfate (M6SU) in a rat model of inflammatory pain induced by an injection of complete Freund's adjuvant and in a mouse model of visceral pain evoked by acetic acid. Subcutaneous doses of 14-O-MeM6SU and M6SU up to 126 and 547 nmol/kg, respectively, produced significant and subcutaneous or intraplantar naloxone methiodide (NAL-M)-reversible antinociception in inflamed paws compared with noninflamed paws. Neither of these doses significantly affected thiobutabarbital-induced sleeping time or rat pulmonary parameters. However, the antinociceptive effects of higher doses were only partially reversed by NAL-M, indicating contribution of the central nervous system. In the mouse writhing test, 14-O-MeM6SU was more potent than M6SU after subcutaneous or intracerebroventricular injections. Both displayed high subcutaneous/intracerebroventricular ED50 ratios. The antinociceptive effects of subcutaneous 14-O-MeM6SU and M6SU up to 136 and 3043 nmol/kg, respectively, were fully antagonized by subcutaneous NAL-M. In addition, the test compounds inhibited mouse gastrointestinal transit in antinociceptive doses. Taken together, these findings suggest that systemic administration of the novel compound 14-O-MeM6SU similar to M6SU in specific dose ranges shows peripheral antinociception in rat and mouse inflammatory pain models without central adverse effects. These findings apply to male animals and must be confirmed in female animals. Therefore, titration of systemic doses of opioid compounds with limited access to the brain might offer peripheral antinociception of clinical importance.


Subject(s)
Analgesics/administration & dosage , Analgesics/pharmacology , Morphine/administration & dosage , Morphine/pharmacology , Analgesics/chemistry , Analgesics/therapeutic use , Animals , Dose-Response Relationship, Drug , Drug Interactions , Female , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/physiology , Male , Mice , Morphine/chemistry , Morphine/therapeutic use , Pain/drug therapy , Rats , Rats, Wistar , Respiration/drug effects , Thiopental/analogs & derivatives , Thiopental/pharmacology
8.
Neuropsychopharmacol Hung ; 15(4): 189-205, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24380960

ABSTRACT

Much progress has been made in the last decade in the understanding the neural substrates of drug addiction, transmitters involved, epigenetic background and their relation to learning and memory but much remains to be elucidated and strong effort is necessary to integrate the rich information at the molecular, cellular systems, and behavioral levels to further clarify the mechanisms and therapy of this complex disease. The aim of this review is to collect and interpret the latest opinions in the development, the underlying mechanisms and therapy of addiction as a disease of central nervous system. The neurocircuitry, the transmitters and the epigenetics of addiction are discussed.


Subject(s)
Behavior, Addictive/metabolism , Brain/metabolism , Chromatin/metabolism , Nerve Tissue Proteins/metabolism , Psychotropic Drugs/therapeutic use , Substance-Related Disorders/drug therapy , Substance-Related Disorders/metabolism , Alcoholism/drug therapy , Alcoholism/metabolism , Behavior, Addictive/genetics , Benzodiazepines/adverse effects , Brain/physiopathology , Brain-Derived Neurotrophic Factor/metabolism , Central Nervous System Stimulants/adverse effects , Chromatin/genetics , Epigenesis, Genetic , Humans , Learning , Marijuana Abuse/drug therapy , Marijuana Abuse/metabolism , Memory , Neurotransmitter Agents/metabolism , Neurotransmitter Agents/pharmacology , Opioid-Related Disorders/drug therapy , Opioid-Related Disorders/metabolism , Psychotropic Drugs/pharmacology , Reward , Signal Transduction/drug effects
9.
Neuropsychopharmacol Hung ; 15(4): 239-51, 2013 Dec.
Article in Hungarian | MEDLINE | ID: mdl-24380965

ABSTRACT

UNLABELLED: Opioids impair the maternal behaviour of experimental animals. The effect of morphine on maternal behaviour in rat dams treated chronically with morphine during the whole pregnancy and lactation has not been yet analysed systematically. OBJECTIVE: The aim of our work was to investigate the behavioural effects of moderate dose morphine administered constantly in the whole perinatal period in rats. METHODS: Nulliparous female rats were treated with 10 mg/kg morphine s.c. once daily, from the day of mating. Maternal behaviour was observed, the effects of acute morphine treatment on the maternal behaviour and whether this effect could be antagonised by naloxone were also investigated. Physical and other behavioural (anxiety-like signals in elevated plus maze, changes in locomotor activity) withdrawal signs precipitated by naloxone were registered. After weaning sensitivity to the rewarding effect of morphine was measured by conditioned place preference and to the aversive effect of naloxone by conditioned place aversion tests. Antinociceptive test on tail-flick apparatus was performed to investigate the changes in morphine antinociceptive effects due to chronic morphine treatment. RESULTS: Maternal behaviour was significantly impaired in morphine-treated dams. This effect of morphine lasted c.a. 2-3 hours a day, it showed dose-dependency and was enhanced in MO-treated group (sensitisation). Only weak physical and no other behavioural (anxiety-like behaviour or hypolocomotion) withdrawal signs were precipitated by naloxone. The positive reinforcing effect of morphine and aversive effect of naloxone were markedly increased on conditioned place paradigm. Significant antinociceptive tolerance was not seen. CONCLUSION: Although human drug abuse can be hardly modelling under experimental circumstances, our constant, relatively moderate dose morphine treatment administered once daily during the whole pregnancy and lactation resulted in several subtle behavioural changes in dams. In perinatally opioid-exposed offspring short- and long-term behavioural disturbances can be detected which is well-known from literature. Besides direct pharmacological effects of morphine impaired maternal responsiveness and pup care could play a role in these disturbances.


Subject(s)
Behavior, Animal/drug effects , Drug Tolerance , Lactation/drug effects , Morphine/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Narcotics/pharmacology , Substance-Related Disorders , Animals , Female , Maternal Behavior/drug effects , Morphine/administration & dosage , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Narcotics/administration & dosage , Pregnancy , Rats , Rats, Wistar , Substance-Related Disorders/diagnosis
10.
Pharmaceuticals (Basel) ; 16(8)2023 Aug 07.
Article in English | MEDLINE | ID: mdl-37631030

ABSTRACT

The current treatment of neuropathic pain (NP) is unsatisfactory; therefore, effective novel agents or combination-based analgesic therapies are needed. Herein, oral tolperisone, pregabalin, and duloxetine were tested for their antinociceptive effect against rat partial sciatic nerve ligation (pSNL)-induced tactile allodynia described by a decrease in the paw withdrawal threshold (PWT) measured by a dynamic plantar aesthesiometer. On day 7 after the operation, PWTs were assessed at 60, 120, and 180 min post-treatment. Chronic treatment was continued for 2 weeks, and again, PWTs were measured on day 14 and 21. None of the test compounds produced an acute antiallodynic effect. In contrast, after chronic treatment, tolperisone and pregabalin alleviated allodynia. In other experiments, on day 14, the acute antiallodynic effect of the tolperisone/pregabalin or duloxetine combination was measured. As a novel finding, a single dose of the tolperisone/pregabalin combination could remarkably alleviate allodynia acutely. It also restored the neuropathy-induced elevated CSF glutamate content. Furthermore, the combination is devoid of adverse effects related to motor and gastrointestinal transit functions. Tolperisone and pregabalin target voltage-gated sodium and calcium channels, respectively. The dual blockade effect of the combination might explain its advantageous acute analgesic effect in the present work.

11.
Behav Pharmacol ; 21(1): 58-68, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20038835

ABSTRACT

This study investigated the behavioural consequences of peri, pre and postnatal morphine (MO) exposure in rats. From gestational day 1 dams were treated with either saline or MO subcutaneously once a day (5 mg/kg on the first 2 days, 10 mg/kg subsequently). Spontaneous locomotor activity in a new environment (habituation) and antinociceptive effects of MO were measured separately in male and female pups after weaning and also in late adolescence or adulthood. The rewarding effect of MO was assessed by conditioned place preference in adult animals. Both exposure-induced and sex differences were observed. A significant delay in habituation to a new environment and decreased sensitivity to the antinociceptive effect of MO were found in male offspring of MO-treated dams. In contrast, the place preference induced by MO was enhanced in the MO-exposed adult animals and this effect was more marked in females. Prenatal exposure to MO resulted in more marked changes than the postnatal exposure through maternal milk. The results indicate that a medium MO dose administered once-daily results in long-term consequences in offspring and may make them more vulnerable to MO abuse in adulthood.


Subject(s)
Habituation, Psychophysiologic/drug effects , Morphine/adverse effects , Narcotics/adverse effects , Prenatal Exposure Delayed Effects/psychology , Reward , Sex Characteristics , Animals , Animals, Suckling , Birth Weight/drug effects , Choice Behavior/drug effects , Conditioning, Operant/drug effects , Female , Male , Morphine/pharmacology , Narcotics/pharmacology , Pain Threshold/drug effects , Pregnancy , Rats , Rats, Wistar , Time Factors
12.
Brain Res Bull ; 147: 78-85, 2019 04.
Article in English | MEDLINE | ID: mdl-30738866

ABSTRACT

Dipeptidyl-peptidase 4 (DPP4) enzyme is involved in the degradation of many biologically active peptides including opioids. Its role in pain transmission is poorly elucidated. Recently we reported on the spinal antihyperalgesic effects of DPP4 inhibitors, Ile-Pro-Ile (Diprotin A) and vildagliptin in carrageenan-evoked acute inflammatory pain in rats. The present study investigated the effects of intrathecal (it.) diprotin A and vildagliptin in Complete Freund's Adjuvant- (CFA) and formalin induced pain in rats. The former assay can model the subchronic inflammatory pain condition and the later one reflects both acute tonic and inflammatory pain conditions. The involvement of opioid receptor (OR) subtypes, Y1-, and GLP1 receptors were also investigated. In CFA pain model it. diprotin A or vildagliptin dose-dependently inhibits hyperalgesia in ipsilateral while has no effect in contralateral paws. The peak effect was achieved 30 min following drug administration which was used for further analysis. Both compounds showed naltrexone reversible antihyperalgesia. Co-administration of OR-subtype-selective antagonists with diprotin A and vildagliptin revealed involvement of µ and δ > µ opioid receptors, respectively. Co-administered Y1 but not GLP1 receptor antagonists reversed the antihyperalgesic action of both DPP4 inhibitors. In touch-hypersensitivity both compounds were ineffective. In formalin test only diprotin A showed µ and δ OR-mediated antinociception and only in the 2nd phase. This effect was Y1 or GLP-1 receptor antagonist insensitive. In conclusion, diprotin A and vildagliptin display antinociception of different mechanisms of action in subchronic inflammatory pain. Furthermore, the spinal pain relay points of inflammatory pain of acute or subchronic conditions were more effectively affected by diprotin A than vildagliptin which needs future elucidation.


Subject(s)
Oligopeptides/pharmacology , Pain/drug therapy , Vildagliptin/pharmacology , Analgesics/pharmacology , Analgesics, Opioid/pharmacology , Animals , Dipeptidyl-Peptidase IV Inhibitors/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Hyperalgesia/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Male , Narcotic Antagonists/pharmacology , Oligopeptides/metabolism , Pain/physiopathology , Pain Measurement , Rats , Rats, Wistar , Receptors, Opioid/metabolism , Receptors, Opioid, mu , Vildagliptin/metabolism
13.
Front Pharmacol ; 10: 347, 2019.
Article in English | MEDLINE | ID: mdl-31024314

ABSTRACT

Reduction of the opioid analgesia in diabetic neuropathic pain (DNP) results from µ-opioid receptor (MOR) reserve reduction. Herein, we examined the antinociceptive and antiallodynic actions of a novel opioid agonist 14-O-methymorphine-6-O-sulfate (14-O-MeM6SU), fentanyl and morphine in rats with streptozocin-evoked DNP of 9-12 weeks following their systemic administration. The antinociceptive dose-response curve of morphine but not of 14-O-MeM6SU or fentanyl showed a significant right-shift in diabetic compared to non-diabetic rats. Only 14-O-MeM6SU produced antiallodynic effects in doses matching antinociceptive doses obtained in non-diabetic rats. Co-administered naloxone methiodide (NAL-M), a peripherally acting opioid receptor antagonist failed to alter the antiallodynic effect of test compounds, indicating the contribution of central opioid receptors. Reduction in spinal MOR binding sites and loss in MOR immunoreactivity of nerve terminals in the spinal cord and dorsal root ganglia in diabetic rats were observed. G-protein coupling assay revealed low efficacy character for morphine and high efficacy character for 14-O-MeM6SU or fentanyl at spinal or supraspinal levels (E max values). Furthermore, at the spinal level only 14-O-MeM6SU showed equal efficacy in G-protein activation in tissues of diabetic- and non-diabetic animals. Altogether, the reduction of spinal opioid receptors concomitant with reduced analgesic effect of morphine may be circumvented by using high efficacy opioids, which provide superior analgesia over morphine. In conclusion, the reduction in the analgesic action of opioids in DNP might be a consequence of MOR reduction, particularly in the spinal cord. Therefore, developing opioids of high efficacy might provide analgesia exceeding that of currently available opioids.

14.
Brain Res Bull ; 74(5): 369-75, 2007 Oct 19.
Article in English | MEDLINE | ID: mdl-17845912

ABSTRACT

Peripheral micro-opioid receptors (MOR) have emerged as important components of inhibitory nociceptive pathways. Here, the antinociceptive effects of MOR agonists, the 6beta-glycine derivative of 14-O-methyloxymorphone (HS-731), DAMGO and morphine were evaluated in a mouse model of visceral pain. The abdominal acetic acid-induced writhing test was used to examine the peripheral, preemptive antinociceptive opioid action on visceral nociception. HS-731 administered subcutaneously (s.c.) or intracerebroventricularly (i.c.v.) dose-dependently and completely inhibited writhing, being 24-598-fold more potent, depending on the administration route, than two selective MOR agonists, the enkephalin analogue [D-Ala(2),N-Me-Phe(4),Gly-ol(5)]enkephalin (DAMGO) and morphine. A longer duration of action (2-3 h) was induced by HS-731 given before acetic acid, while shorter effect was produced by morphine (30-60 min) and DAMGO (30-45 min). The antinociceptive effects of systemic opioids were reversed by the s.c. opioid antagonist, naloxone. Blocking of central MOR by the selective MOR antagonist D-Phe-Cys-Tyr-d-Trp-Arg-Thr-Pen-Thr-NH(2) (CTAP, i.c.v.) resulted in a significant reduction of antinociception of s.c. morphine, whereas it completely failed to antagonize the effects of systemic HS-731 or DAMGO. In in vitro studies, HS-731 and DAMGO, but not morphine showed high intrinsic efficacy, naltrexone-sensitive agonist effect at MOR of the rat vas deferens. These data demonstrate that selective activation of peripheral MOR by systemic s.c. HS-731 or DAMGO produces potent peripheral, preemptive visceral antinociception, while morphine's effects are mediated primarily through central mechanisms. Our findings support the role of peripheral MOR in the pathology of pain states involving sensitization of peripheral nociceptors.


Subject(s)
Analgesics, Opioid/administration & dosage , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage , Epoxy Compounds/administration & dosage , Morphinans/administration & dosage , Morphine/administration & dosage , Pain/drug therapy , Vas Deferens/drug effects , Acetic Acid , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Routes , Drug Administration Schedule , Drug Interactions , In Vitro Techniques , Male , Mice , Pain/chemically induced , Pain Measurement , Peptide Fragments , Peptides/therapeutic use , Rats , Rats, Wistar , Somatostatin , Time Factors , Vas Deferens/physiology
15.
Eur J Pharmacol ; 814: 264-273, 2017 Nov 05.
Article in English | MEDLINE | ID: mdl-28864212

ABSTRACT

14-O-methyl (14-O-Me) group in morphine-6-O-sulfate (M6SU) or oxymorphone has been reported to be essential for enhanced affinity, potency and antinociceptive effect of these opioids. Herein we report on the pharmacological properties (potency, affinity and efficacy) of the new compound, 14-O-methylmorphine (14-O-MeM) in in vitro. Additionally, we also investigated the antinociceptive effect of the novel compound, as well as its inhibitory action on gastrointestinal transit in in vivo. The potency and efficacy of test compound were measured by [35S]GTPγS binding, isolated mouse vas deferens (MVD) and rat vas deferens (RVD) assays. The affinity of 14-O-MeM for opioid receptors was assessed by radioligand binding and MVD assays. The antinociceptive and gastrointestinal effects of the novel compound were evaluated in the rat tail-flick test and charcoal meal test, respectively. Morphine, DAMGO, Ile5,6 deltorphin II, deltorphin II and U-69593 were used as reference compounds. 14-O-MeM showed higher efficacy (Emax) and potency (EC50) than morphine in MVD, RVD or [35S]GTPγS binding. In addition, 14-O-MeM compared to morphine showed higher affinity for µ-opioid receptor (MOR). In vivo, in rat tail-flick test 14-O-MeM proved to be stronger antinociceptive agent than morphine after peripheral or central administration. Additionally, both compounds inhibited the gastrointestinal peristalsis. However, when the antinociceptive and antitransit doses for each test compound are compared, 14-O-MeM proved to have slightly more favorable pharmacological profile. Our results affirm that 14-O-MeM, an opioid of high efficacy and affinity for MOR can be considered as a novel analgesic agent of potential clinical value.


Subject(s)
Analgesics/metabolism , Analgesics/pharmacology , Codeine/metabolism , Codeine/pharmacology , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Animals , Binding, Competitive , Gastrointestinal Transit/drug effects , Inhibitory Concentration 50 , Male , Mice , Rats , Substrate Specificity , Vas Deferens/drug effects , Vas Deferens/metabolism
16.
Eur J Pharmacol ; 809: 111-121, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28502630

ABSTRACT

Opioid antagonists, naloxone and naltrexone have long been used in clinical practice and research. In addition to their low selectivity, they easily pass through the blood-brain barrier. Quaternization of the amine group in these molecules, (e.g. methylnaltrexone) results in negligible CNS penetration. In addition, zwitterionic compounds have been reported to have limited CNS access. The current study, for the first time gives report on the synthesis and the in vitro [competition binding, G-protein activation, isolated mouse vas deferens (MVD) and mouse colon assay] pharmacology of the zwitterionic compound, naltrexone-14-O-sulfate. Naltrexone, naloxone, and its 14-O-sulfate analogue were used as reference compounds. In competition binding assays, naltrexone-14-O-sulfate showed lower affinity for µ, δ or κ opioid receptor than the parent molecule, naltrexone. However, the µ/κ opioid receptor selectivity ratio significantly improved, indicating better selectivity. Similar tendency was observed for naloxone-14-O-sulfate when compared to naloxone. Naltrexone-14-O-sulfate failed to activate [35S]GTPγS-binding but inhibit the activation evoked by opioid agonists (DAMGO, Ile5,6deltorphin II and U69593), similarly to the reference compounds. Schild plot constructed in MVD revealed that naltrexone-14-O-sulfate acts as a competitive antagonist. In mouse colon, naltrexone-14-O-sulfate antagonized the inhibitory effect of morphine with lower affinity compared to naltrexone and higher affinity when compared to naloxone or naloxone-14-O-sulfate. In vivo (mouse tail-flick test), subcutaneously injected naltrexone-14-O-sulfate antagonized morphine's antinociception in a dose-dependent manner, indicating it's CNS penetration, which was unexpected from such zwitter ionic structure. Future studies are needed to evaluate it's pharmacokinetic profile.


Subject(s)
Analgesics/chemical synthesis , Analgesics/pharmacology , Naltrexone/chemical synthesis , Naltrexone/pharmacology , Narcotic Antagonists/chemical synthesis , Narcotic Antagonists/pharmacology , Receptors, Opioid/metabolism , Animals , Behavior, Animal/drug effects , Chemistry Techniques, Synthetic , Colon/drug effects , Colon/metabolism , Male , Mice , Morphine/pharmacology , Naltrexone/analogs & derivatives , Rats , Rats, Wistar , Vas Deferens/drug effects , Vas Deferens/metabolism
17.
Brain Res Bull ; 67(1-2): 126-32, 2005 Sep 30.
Article in English | MEDLINE | ID: mdl-16140171

ABSTRACT

Norfluoxetine is the most important active metabolite of the widely used antidepressant fluoxetine but little is known about its pharmacological actions. In this study the anticonvulsant actions of norfluoxetine and fluoxetine were studied and compared to those of phenytoin and clonazepam in pentylenetetrazol-induced mouse epilepsy models. Pretreatment with fluoxetine or norfluoxetine (20mg/kg s.c.), as well as phenytoin (30 mg/kg s.c.) and clonazepam (0.1mg/kg s.c.) significantly increased both the rate and duration of survival, demonstrating a significant protective effect against pentylenetetrazol-induced epilepsy. These effects of norfluoxetine were similar to those of fluoxetine. According to the calculated combined protection scores, both norfluoxetine and fluoxetine were effective from the concentration of 10mg/kg, while the highest protective action was observed with clonazepam. Effects of norfluoxetine and fluoxetine on voltage-gated Ca2+ channels were evaluated by measuring peak Ba2+ current flowing through the Ca2+ channels upon depolarization using whole cell voltage clamp in enzymatically isolated rat cochlear neurons. The current was reduced equally in a concentration-dependent manner by norfluoxetine (EC50=20.4+/-2.7 microM, Hill coefficient=0.86+/-0.1) and fluoxetine (EC50=22.3+/-3.6 microM, Hill coefficient=0.87+/-0.1). It was concluded that the efficacy of the two compounds in neuronal tissues was equal, either in preventing seizure activity or in blocking the neuronal Ca2+ channels.


Subject(s)
Calcium Channels/drug effects , Cochlear Nucleus/drug effects , Epilepsy/drug therapy , Fluoxetine/analogs & derivatives , Fluoxetine/pharmacology , Neurons, Afferent/drug effects , Animals , Anticonvulsants/pharmacology , Barium/metabolism , Barium/pharmacology , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Clonazepam/pharmacology , Cochlear Nucleus/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Epilepsy/chemically induced , Epilepsy/physiopathology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Neurons, Afferent/metabolism , Patch-Clamp Techniques , Phenytoin/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology
18.
Brain Res Bull ; 117: 32-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26235542

ABSTRACT

The therapeutic use of opioids is limited by the development of tolerance to the analgesic effect and the cellular and molecular mechanisms underlying this phenomenon are still not completely understood. For this reason the search for new analgesic derivatives, endowed with lower tolerance, is always an active field. The newly synthesized 14-O-Methylmorphine-6-sulfate (14-O-MeM6SU) shows high efficacy in in vitro assays and a strong analgesic action in the rat tail flick test. The aim of present work was to investigate: the analgesic effect of 14-O-MeM6SU in mouse tail-flick test; the tolerance to analgesic effect of 14-O-MeM6SU compared to morphine in mice, the effects of test compounds on glutamatergic neurotransmission by measuring spontaneous excitatory postsynaptic currents (sEPSCs) of layer V pyramidal cells from rat prefrontal cortices; and the effect of acute and chronic 14-O-MeM6SU treatments on opioid receptor gene expression in SH-SY5Y neuroblastoma cells expressing µ-opioid (MOP) and nociceptin/opioid receptor-like 1 (NOP) receptors. 14-O-MeM6SU was 17 times more potent than morphine in analgesia and had long duration of action in analgesic dose equipotent to morphine. Mice were treated subcutaneously (s.c.) either with 200 µmol/kg morphine or with 14-O-MeM6SU (12 µmol/kg) twice daily for three days. The magnitude of tolerance or cross-tolerance indicated by the shift in antinociceptive ED50 measured was greater for morphine compared to 14-O-MeM6SU. Subsequent to behavioral testing, patch-clamp experiments in layer V pyramidal neurons of rat prefrontal cortical slices in the presence of bicuculline were performed. Both 14-O-MeM6SU (0.1 µM) and morphine (1 µM) decreased the frequency of sEPSCs, indicating reduction of glutamate release. The effect of the novel compound was reversed by the opioid receptor antagonist naloxone, indicating an opioid mediated action. In contrast, the amplitude was not affected. Finally, gene expression data showed a dose dependent down-regulation of MOP receptor after 24h and 48 h exposure to 14-O-MeM6SU. Interestingly, no changes were detected for NOP receptor gene expression. The specific lack of this effect could be related to the lower tolerance development to analgesic effect of 14-O-MeM6SU. Furthermore, 14-O-MeM6SU displayed high intrinsic efficacy possibly an important factor in the observed effects. Further, the observed inhibition of glutamatergic signaling might be attributed also to the reduction of opioid tolerance. Based on our results the development of a new clinically important, safe analgesic agent might be possible.


Subject(s)
Analgesics, Opioid/pharmacology , Codeine/analogs & derivatives , Morphine/pharmacology , Analgesics, Opioid/adverse effects , Animals , Cell Line, Tumor , Codeine/adverse effects , Codeine/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Tolerance , Excitatory Postsynaptic Potentials/drug effects , Glutamic Acid/metabolism , Humans , Male , Mice , Morphine/adverse effects , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nociceptive Pain/drug therapy , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats, Wistar , Receptors, Opioid/genetics , Receptors, Opioid/metabolism , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tissue Culture Techniques , Nociceptin Receptor
19.
Curr Med Chem ; 9(1): 31-9, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11860345

ABSTRACT

The clinical effectiveness of morphine is limited by several side effects, including the development of tolerance and dependence. Most of these side effects are believed to be mediated by central opioid receptors; therefore, hydrophilic opioids, which don't cross the blood-brain barrier, may have advantages over morphine in some clinical applications. We recently synthesized several analogues of DALDA (Tyr-D-Arg-Phe-Lys-NH2), a highly hydrophilic peptide derived from the endogenous opioid peptide dermorphin; all of them, particularly [Dmt(1)] DALDA (Dmt - 2',6'-dimethyl tyrosine), had high potency and selectivity at mu receptors, the target of morphine, in activity assays. Here we report the pharmacological characterization of [Dmt(1)] DALDA in the whole animal. [Dmt(1)]DALDA was 40 times more potent than morphine in inducing antinociception in mice when both drugs were given s.c., and 6-14 times more potent than DAMGO, a selective m agonist, when both drugs were given it. However, [Dmt(1)]DALDA showed poor cross-tolerance to morphine; thus chronic morphine treatment of animals increased the antinociceptive AD(50) of systemic [Dmt(1)]DALDA two fold or less, as compared to an 8-9-fold increase for morphine and a 4-5-fold increase for DAMGO. The antinociceptive activity of [Dmt(1)]DALDA (i.t) was blocked by CTAP, a selective mu antagonist, but not by TIPP psi, a selective delta antagonist, nor by nor-BNI, a selective kappa antagonist. [Dmt(1)]DALDA-induced antinociception was also blocked by naloxone methiodide, an antagonist that does not cross the blood-brain barrier, when agonist and antagonist were given i.t. or i.c.v., but not when they were given s.c. We conclude that [Dmt(1)] DALDA is a highly potent analgesic acting at mu receptors. Though it appears to penetrate the blood-brain barrier, it exhibits low cross-tolerance to morphine, suggesting that it may have advantages over the latter in certain clinical applications.


Subject(s)
Analgesics, Opioid/pharmacology , Morphine/pharmacology , Oligopeptides/pharmacology , Receptors, Opioid, mu/agonists , Analgesics, Opioid/antagonists & inhibitors , Analgesics, Opioid/chemical synthesis , Animals , Drug Tolerance , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Injections, Intraventricular , Injections, Spinal , Injections, Subcutaneous , Male , Mice , Mice, Inbred ICR , Morphine/administration & dosage , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Oligopeptides/antagonists & inhibitors , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Pain Measurement/drug effects , Spinal Cord/drug effects , Spinal Cord/metabolism , Time Factors
20.
Eur J Pharmacol ; 483(2-3): 301-8, 2004 Jan 12.
Article in English | MEDLINE | ID: mdl-14729121

ABSTRACT

A series of 6-amino acid conjugates (glycine, alanine and phenylalanine) of the highly potent opioid analgesic 14-O-methyloxymorphone was developed in an effort to obtain agonists that would have potentially limited ability to cross the blood-brain barrier. Binding studies revealed that all derivatives displayed high affinities (0.77-2.58 nM) at the mu-opioid receptor in rat brain membranes. They were potent agonists in mouse vas deferens preparation (IC(50)=5.52-26.8 nM). While the alpha-amino acid epimers are favoured by mu-opioid receptors, the beta-epimers proved to have increased interaction with delta-sites. Only the beta-phenylalanine conjugate showed some preference for delta- over mu-opioid receptors and delta-opioid receptor agonist activity. The relatively high delta-opioid receptor affinity of this analogue was also predicted by molecular modelling studies. The newly developed ionizable derivatives could find clinical applications as potent analgesics without the adverse actions of centrally acting opioids.


Subject(s)
Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Oxymorphone/chemistry , Oxymorphone/pharmacology , Analgesics, Opioid/metabolism , Animals , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Mice , Narcotic Antagonists/metabolism , Narcotic Antagonists/pharmacology , Oxymorphone/metabolism , Protein Binding/drug effects , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Vas Deferens/drug effects , Vas Deferens/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL