1
|
Arai I, Tsuji M, Saito S, Takeda H. Experimental Study: Interleukin-31 Augments Morphine-Induced Antinociceptive Activity and Suppress Tolerance Development in Mice. Int J Mol Sci 2023; 24:16548. [PMID: 38003738 PMCID: PMC10671644 DOI: 10.3390/ijms242216548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Morphine-induced antinociception is partially reduced in interleukin-31 (IL-31) receptor A (IL-31RA)-deficient mice, indicating that IL-31RA is crucial for morphine-induced peripheral antinociception. Herein, we examined the combined effects of IL-31 and morphine on the antinociceptive activity and itch-associated scratching behavior (LLS) in mice and elucidated the regulatory mechanisms. A hot-plate test was used to assess antinociception. LLS was automatically detected and recorded via a computer. IL-31RA mRNA expression was assessed using real-time polymerase chain reaction. Repeated pre-treatment with IL-31 resulted in significant antinociceptive activity. Repeated administration of morphine decreased the morphine-induced antinociceptive activity, LLS counts, and regular dose and inhibited IL-31-induced LLS. These results suggested that the repeated administration of morphine depleted inter-neuronal IL-31RA levels, preventing morphine-induced antinociception. Therefore, IL-31 may be helpful as an adjunct analgesic to morphine. To explore the benefits of IL-31, its influence on morphine-induced antinociceptive tolerance in mice was examined. An IL-31 and morphine combination increased the analgesic action, which increased the expression of DRG neuronal IL-31RA, elucidating the site of peripheral antinociception of morphine. This site may induce exocytosis of IL-31RA in the sensory nervous system. Collectively, the suppressive effect of IL-31 on morphine-induced antinociceptive tolerance may result from IL-31RA supplementation in sensory nerves.
Collapse
Affiliation(s)
- Iwao Arai
- Department of Pharmacology, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara 324-8510, Japan
- Division of Environmental Allergy, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Tokyo 105-8461, Japan
| | - Minoru Tsuji
- Department of Pharmacology, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara 324-8510, Japan
| | - Saburo Saito
- Division of Environmental Allergy, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Tokyo 105-8461, Japan
| | - Hiroshi Takeda
- Department of Pharmacology, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara 324-8510, Japan
| |
Collapse
|
2
|
Arai I, Tsuji M, Takahashi K, Saito S, Takeda H. Analyzing the Antinociceptive Effect of Interleukin-31 in Mice. Int J Mol Sci 2023; 24:11563. [PMID: 37511321 PMCID: PMC10380705 DOI: 10.3390/ijms241411563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/04/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
The theory that an itch inhibits pain has been refuted; however, previous research did not investigate this theory for an interleukin-31 (IL-31)-induced itch. Previously, we have found that morphine-induced antinociception was partially reduced in IL-31 receptor A (IL-31RA)-deficient (IL-31RAKI) mice, indicating that IL-31RA may play an important role in morphine-induced peripheral antinociception. In the present study, we evaluated the effect of IL-31-induced analgesia on a 2,4,6-trinitrochlorobenzene (TNCB)-sensitized mice using a hot-plate test. This test evaluated the antinociceptive activity of morphine and non-steroidal anti-inflammatory drugs (NSAIDs). Repeated pretreatment with IL-31 showed significant antinociceptive action. Furthermore, its combination with morphine, but not with NSAIDs, increased the analgesic action. In contrast, treatment with TNCB and capsaicin decreased antinociception. Moreover, TNCB increased IL-31RA expression in the dorsal root ganglia at 24 h, whereas capsaicin inhibited it. The comparative action of several analgesics on TNCB or capsaicin was evaluated using a hot-plate test, which revealed that the antinociceptive activity was decreased or disappeared in response to capsaicin-induced pain in IL-31RAKI mice. These results indicate that the analgesic action of IL-31 involves the peripheral nervous system, which affects sensory nerves. These results provide a basis for developing novel analgesics using this mechanism.
Collapse
Affiliation(s)
- Iwao Arai
- Department of Pharmacology, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara 324-8510, Japan
- Division of Environmental Allergy, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Tokyo 105-8461, Japan
| | - Minoru Tsuji
- Department of Pharmacology, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara 324-8510, Japan
| | - Kohei Takahashi
- Department of Pharmacology, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara 324-8510, Japan
| | - Saburo Saito
- Division of Environmental Allergy, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Tokyo 105-8461, Japan
| | - Hiroshi Takeda
- Department of Pharmacology, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara 324-8510, Japan
| |
Collapse
|
3
|
Heitmann PT, Keightley L, Wiklendt L, Wattchow DA, Brookes SSJ, Spencer NJ, Costa M, Dinning PG. The effects of loperamide on excitatory and inhibitory neuromuscular function in the human colon. Neurogastroenterol Motil 2022; 34:e14442. [PMID: 36054796 DOI: 10.1111/nmo.14442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 06/15/2022] [Accepted: 07/21/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND In most animal species, opioids alter colonic motility via the inhibition of excitatory enteric motor neurons. The mechanisms by which opioids alter human colonic motility are unclear. The aim of this study was to describe the effects of loperamide on neuromuscular function in the human colon. METHODS Tissue specimens of human colon from 10 patients undergoing an anterior resection were divided into three inter-taenial circular muscle strips. Separate organ baths were used to assess: (1) excitatory transmission (selective blockade of inhibitory transmission: L-NOARG/MRS2179); (2) inhibitory transmission (selective blockade of excitatory transmission: hyoscine hydrobromide); and (3) a control bath (no drug additions). Neuromuscular function was assessed using force transducer recordings and electrical field stimulation (EFS; 20 V, 10 Hz, 0.5 ms, 10 s) prior to and following loperamide and naloxone. KEY RESULTS In human preparations with L-NOARG/MRS2179, loperamide had no significant effects on isometric contractions. In preparations with hyoscine hydrobromide, loperamide reduced isometric relaxation during EFS (median difference + 0.60 g post-loperamide, Z = -2.35, p = 0.019). CONCLUSIONS AND INFERENCES Loperamide had no effect on excitatory neuromuscular function in human colonic circular muscle. These findings suggest that loperamide alters colonic function by acting primarily on inhibitory motor neurons, premotor enteric neurons, or via alternative non-opioid receptor pathways.
Collapse
Affiliation(s)
- Paul T Heitmann
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.,Departments of Surgery and Gastroenterology, Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Lauren Keightley
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Lukasz Wiklendt
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - David A Wattchow
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.,Departments of Surgery and Gastroenterology, Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Simon S J Brookes
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Nicholas J Spencer
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Marcello Costa
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Phil G Dinning
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.,Departments of Surgery and Gastroenterology, Flinders Medical Centre, Adelaide, South Australia, Australia
| |
Collapse
|
4
|
Pantouli F, Grim TW, Schmid CL, Acevedo-Canabal A, Kennedy NM, Cameron MD, Bannister TD, Bohn LM. Comparison of morphine, oxycodone and the biased MOR agonist SR-17018 for tolerance and efficacy in mouse models of pain. Neuropharmacology 2021; 185:108439. [PMID: 33345829 PMCID: PMC7887086 DOI: 10.1016/j.neuropharm.2020.108439] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022]
Abstract
The mu opioid receptor-selective agonist, SR-17018, preferentially activates GTPγS binding over βarrestin2 recruitment in cellular assays, thereby demonstrating signaling bias. In mice, SR-17018 stimulates GTPγS binding in brainstem and produces antinociception with potencies similar to morphine. However, it produces much less respiratory suppression and mice do not develop antinociceptive tolerance in the hot plate assay upon repeated dosing. Herein we evaluate the effects of acute and repeated dosing of SR-17018, oxycodone and morphine in additional models of pain-related behaviors. In the mouse warm water tail immersion assay, an assessment of spinal reflex to thermal nociception, repeated administration of SR-17018 produces tolerance as does morphine and oxycodone. SR-17018 retains efficacy in a formalin-induced inflammatory pain model upon repeated dosing, while oxycodone does not. In a chemotherapeutic-induced neuropathy pain model SR-17018 is more potent and efficacious than morphine or oxycodone, moreover, this efficacy is retained upon repeated dosing of SR-17018. These findings demonstrate that, with the exception of the tail flick test, SR-17018 retains efficacy upon chronic treatment across several pain models.
Collapse
Affiliation(s)
- Fani Pantouli
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Travis W Grim
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Cullen L Schmid
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Agnes Acevedo-Canabal
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Nicole M Kennedy
- Departments of Molecular Medicine and Chemistry, The Scripps Research Institute, Jupiter, FL, USA
| | - Michael D Cameron
- Departments of Molecular Medicine and Chemistry, The Scripps Research Institute, Jupiter, FL, USA
| | - Thomas D Bannister
- Departments of Molecular Medicine and Chemistry, The Scripps Research Institute, Jupiter, FL, USA
| | - Laura M Bohn
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, FL, USA.
| |
Collapse
|
5
|
Abstract
The management of pain, particularly chronic pain, is still an area of medical need. In this context, opioids remain a gold standard for the treatment of pain. However, significant side effects, mainly of central origin, limit their clinical use. Here, we review recent progress to improve the therapeutic and safety profiles of opioids for pain management. Characterization of peripheral opioid-mediated pain mechanisms have been a key component of this process. Several studies identified peripheral µ, δ, and κ opioid receptors (MOR, DOR, and KOR, respectively) and nociceptin/orphanin FQ (NOP) receptors as significant players of opioid-mediated antinociception, able to achieve clinically significant effects independently of any central action. Following this, particularly from a medicinal chemistry point of view, main efforts have been directed towards the peripheralization of opioid receptor agonists with the objective of optimizing receptor activity and minimizing central exposure and the associated undesired effects. These activities have allowed the characterization of a great variety of compounds and investigational drugs that show low central nervous system (CNS) penetration (and therefore a reduced side effect profile) yet maintaining the desired opioid-related peripheral antinociceptive activity. These include highly hydrophilic/amphiphilic and massive molecules unable to easily cross lipid membranes, substrates of glycoprotein P (a extrusion pump that avoids CNS penetration), nanocarriers that release the analgesic agent at the site of inflammation and pain, and pH-sensitive opioid agonists that selectively activate at those sites (and represent a new pharmacodynamic paradigm). Hopefully, patients with pain will benefit soon from the incorporation of these new entities.
Collapse
|
6
|
Speltz R, Lunzer MM, Shueb SS, Akgün E, Reed R, Kalyuzhny A, Portoghese PS, Simone DA. The bivalent ligand, MMG22, reduces neuropathic pain after nerve injury without the side effects of traditional opioids. Pain 2020; 161:2041-2057. [PMID: 32345918 PMCID: PMC7606301 DOI: 10.1097/j.pain.0000000000001902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/21/2020] [Indexed: 11/26/2022]
Abstract
ABSTRACT Functional interactions between the mu opioid receptor (MOR) and the metabotropic glutamate receptor 5 (mGluR5) in pain and analgesia have been well established. MMG22 is a bivalent ligand containing MOR agonist (oxymorphamine) and mGluR5 antagonist (MPEP) pharmacophores tethered by a 22-atom linker. MMG22 has been shown to produce potent analgesia in several models of chronic inflammatory and neuropathic pain (NP). This study assessed the efficacy of systemic administration of MMG22 at reducing pain behavior in the spared nerve injury (SNI) model of NP in mice, as well as its side-effect profile and abuse potential. MMG22 reduced mechanical hyperalgesia and spontaneous ongoing pain after SNI, with greater potency early (10 days) as compared to late (30 days) after injury. Systemic administration of MMG22 did not induce place preference in naive animals, suggesting absence of abuse liability when compared to traditional opioids. MMG22 also lacked the central locomotor, respiratory, and anxiolytic side effects of its monomeric pharmacophores. Evaluation of mRNA expression showed the transcripts for both receptors were colocalized in cells in the dorsal horn of the lumbar spinal cord and dorsal root ganglia. Thus, MMG22 reduces hyperalgesia after injury in the SNI model of NP without the typical centrally mediated side effects associated with traditional opioids.
Collapse
Affiliation(s)
- Rebecca Speltz
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Mary M Lunzer
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Sarah S Shueb
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| | - Eyup Akgün
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | | | - Alex Kalyuzhny
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
- Bio-Techne, Minneapolis, MN, United States
| | - Philip S Portoghese
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Donald A Simone
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
7
|
On the Role of Peripheral Sensory and Gut Mu Opioid Receptors: Peripheral Analgesia and Tolerance. Molecules 2020; 25:molecules25112473. [PMID: 32466522 PMCID: PMC7321260 DOI: 10.3390/molecules25112473] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/22/2020] [Accepted: 05/24/2020] [Indexed: 02/06/2023] Open
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, β-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.
Collapse
|
8
|
Inhibition of Fast Nerve Conduction Produced by Analgesics and Analgesic Adjuvants-Possible Involvement in Pain Alleviation. Pharmaceuticals (Basel) 2020; 13:ph13040062. [PMID: 32260535 PMCID: PMC7243109 DOI: 10.3390/ph13040062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/29/2020] [Accepted: 04/01/2020] [Indexed: 02/07/2023] Open
Abstract
Nociceptive information is transmitted from the periphery to the cerebral cortex mainly by action potential (AP) conduction in nerve fibers and chemical transmission at synapses. Although this nociceptive transmission is largely inhibited at synapses by analgesics and their adjuvants, it is possible that the antinociceptive drugs inhibit nerve AP conduction, contributing to their antinociceptive effects. Many of the drugs are reported to inhibit the nerve conduction of AP and voltage-gated Na+ and K+ channels involved in its production. Compound action potential (CAP) is a useful measure to know whether drugs act on nerve AP conduction. Clinically-used analgesics and analgesic adjuvants (opioids, non-steroidal anti-inflammatory drugs, 2-adrenoceptor agonists, antiepileptics, antidepressants and local anesthetics) were found to inhibit fast-conducting CAPs recorded from the frog sciatic nerve by using the air-gap method. Similar actions were produced by antinociceptive plant-derived chemicals. Their inhibitory actions depended on the concentrations and chemical structures of the drugs. This review article will mention the inhibitory actions of the antinociceptive compounds on CAPs in frog and mammalian peripheral (particularly, sciatic) nerves and on voltage-gated Na+ and K+ channels involved in AP production. Nerve AP conduction inhibition produced by analgesics and analgesic adjuvants is suggested to contribute to at least a part of their antinociceptive effects.
Collapse
|
9
|
pK a of opioid ligands as a discriminating factor for side effects. Sci Rep 2019; 9:19344. [PMID: 31852967 PMCID: PMC6920366 DOI: 10.1038/s41598-019-55886-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 12/03/2019] [Indexed: 01/18/2023] Open
Abstract
The non-selective activation of central and peripheral opioid receptors is a major shortcoming of currently available opioids. Targeting peripheral opioid receptors is a promising strategy to preclude side effects. Recently, we showed that fentanyl-derived μ-opioid receptor (MOR) agonists with reduced acid dissociation constants (pKa) due to introducing single fluorine atoms produced injury-restricted antinociception in rat models of inflammatory, postoperative and neuropathic pain. Here, we report that a new double-fluorinated compound (FF6) and fentanyl show similar pKa, MOR affinity and [35S]-GTPγS binding at low and physiological pH values. In vivo, FF6 produced antinociception in injured and non-injured tissue, and induced sedation and constipation. The comparison of several fentanyl derivatives revealed a correlation between pKa values and pH-dependent MOR activation, antinociception and side effects. An opioid ligand’s pKa value may be used as discriminating factor to design safer analgesics.
Collapse
|
10
|
Vera G, Girón R, Martín-Fontelles MI, Abalo R. Radiographic dose-dependency study of loperamide effects on gastrointestinal motor function in the rat. Temporal relationship with nausea-like behavior. Neurogastroenterol Motil 2019; 31:e13621. [PMID: 31117152 DOI: 10.1111/nmo.13621] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/09/2019] [Accepted: 04/24/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Loperamide is a potent mu opioid receptor agonist available over the counter to treat diarrhea. Although at therapeutic doses loperamide is devoid of central effects, it may exert them if used at high doses or combined with drugs that increase its systemic and/or central bioavailability. Recently, public health and scientific interest on loperamide has increased due to a growing trend of misuse and abuse, and consequent reports on its toxicity. Our aim was to evaluate in the rat the effects of increasing loperamide doses, with increasing likelihood to induce central effects, on gastrointestinal motor function (including gastric dysmotility and nausea-like behavior). METHODS Male Wistar rats received an intraperitoneal injection of vehicle or loperamide (0.1, 1, or 10 mg kg-1 ). Three sets of experiments were performed to evaluate: (a) central effects (somatic nociceptive thresholds, immobility time, core temperature, spontaneous locomotor activity); (b) general gastrointestinal motility (serial X-rays were taken 0-8 hours after intragastric barium administration and analyzed semiquantitatively, morphometrically, and densitometrically); and (c) bedding intake (a rodent indirect marker of nausea). Animals from sets 1 and 3 were used to evaluate gastric dysmotility ex vivo at 2 and 4 hours after administration, respectively. KEY RESULTS Loperamide significantly induced antinociception, hypothermia, and hypolocomotion (but not catalepsy) at high doses and dose-dependently reduced gastrointestinal motor function, with the intestine exhibiting higher sensitivity than the stomach. Whereas bedding intake occurred early and transiently, gastric dysmotility was much more persistent. CONCLUSIONS AND INFERENCES Our results suggest that loperamide-induced nausea and gastric dysmotility might be temporally dissociated.
Collapse
Affiliation(s)
- Gema Vera
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain.,Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL), Alcorcón, Spain.,Unidad Asociada I+D+i al Instituto de Química Médica (IQM), Centro Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Rocío Girón
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain.,Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL), Alcorcón, Spain.,Unidad Asociada I+D+i al Instituto de Química Médica (IQM), Centro Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - María Isabel Martín-Fontelles
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain.,Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL), Alcorcón, Spain.,Unidad Asociada I+D+i al Instituto de Química Médica (IQM), Centro Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Raquel Abalo
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain.,Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL), Alcorcón, Spain.,Unidad Asociada I+D+i al Instituto de Química Médica (IQM), Centro Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
11
|
Fakhraei N, Javadian N, Rahimian R, Nili F, Rahimi N, Hashemizadeh S, Dehpour AR. Involvement of central opioid receptors in protective effects of methadone on experimental colitis in rats. Inflammopharmacology 2018; 26:1399-1413. [PMID: 30318564 DOI: 10.1007/s10787-018-0538-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 10/01/2018] [Indexed: 01/16/2023]
Abstract
PURPOSE There are several lines of evidence on the protective roles of opioids in gastrointestinal inflammatory conditions. This study aims to distinguish the central and peripheral roles of methadone, a non-selective opioid receptor agonist, in an acute model of ulcerative colitis in male rats. METHODS Ulcerative colitis was induced by intrarectal administration of acetic acid 4%. Methadone was injected subcutaneously (s.c.), 5 and 10 mg/kg, and intracerebroventricular (i.c.v.), 50 and 300 ng/rat. Opioid antagonists were employed. Methylnaltrexone (MNTX; 5 mg/kg, i.p.), a peripherally acting opioid receptor antagonist, and naltrexone (NTX; 5 mg/kg, i.p. and 10 ng/rat, i.c.v.), a peripherally and centrally acting opioid receptor antagonist were injected before methadone (10 mg/kg, s.c. and or 300 ng/rat, i.c.v.) administration. NTX (5 mg/kg, i.p. and 10 ng/rat, i.c.v.) were administered 30 min prior to administration of methadone (10 mg/kg, s.c. and 300 ng/rat, i.c.v.), respectively. MNTX (5 mg/kg, i.p.) was injected 30 min prior to methadone (10 mg/kg, s.c.). Seventy-two hours following colitis induction, macroscopic and microscopic mucosal lesions, and the colonic levels of tumor necrosis factor-alpha (TNF-α) and interleukin-1β (IL-1β) were determined. RESULTS Methadone (300 ng/rat, i.c.v.) and Methadone (5 and 10 mg/kg, s.c.) improved the macroscopic and microscopic scores through opioid receptors. Also, a significant reduction in TNF-α and IL-1β was observed. Peripherally and centrally injected NTX significantly reversed methadone 10 mg/kg s.c. anti-inflammatory effects while MNTX could not completely reverse this effect. Moreover, centrally administered methadone (300 ng/rat) showed the anti-inflammatory effect which was reversed by central administration of NTX (10 ng/rat). CONCLUSIONS The opioid receptors mainly the central opioid receptors may mediate the protective actions of methadone on the experimental model of inflammatory bowel disease in rat.
Collapse
MESH Headings
- Acetic Acid
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/therapeutic use
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Colitis, Ulcerative/chemically induced
- Colitis, Ulcerative/mortality
- Colitis, Ulcerative/prevention & control
- Injections, Intraventricular
- Injections, Subcutaneous
- Interleukin-1beta/biosynthesis
- Intestinal Mucosa/pathology
- Male
- Methadone/administration & dosage
- Methadone/therapeutic use
- Naltrexone/pharmacology
- Narcotic Antagonists/pharmacology
- Rats
- Rats, Wistar
- Receptors, Opioid/drug effects
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Nahid Fakhraei
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Nina Javadian
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Rahimian
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Laval University, Quebec, G1J 2G3, Canada
| | - Fatemeh Nili
- Department of Pathology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Nastaran Rahimi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shiva Hashemizadeh
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
The Peripheral Versus Central Antinociception of a Novel Opioid Agonist: Acute Inflammatory Pain in Rats. Neurochem Res 2018; 43:1250-1257. [DOI: 10.1007/s11064-018-2542-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/26/2018] [Accepted: 04/29/2018] [Indexed: 10/17/2022]
|
13
|
Kerui G, Jasmin L. Dual effects of brain sparing opioid in newborn rats: Analgesia and hyperalgesia. NEUROBIOLOGY OF PAIN 2018; 3:1-7. [PMID: 31194154 PMCID: PMC6550121 DOI: 10.1016/j.ynpai.2018.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 01/16/2023]
Abstract
The peripherally acting opioid loperamide produces sustained antinociception in the newborn rat. Loperamide minimally crosses the blood brain barrier in the newborn rat. Daily systemic administration of loperamide produces opioid induced hyperalgesia in the newborn rat.
Effective pain management in neonates without the unwanted central nervous system (CNS) side effects remains an unmet need. To circumvent these central effects we tested the peripherally acting (brain sparing) opioid agonist loperamide in neonate rats. Our results show that: 1) loperamide (1 mg/kg, s.c.) does not affect the thermal withdrawal latency in the normal hind paw while producing antinociception in all pups with an inflamed hind paw. 2) A dose of loperamide 5 times higher resulted in only 6.9 ng/mL of loperamide in the cerebrospinal fluid (CSF), confirming that loperamide minimally crosses the blood–brain barrier (BBB). 3) Unexpectedly, sustained administration of loperamide for 5 days resulted in a hyperalgesic behavior, as well as increased excitability (sensitization) of dorsal root ganglia (DRGs) and spinal nociceptive neurons. This indicates that opioid induced hyperalgesia (OIH) can be induced through the peripheral nervous system. Unless prevented, OIH could in itself be a limiting factor in the use of brain sparing opioids in the neonate.
Collapse
Affiliation(s)
- Gong Kerui
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Luc Jasmin
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
14
|
Wu Y, Zou B, Liang L, Li M, Tao YX, Yu H, Wang X, Li M. Loperamide inhibits sodium channels to alleviate inflammatory hyperalgesia. Neuropharmacology 2017; 117:282-291. [PMID: 28216001 DOI: 10.1016/j.neuropharm.2017.02.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 02/10/2017] [Accepted: 02/12/2017] [Indexed: 12/25/2022]
Abstract
Previous studies demonstrated that Loperamide, originally known as an anti-diarrheal drug, is a promising analgesic agent primarily targeting mu-opioid receptors. However some evidences suggested that non-opioid mechanisms may be contributing to its analgesic effect. In the present study, Loperamide was identified as a Nav1.7 blocker in a pilot screen. In HEK293 cells expressing Nav1.7 sodium channels, Loperamide blocked the resting state of Nav1.7 channels (IC50 = 1.86 ± 0.11 μM) dose-dependently and reversibly. Loperamide produced a 10.4 mV of hyperpolarizing shift for the steady-state inactivation of Nav1.7 channels without apparent effect on the voltage-dependent activation. The drug displayed a mild use- and state-dependent inhibition on Nav1.7 channels, which was removed by the local anesthetic-insensitive construct Nav1.7-F1737A. Inhibition of Nav1.7 at resting state was not altered significantly by the F1737A mutation. Compared to its effects on Nav1.7, Loperamide exhibited higher potency on recombinant Nav1.8 channels in ND7/23 cells (IC50 = 0.60 ± 0.10 μM) and weaker potency on Nav1.9 channels (3.48 ± 0.33 μM). Notably more pronounced inhibition was observed in the native Nav1.8 channels (0.11 ± 0.08 μM) in DRG neurons. Once mu-opioid receptor was antagonized by Naloxone in DRG neurons, potency of Loperamide on Nav1.8 was identical to that of recombinant Nav1.8 channels. The inhibition on Nav channels may be the main mechanism of Loperamide for pain relief beyond mu-opioid receptor. In the meanwhile, the opioid receptor pathway may also influence the blocking effect of Loperamide on sodium channels, implying a cross-talk between sodium channels and opioid receptors in pain processing.
Collapse
Affiliation(s)
- Ying Wu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Beiyan Zou
- The Solomon H. Snyder Department of Neuroscience, High Throughput Biology Center and Johns Hopkins Ion Channel Center, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Lingli Liang
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Min Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Haibo Yu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xiaoliang Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Min Li
- The Solomon H. Snyder Department of Neuroscience, High Throughput Biology Center and Johns Hopkins Ion Channel Center, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
15
|
Ronsisvalle S, Aricò G, Panarello F, Spadaro A, Pasquinucci L, Pappalardo MS, Parenti C, Ronsisvalle N. An LP1 analogue, selective MOR agonist with a peculiar pharmacological profile, used to scrutiny the ligand binding domain. Bioorg Med Chem 2016; 24:5280-5290. [DOI: 10.1016/j.bmc.2016.08.057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/26/2016] [Accepted: 08/27/2016] [Indexed: 12/18/2022]
|
16
|
Oberdick J, Ling Y, Phelps MA, Yudovich MS, Schilling K, Sadee W. Preferential Delivery of an Opioid Antagonist to the Fetal Brain in Pregnant Mice. J Pharmacol Exp Ther 2016; 358:22-30. [PMID: 27189967 PMCID: PMC4931874 DOI: 10.1124/jpet.115.231902] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 04/27/2016] [Indexed: 02/01/2023] Open
Abstract
Prolonged fetal exposure to opioids results in neonatal abstinence syndrome (NAS), a major medical problem requiring intensive care and increased hospitalization times for newborns with NAS. Multiple strategies are currently available to alleviate withdrawal in infants with NAS. To prevent NAS caused by opioid maintenance programs in pregnant women, blocking fetal dependence without compromising the mother's opiate therapy is desirable. Here we tested in pregnant mice whether a peripherally selective opioid antagonist can preferentially enter the fetal brain and, thereby, in principle, selectively protect the fetus. We show using mass spectrometry that 6β-naltrexol, a neutral opioid antagonist with very limited ability to cross the blood-brain barrier (BBB), readily crosses the placental barrier and enters the fetal brain at high levels, although it is relatively excluded from the maternal brain. Furthermore, owing to the late development of the BBB in postnatal mice, we show that 6β-naltrexol can readily enter the juvenile mouse brain until at least postnatal day 14. Taking advantage of this observation, we show that long-term exposure to morphine starting in the second postnatal week causes robust and quantifiable dependence behaviors that are suppressed by concomitant administration of 6β-naltrexol with much greater potency (ID50 0.022-0.044 mg/kg, or 1/500 the applied dose of morphine) than previously demonstrated for either the suppression of central nervous system opioid effects or the induction of withdrawal in adults. These results indicate that peripherally selective opioid antagonists capable of penetrating the placenta may be beneficial for preventing or reducing neonatal dependence and NAS in a dose range that should not interfere with maternal opioid maintenance.
Collapse
Affiliation(s)
- John Oberdick
- Department of Neuroscience (J.O., M.S.Y.), and College of Medicine Center for Pharmacogenomics (W.S.), Wexner Medical Center, and College of Pharmacy, Division of Pharmaceutics and Pharmaceutical Chemistry (Y.L., M.A.P.), The Ohio State University, Columbus, Ohio; and Anatomisches Institut, Anatomie und Zellbiologie (K.S.), Universität Bonn, Bonn, Germany
| | - Yonghua Ling
- Department of Neuroscience (J.O., M.S.Y.), and College of Medicine Center for Pharmacogenomics (W.S.), Wexner Medical Center, and College of Pharmacy, Division of Pharmaceutics and Pharmaceutical Chemistry (Y.L., M.A.P.), The Ohio State University, Columbus, Ohio; and Anatomisches Institut, Anatomie und Zellbiologie (K.S.), Universität Bonn, Bonn, Germany
| | - Mitch A Phelps
- Department of Neuroscience (J.O., M.S.Y.), and College of Medicine Center for Pharmacogenomics (W.S.), Wexner Medical Center, and College of Pharmacy, Division of Pharmaceutics and Pharmaceutical Chemistry (Y.L., M.A.P.), The Ohio State University, Columbus, Ohio; and Anatomisches Institut, Anatomie und Zellbiologie (K.S.), Universität Bonn, Bonn, Germany
| | - Max S Yudovich
- Department of Neuroscience (J.O., M.S.Y.), and College of Medicine Center for Pharmacogenomics (W.S.), Wexner Medical Center, and College of Pharmacy, Division of Pharmaceutics and Pharmaceutical Chemistry (Y.L., M.A.P.), The Ohio State University, Columbus, Ohio; and Anatomisches Institut, Anatomie und Zellbiologie (K.S.), Universität Bonn, Bonn, Germany
| | - Karl Schilling
- Department of Neuroscience (J.O., M.S.Y.), and College of Medicine Center for Pharmacogenomics (W.S.), Wexner Medical Center, and College of Pharmacy, Division of Pharmaceutics and Pharmaceutical Chemistry (Y.L., M.A.P.), The Ohio State University, Columbus, Ohio; and Anatomisches Institut, Anatomie und Zellbiologie (K.S.), Universität Bonn, Bonn, Germany
| | - Wolfgang Sadee
- Department of Neuroscience (J.O., M.S.Y.), and College of Medicine Center for Pharmacogenomics (W.S.), Wexner Medical Center, and College of Pharmacy, Division of Pharmaceutics and Pharmaceutical Chemistry (Y.L., M.A.P.), The Ohio State University, Columbus, Ohio; and Anatomisches Institut, Anatomie und Zellbiologie (K.S.), Universität Bonn, Bonn, Germany
| |
Collapse
|
17
|
Kulyk VB, Chizhmakov IV, Volkova TM, Maximyuk OP, Krishtal OA. [ROLE PHOSPHOINOSITID SIGNALING PATHWAY IN OPIOIDS CONTROL OF P2X3 RECEPTORS IN THE PRIMARY SENSORY NEURONS]. ACTA ACUST UNITED AC 2015; 61:22-9. [PMID: 26552301 DOI: 10.15407/fz61.04.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Homomeric P2X3 receptors expressed in primary nociceptive neurons are crucial elements in the pain signal generation. In turn, opioid system regulates the intensity of this signal in both CNS and PNS. Here we describe the effects of opioids on P2X3 receptors in DRG neurons studied by using patch clamp technique. Activation of G-protein coupled opioid receptors by endogenous opioid Leu-enkephalin (Leu), resulted in the two opposite effects on P2X3 receptor-mediated currents (P2X3 currents). In particular, application of 1 µM Leu lead to the complete inhibition of P2X3 currents. However, after pretreatment of the neurons with a Gi/o-protein inhibitor pertussis toxin (PT), the same concentration of Leu caused facilitation of P2X3 currents. PLC inhibitor U-73122 at concentration of 1 µM completely eliminated both facilitating and inhibitory effects of Leu on P2X3 currents. Thus, opioid receptor agonists cause two oppositely directed effects on P2X3 receptors in DRG neurons of rats and both of them are mediated through PLC signaling pathway. Our results point to a possible molecular basis of the mechanism for the well-known transition inhibitory action of opioids (analgesia) to facilitating (hyperalgesia).
Collapse
|
18
|
Deuis JR, Whately E, Brust A, Inserra MC, Asvadi NH, Lewis RJ, Alewood PF, Cabot PJ, Vetter I. Activation of κ Opioid Receptors in Cutaneous Nerve Endings by Conorphin-1, a Novel Subtype-Selective Conopeptide, Does Not Mediate Peripheral Analgesia. ACS Chem Neurosci 2015. [PMID: 26225903 DOI: 10.1021/acschemneuro.5b00113] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Selective activation of peripheral κ opioid receptors (KORs) may overcome the dose-limiting adverse effects of conventional opioid analgesics. We recently developed a vicinal disulfide-stabilized class of peptides with subnanomolar potency at the KOR. The aim of this study was to assess the analgesic effects of one of these peptides, named conorphin-1, in comparison with the prototypical KOR-selective small molecule agonist U-50488, in several rodent pain models. Surprisingly, neither conorphin-1 nor U-50488 were analgesic when delivered peripherally by intraplantar injection at local concentrations expected to fully activate the KOR at cutaneous nerve endings. While U-50488 was analgesic when delivered at high local concentrations, this effect could not be reversed by coadministration with the selective KOR antagonist ML190 or the nonselective opioid antagonist naloxone. Instead, U-50488 likely mediated its peripheral analgesic effect through nonselective inhibition of voltage-gated sodium channels, including peripheral sensory neuron isoforms NaV1.8 and NaV1.7. Our study suggests that targeting the KOR in peripheral sensory nerve endings innervating the skin is not an alternative analgesic approach.
Collapse
Affiliation(s)
- Jennifer R. Deuis
- School
of Pharmacy, University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Ella Whately
- School
of Pharmacy, University of Queensland, Woolloongabba, QLD 4102, Australia
| | | | - Marco C. Inserra
- School
of Pharmacy, University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Naghmeh H. Asvadi
- School
of Pharmacy, University of Queensland, Woolloongabba, QLD 4102, Australia
| | | | | | - Peter J. Cabot
- School
of Pharmacy, University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Irina Vetter
- School
of Pharmacy, University of Queensland, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
19
|
Abstract
Chronic diarrhea is usually associated with a number of non-infectious causes. When definitive treatment is unavailable, symptomatic drug therapy is indicated. Pharmacologic agents for chronic diarrhea include loperamide, 5-hydroxytryptamine type 3 (5-HT3) receptor antagonists, diosmectite, cholestyramine, probiotics, antispasmodics, rifaximin, and anti-inflammatory agents. Loperamide, a synthetic opiate agonist, decreases peristaltic activity and inhibits secretion, resulting in the reduction of fluid and electrolyte loss and an increase in stool consistency. Cholestyramine is a bile acid sequestrant that is generally considered as the first-line treatment for bile acid diarrhea. 5-HT3 receptor antagonists have significant benefits in patients with irritable bowel syndrome (IBS) with diarrhea. Ramosetron improves stool consistency as well as global IBS symptoms. Probiotics may have a role in the prevention of antibiotic-associated diarrhea. However, data on the role of probiotics in the treatment of chronic diarrhea are lacking. Diosmectite, an absorbent, can be used for the treatment of chronic functional diarrhea, radiation-induced diarrhea, and chemotherapy-induced diarrhea. Antispasmodics including alverine citrate, mebeverine, otilonium bromide, and pinaverium bromide are used for relieving diarrheal symptoms and abdominal pain. Rifaximin can be effective for chronic diarrhea associated with IBS and small intestinal bacterial overgrowth. Budesonide is effective in both lymphocytic colitis and collagenous colitis. The efficacy of mesalazine in microscopic colitis is weak or remains uncertain. Considering their mechanisms of action, these agents should be prescribed properly.
Collapse
Affiliation(s)
- Kwang Jae Lee
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
20
|
Kulyk VB, Chyzhmakov IV, Volkova TM, Kryshtal OA. Ambivalent Effects on P2X3 Receptors in Rat Sensory Neurons in the Presence of Opioid Receptor Antagonists. NEUROPHYSIOLOGY+ 2015. [DOI: 10.1007/s11062-015-9519-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
21
|
Saliminia A, Azimaraghi O, Babayipour S, Ardavan K, Movafegh A. Efficacy of transverse abdominis plane block in reduction of postoperation pain in laparoscopic cholecystectomy. ACTA ACUST UNITED AC 2015; 53:119-22. [PMID: 26282913 DOI: 10.1016/j.aat.2015.07.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 06/19/2015] [Accepted: 07/03/2015] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Transversus abdominis plane (TAP) block is a recently introduced regional anesthesia technique that is used for postoperative pain reduction in some abdominal surgeries. The present study evaluated the efficacy of the TAP block on the post laparoscopic cholecystectomy pain intensity and analgesic consumption. METHODS Fifty-four patients were enrolled in three groups: TAP block with normal saline (Group 1, n = 18); TAP block with bupivacaine (Group 2, n = 18); and TAP block with bupivacaine plus sufentanil (Group 3, n = 18). The time to the first fentanyl request, fentanyl consumption in the 24 hours following surgery, and postoperative pain intensity at 30 minutes, 1 hour, 6 hours, 12 hours, and 24 hours following discharge for recovery were measured and recorded. RESULTS The total amount of 24-hour fentanyl consumption was higher in Group 1 (877.8 ± 338.8 μg) than either Group 2 (566.7 ± 367.8 μg) or Group 3 (555.5 ± 356.8 μg; p = 0.03). Postoperative pain score was higher in Group 1 than intervention groups (p = 0.006); however, there was no significant difference in intervention groups. The time to the first fentanyl request in Group 1 (79.44 ± 42.2) was significantly lower than Group 3 (206.38 ± 112.7; p = 0.001). CONCLUSION The present study demonstrated that bilateral TAP block with 0.5% bupivacaine reduces post laparoscopic cholecystectomy pain intensity and fentanyl request and prolongs time to the first analgesic request. Adding sufentanil to the block solution reduced neither pain intensity nor fentanyl further consumption.
Collapse
Affiliation(s)
- Alireza Saliminia
- Anesthesiology and Critical Care, Dr. Ali Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Omid Azimaraghi
- Anesthesiology and Critical Care, Dr. Ali Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Shiva Babayipour
- Research Development Center, Dr. Ali Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Kamelia Ardavan
- Department of Cardiology, Research Committee, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ali Movafegh
- Anesthesiology and Critical Care, Dr. Ali Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Azimaraghi O, Marashi SM, Khazaei N, Pourhassan S, Movafegh A. The Effect of Adding Sufentanil to 0.5% Hyperbaric Bupivacaine on Duration of Brachial Plexus Blockade in Chronic Opium Abusers: a Randomized Clinical Trial. Anesth Pain Med 2015; 5:e21960. [PMID: 26161314 PMCID: PMC4493736 DOI: 10.5812/aapm.21960v2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 10/04/2014] [Accepted: 11/11/2014] [Indexed: 01/19/2023] Open
Abstract
Background: Anesthesia induction in patients with current substance abuse can be a challenge for anesthesiologists. Objectives: This study aimed to evaluate the effect of adding Sufentanil to Bupivacaine on duration of brachial plexus nerve block. Patients and Methods: One hundred and twenty patients with (Groups C and D) and without (Groups A and B) a history of opium abuse (60 in each group) scheduled for elective upper extremity procedures were randomly assigned to either receive 30 mL bupivacaine alone (Groups A and C) or in combination with additional 10 µg sufentanil (Groups B and D). An ultrasound-guided technique was applied to perform upper extremity brachial plexus blockade. The onset and duration of sensory and motor blocks were recorded and compared between the four groups. Results: The duration of sensory and motor block were significantly less in Group C (537.0 ± 40.1 minutes, 479.0 ± 34.8 minutes) and the longest duration of sensory and motor block was observed in group B (705.0 ± 43.8 minutes, 640.0 ± 32.5 minutes). The duration of sensory and motor block in Group B (705.0 ± 43.8 minutes, 640.0 ± 32.5 minutes) was longer and statistically higher than group A (619.5 ± 48.0 minutes, 573.2 ± 31.5 minutes), the same trend was observed in group D (598.6 ± 53.2 minutes, 569.3 ± 39.9 minutes) over group C (537.0 ± 40.1 minutes, 479.0 ± 34.8 minutes) (P < 0.001, one-way ANOVA). Conclusions: The length of sensory and motor blockade is shorter in chronic opioid abusers. Adding 10 µg sufentanil to hyperbaric bupivacaine in opium abusers lengthened the sensory and motor block duration.
Collapse
Affiliation(s)
- Omid Azimaraghi
- Department of Anesthesiology, Pain and Critical Care, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mojtaba Marashi
- Department of Anesthesiology, Pain and Critical Care, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Noushin Khazaei
- Department of Anesthesiology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sarah Pourhassan
- Department of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Movafegh
- Department of Anesthesiology, Pain and Critical Care, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Corresponding author: Ali Movafegh, Department of Anesthesiology, Pain and Critical Care, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran. Tel: +98-9123021389, Fax: +98-88220032, E-mail:
| |
Collapse
|
23
|
Eans SO, Ganno ML, Mizrachi E, Houghten RA, Dooley CT, McLaughlin JP, Nefzi A. Parallel Synthesis of Hexahydrodiimidazodiazepines Heterocyclic Peptidomimetics and Their in Vitro and in Vivo Activities at μ (MOR), δ (DOR), and κ (KOR) Opioid Receptors. J Med Chem 2015; 58:4905-17. [PMID: 25996309 DOI: 10.1021/jm501637c] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In the development of analgesics with mixed-opioid agonist activity, peripherally selective activity is expected to decrease side effects, minimizing respiratory depression and reinforcing properties generating significantly safer analgesic therapeutics. We synthesized diazaheterocyclics from reduced tripeptides. In vitro screening with radioligand competition binding assays demonstrated variable affinity for μ (MOR), δ (DOR), and κ (KOR) opioid receptors across the series, with the diimidazodiazepine 14 (2065-14) displaying good affinity for DOR and KOR. Central (icv), intraperitoneal (ip), or oral (po) administration of 14 produced dose-dependent, opioid-receptor mediated antinociception in the mouse, as determined from a 55 °C warm-water tail-withdrawal assay. Only trace amounts of compound 14 was found in brain up to 90 min later, suggesting poor BBB penetration and possible peripherally restricted activity. Central administration of 14 did not produce locomotor effects, acute antinociceptive tolerance, or conditioned-place preference or aversion. The data suggest these diazaheterocyclic mixed activity opioid receptor agonists may hold potential as new analgesics with fewer liabilities of use.
Collapse
Affiliation(s)
- Shainnel O Eans
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Michelle L Ganno
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Elisa Mizrachi
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Richard A Houghten
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Colette T Dooley
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Jay P McLaughlin
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Adel Nefzi
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| |
Collapse
|
24
|
de Oliveira AM, de Araújo AF, Lyra Lemos RP, Conserva LM, de Souza Ferro JN, Barreto E. Antinociceptive and anti-inflammatory activity of the siaresinolic acid, a triterpene isolated from the leaves of Sabicea grisea Cham. & Schltdl. var. grisea. J Nat Med 2015; 69:232-40. [DOI: 10.1007/s11418-014-0883-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 12/25/2014] [Indexed: 10/24/2022]
|
25
|
Chizhmakov I, Kulyk V, Khasabova I, Khasabov S, Simone D, Bakalkin G, Gordienko D, Verkhratsky A, Krishtal O. Molecular mechanism for opioid dichotomy: bidirectional effect of μ-opioid receptors on P2X₃ receptor currents in rat sensory neurones. Purinergic Signal 2015; 11:171-81. [PMID: 25592684 DOI: 10.1007/s11302-015-9443-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/05/2015] [Indexed: 12/28/2022] Open
Abstract
Here, we describe a molecular switch associated with opioid receptors-linked signalling cascades that provides a dual opioid control over P2X3 purinoceptor in sensory neurones. Leu-enkephalin inhibited P2X3-mediated currents with IC50 ~10 nM in ~25% of small nociceptive rat dorsal root ganglion (DRG) neurones. In contrast, in neurones pretreated with pertussis toxin leu-enkephalin produced stable and significant increase of P2X3 currents. All effects of opioid were abolished by selective μ-opioid receptor antagonist D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP), nonselective inhibitor naloxone, and by PLC inhibitor U73122. Thus, we discovered a dual link between purinoceptors and μ-opioid receptors: the latter exert both inhibitory (pertussis toxin-sensitive) and stimulatory (pertussis toxin-insensitive) actions on P2X3 receptors through phospholipase C (PLC)-dependent pathways. This dual opioid control of P2X3 receptors may provide a molecular explanation for dichotomy of opioid therapy. Pharmacological control of this newly identified facilitation/inhibition switch may open new perspectives for the adequate medical use of opioids, the most powerful pain-killing agents known today.
Collapse
Affiliation(s)
- Igor Chizhmakov
- State Key Laboratory for Molecular Biology, Bogomoletz Institute of Physiology, 4 Bogomoletz str., Kiev, 01024, Ukraine
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Sukhtankar DD, Lagorio CH, Ko MC. Effects of the NOP agonist SCH221510 on producing and attenuating reinforcing effects as measured by drug self-administration in rats. Eur J Pharmacol 2014; 745:182-9. [PMID: 25446568 DOI: 10.1016/j.ejphar.2014.10.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 10/15/2014] [Accepted: 10/16/2014] [Indexed: 10/24/2022]
Abstract
Nociceptin/orphanin FQ peptide (NOP) receptor agonists attenuate morphine-induced conditioned place preference in rodents. However, it is not known whether NOP agonists have reinforcing properties or can inhibit mu opioid receptor (MOP)-mediated reinforcement as measured by drug self-administration in rodents. Further understanding the behavioral effects of NOP agonists could suggest them as having potential in attenuating reinforcing effects of opioids. In the first part of the study, reinforcing properties of selective NOP agonist SCH221510 were determined and compared with the full MOP agonist remifentanil under fixed-ratio 5 (FR5) and progressive-ratio (PR) schedules of drug self-administration. In the second part, effects of systemic and intracisternal pretreatment of SCH221510 were determined and compared with MOP antagonist naltrexone in attenuating reinforcing effects of remifentanil and a non-drug reinforcer (sucrose pellets). Remifentanil self-administration (0.3-10 µg/kg/infusion) generated a biphasic dose-response curve, characteristic of drugs with reinforcing properties. SCH221510 (3-300 µg/kg/infusion) self-administration resulted in flat dose-response curves and early break-points under the PR, indicative of drugs lacking reinforcing value. Intracisternally, but not systemically, administered SCH221510 (0.3-3 µg) attenuated remifentanil self-administration, comparable with systemic naltrexone (0.03-0.3 mg/kg). SCH221510 (1-3 µg), unlike naltrexone (0.03-1 mg/kg), attenuated responding for sucrose pellets. Both effects of SCH221510 were reversed by the NOP antagonist J-113397 (0.3-3 µg). These results suggest that SCH221510 does not function as a reinforcer in rats, and that it can attenuate the reinforcing value of MOP agonists; therefore, the potential utility of NOP agonists for the treatment of drug addiction warrants further evaluation.
Collapse
Affiliation(s)
- Devki D Sukhtankar
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | - Carla H Lagorio
- Department of Psychology, University of Wisconsin-Eau Claire, Eau Claire, WI 54702, USA
| | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Center for Comparative Medicine Research, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
27
|
Coletta R, Khalil BA, Morabito A. Short bowel syndrome in children: surgical and medical perspectives. Semin Pediatr Surg 2014; 23:291-7. [PMID: 25459014 DOI: 10.1053/j.sempedsurg.2014.09.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The main cause of intestinal failure in children is due to short bowel syndrome (SBS) resulting from congenital or acquired intestinal lesions. From the first lengthening procedure introduced by Bianchi, the last three decades have seen lengthening procedures established as fundamental components of multidisciplinary intestinal rehabilitation programs. Debate on indications and timing of the procedures is still open leaving SBS surgical treatment a great challenge. However, enteral autonomy is possible only with an individualized approach remembering that each SBS patient is unique. Current literature on autologous gastrointestinal reconstruction technique was reviewed aiming to assess a comprehensive pathway in SBS non-transplant management.
Collapse
Affiliation(s)
- Riccardo Coletta
- Paediatric Autologous Bowel Reconstruction and Rehabilitation Unit, Royal Manchester Children׳s Hospital, Oxford Rd, Manchester M13 9WL, UK; School of Medicine, University of Manchester, Manchester, UK
| | - Basem A Khalil
- Paediatric Autologous Bowel Reconstruction and Rehabilitation Unit, Royal Manchester Children׳s Hospital, Oxford Rd, Manchester M13 9WL, UK; School of Medicine, University of Manchester, Manchester, UK
| | - Antonino Morabito
- Paediatric Autologous Bowel Reconstruction and Rehabilitation Unit, Royal Manchester Children׳s Hospital, Oxford Rd, Manchester M13 9WL, UK; School of Medicine, University of Manchester, Manchester, UK.
| |
Collapse
|
28
|
Jagla C, Martus P, Stein C. Peripheral opioid receptor blockade increases postoperative morphine demands--a randomized, double-blind, placebo-controlled trial. Pain 2014; 155:2056-62. [PMID: 25046272 DOI: 10.1016/j.pain.2014.07.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 06/11/2014] [Accepted: 07/15/2014] [Indexed: 12/11/2022]
Abstract
Experimental studies suggest that a large proportion of opioid analgesia can be mediated by peripheral opioid receptors. This trial examined the contribution of such receptors to clinical analgesia induced by intravenous morphine. We hypothesized that the selective blockade of peripheral opioid receptors by methylnaltrexone (MNX) would increase the patients' demand for morphine to achieve satisfactory postoperative pain relief. In a double-blind, placebo-controlled, sequential 2-center trial, 50 patients undergoing knee replacement surgery were randomized (1:1) to receive either subcutaneous MNX (0.9 mg/kg) (hospital I: n=14; hospital II: n=11) or saline (hospital I: n=13; hospital II: n=12) at the end of surgery. The primary endpoint was the cumulative amount of intravenous morphine administered during the first 8 hours. Secondary endpoints were pain scores at rest and during movement (by numerical rating scale and McGill Questionnaire), vital signs, adverse side effects, and withdrawal symptoms. After MNX, demands for morphine were strongly (by about 40%) increased (hospital I: 35.31 ± 12.99 mg vs 25.51 ± 7.92 mg, P=0.03; hospital II: 35.42 ± 11.73 mg vs 24.80 ± 7.84 mg, P=0.02; pooled data: P<.001; means ± SD). Secondary endpoints were similar in all groups (P>.05). Thus, a significant proportion of analgesia produced by systemically administered morphine is mediated by peripheral opioid receptors. Drugs that selectively activate such receptors should have the potential to produce powerful clinical pain relief.
Collapse
Affiliation(s)
- Christina Jagla
- Klinik für Anaesthesiologie und Operative Intensivmedizin, Charité, Campus Benjamin Franklin, Freie Universität Berlin, Berlin, Germany; Helmholtz Virtual Institute Multifunctional Biomaterials for Medicine, Teltow, Germany
| | - Peter Martus
- Institut für Klinische Epidemiologie und Angewandte Biometrie, Universität Tübingen, Tübingen, Germany
| | - Christoph Stein
- Klinik für Anaesthesiologie und Operative Intensivmedizin, Charité, Campus Benjamin Franklin, Freie Universität Berlin, Berlin, Germany; Helmholtz Virtual Institute Multifunctional Biomaterials for Medicine, Teltow, Germany.
| |
Collapse
|
29
|
Zand F, Amini A, Asadi S, Farbood A. The effect of methylnaltrexone on the side effects of intrathecal morphine after orthopedic surgery under spinal anesthesia. Pain Pract 2014; 15:348-54. [PMID: 24571193 DOI: 10.1111/papr.12185] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 01/10/2014] [Indexed: 12/26/2022]
Abstract
Methylnaltrexone is a peripheral opioid receptor antagonist that does not cross the blood-brain barrier; so without interference with pain relief, it could reverse the peripheral opioid side effects such as constipation, pruritus, postoperative ileus, and urinary retention. This study has been designed to evaluate the effect of methylnaltrexone on postoperative side effects of intrathecal morphine. In seventy-two 18- to 55-year-old patients scheduled for elective orthopedic operations under spinal anesthesia, neuraxial blockade was achieved using 10 mg 0.5% hyperbaric bupivacaine and 0.1 mg preservative-free morphine sulfate. The first group (M) received 12 mg methylnaltrexone, while the second group (P) received normal saline, subcutaneously, immediately after spinal block in a randomized, double-blind fashion. There was a significant decrease in the rate of nausea and vomiting in group M, but there was no significant difference in the rate of pruritus or urinary retention between the two groups. Pain score was significantly lower in group M. Respiratory depression or decreased level of consciousness was not reported in any patient. Subcutaneous administration of methylnaltrexone was not effective in decreasing postoperative urinary retention and pruritus, but lowered the rate of nausea and vomiting and pain score after intrathecal bupivacaine and morphine.
Collapse
Affiliation(s)
- Farid Zand
- Shiraz Anesthesiology and Critical Care Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | | |
Collapse
|
30
|
Kumpf VJ. Pharmacologic management of diarrhea in patients with short bowel syndrome. JPEN J Parenter Enteral Nutr 2014; 38:38S-44S. [PMID: 24463352 DOI: 10.1177/0148607113520618] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Diarrhea associated with short bowel syndrome (SBS) can have multiple etiologies, including accelerated intestinal transit, gastric acid hypersecretion, intestinal bacterial overgrowth, and malabsorption of fats and bile salts. As a result, patients may need multiple medications to effectively control fecal output. The armamentarium of antidiarrheal drugs includes antimotility agents, antisecretory drugs, antibiotics and probiotics, bile acid-binding resins, and pancreatic enzymes. An antidiarrheal regimen must be individualized for each patient and should be developed using a methodical, stepwise approach. Treatment should be initiated with a single first-line medication at the low end of its dosing range. Dosage and/or dosing frequency can then be slowly escalated to achieve maximal effect while minimizing adverse events. If diarrhea remains poorly controlled, additional agents can be incorporated sequentially. If modification of the regimen is required, a single medication should be altered or exchanged at a time. After each adjustment of the regimen, sufficient time should be permitted to fully assess response (≥3-5 days) before initiating additional changes. SBS-associated malabsorption is a major obstacle to optimization of an antidiarrheal regimen because drug absorption is impaired. Patients may benefit from high dosages and/or frequent dosing intervals, liquid preparations, or nonoral routes of drug delivery. Although the diarrhea associated with SBS can be debilitating, effective pharmaceutical management has the potential to substantially improve health outcomes and quality of life for these patients.
Collapse
|
31
|
Sánchez-Fernández C, Montilla-García Á, González-Cano R, Nieto FR, Romero L, Artacho-Cordón A, Montes R, Fernández-Pastor B, Merlos M, Baeyens JM, Entrena JM, Cobos EJ. Modulation of peripheral μ-opioid analgesia by σ1 receptors. J Pharmacol Exp Ther 2014; 348:32-45. [PMID: 24155346 DOI: 10.1124/jpet.113.208272] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We evaluated the effects of σ1-receptor inhibition on μ-opioid-induced mechanical antinociception and constipation. σ1-Knockout mice exhibited marked mechanical antinociception in response to several μ-opioid analgesics (fentanyl, oxycodone, morphine, buprenorphine, and tramadol) at systemic (subcutaneous) doses that were inactive in wild-type mice and even unmasked the antinociceptive effects of the peripheral μ-opioid agonist loperamide. Likewise, systemic (subcutaneous) or local (intraplantar) treatment of wild-type mice with the selective σ1 antagonists BD-1063 [1-[2-(3,4-dichlorophenyl)ethyl]-4-methylpiperazine dihydrochloride] or S1RA [4-[2-[[5-methyl-1-(2-naphthalenyl)1H-pyrazol-3-yl]oxy]ethyl] morpholine hydrochloride] potentiated μ-opioid antinociception; these effects were fully reversed by the σ1 agonist PRE-084 [2-(4-morpholinethyl)1-phenylcyclohexanecarboxylate) hydrochloride], showing the selectivity of the pharmacological approach. The μ-opioid antinociception potentiated by σ1 inhibition (by σ1-receptor knockout or σ1-pharmacological antagonism) was more sensitive to the peripherally restricted opioid antagonist naloxone methiodide than opioid antinociception under normal conditions, indicating a key role for peripheral opioid receptors in the enhanced antinociception. Direct interaction between the opioid drugs and σ1 receptor cannot account for our results, since the former lacked affinity for σ1 receptors (labeled with [(3)H](+)-pentazocine). A peripheral role for σ1 receptors was also supported by their higher density (Western blot results) in peripheral nervous tissue (dorsal root ganglia) than in several central areas involved in opioid antinociception (dorsal spinal cord, basolateral amygdala, periaqueductal gray, and rostroventral medulla). In contrast to its effects on nociception, σ1-receptor inhibition did not alter fentanyl- or loperamide-induced constipation, a peripherally mediated nonanalgesic opioid effect. Therefore, σ1-receptor inhibition may be used as a systemic or local adjuvant to enhance peripheral μ-opioid analgesia without affecting opioid-induced constipation.
Collapse
MESH Headings
- Analgesics, Opioid/antagonists & inhibitors
- Analgesics, Opioid/pharmacology
- Animals
- Constipation/chemically induced
- Constipation/genetics
- Constipation/metabolism
- Female
- Ganglia, Spinal/drug effects
- Ganglia, Spinal/metabolism
- Ganglia, Spinal/physiology
- Mice
- Mice, Knockout
- Pain Measurement/methods
- Receptors, Opioid, mu/metabolism
- Receptors, Opioid, mu/physiology
- Receptors, sigma/deficiency
- Receptors, sigma/genetics
- Receptors, sigma/physiology
- Sigma-1 Receptor
Collapse
Affiliation(s)
- Cristina Sánchez-Fernández
- Departments of Pharmacology (C.S.-F., A.M.-G., R.G.-C., F.R.N., L.R., A.A.-C., J.M.B., E.J.C.) and Physiology (R.M.), School of Medicine, University of Granada, Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain (C.S.-F., R.G.-C., F.R.N., R.M., J.M.B., J.M.E., E.J.C.); Animal Behavior Research Unit, Scientific Instrumentation Center, University of Granada, Granada, Spain (J.M.E.); and Drug Discovery and Preclinical Development, Barcelona, Spain (B.F.-P., M.M.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
INTRODUCTION Centrally acting opioids are well established in the treatment of acute, surgical and cancer pain. However, their use in chronic noncancer pain (CNCP) is controversial because of side effects such as tolerance, somnolence, respiratory depression, confusion, constipation and addiction. Chronic arthritis and other musculoskeletal diseases are among the leading causes of CNCP. AREAS COVERED This manuscript will discuss the role of conventional opioids in chronic arthritis. In addition, future developments and strategies exploiting peripheral effects of opioids on pain and inflammation will be outlined. EXPERT OPINION Aims in drug development include the design of peripherally restricted opioid agonists, selective targeting of endogenous opioids to sites of painful injury and the augmentation of peripheral ligand and receptor synthesis, for example, by gene therapy. Although a large number of peripherally acting opioid compounds have been developed, clinical Phase III studies have not been published so far. Another strategy is to augment the effects of endogenously released opioid peptides by the inhibition of their degrading enzymes. Technology-oriented research is needed to find novel ways of peripheral restriction of opioids. Such analgesics would be desirable for their lack of central side effects and of adverse effects typical of nonsteroidal anti-inflammatory drugs (gastrointestinal ulcers, bleeding, myocardial infarction and stroke).
Collapse
Affiliation(s)
- Christoph Stein
- Freie Universitaet Berlin, Charite Campus Benjamin Franklin, Department of Anaesthesiology and Critical Care Medicine , Berlin , Germany
| | | |
Collapse
|
33
|
Kumar R, Reeta K, Ray SB. Antinociceptive effect of intrathecal loperamide: Role of mu-opioid receptor and calcium channels. Eur J Pharmacol 2012; 696:77-82. [DOI: 10.1016/j.ejphar.2012.09.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 08/31/2012] [Accepted: 09/17/2012] [Indexed: 10/27/2022]
|
34
|
Rubelt MS, Amasheh S, Grobosch T, Stein C. Liquid chromatography-tandem mass spectrometry for analysis of intestinal permeability of loperamide in physiological buffer. PLoS One 2012; 7:e48502. [PMID: 23144895 PMCID: PMC3493557 DOI: 10.1371/journal.pone.0048502] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 09/26/2012] [Indexed: 11/18/2022] Open
Abstract
Analysis of in vitro samples with high salt concentrations represents a major challenge for fast and specific quantification with liquid chromatography-tandem mass spectrometry (LC-MS/MS). To investigate the intestinal permeability of opioids in vitro employing the Ussing chamber technique, we developed and validated a fast, sensitive and selective method based on LC–MS/MS for the determination of loperamide in HEPES-buffered Ringer's solution. Chromatographic separation was achieved with an Atlantis dC18 column, 2.1 mm×20 mm, 3 µm particle size and a gradient consisting of methanol/0.1% formic acid and ammonium acetate. The flow rate was 0.7 ml/min, and the total run time was 3 min. For quantification, two mass transitions for loperamide and a deuterated internal standard (methadone-d3) were used. The lower limit of loperamide quantification was 0.2 ng/ml. This new LC-MS/MS method can be used for the detection of loperamide in any experimental setup using HEPES-buffered Ringer's solution as a matrix compound.
Collapse
Affiliation(s)
- Miriam S Rubelt
- Department of Anesthesiology, Charité, Campus Benjamin Franklin, Berlin, Germany.
| | | | | | | |
Collapse
|
35
|
Pinheiro MMG, Boylan F, Fernandes PD. Antinociceptive effect of the Orbignya speciosa Mart. (Babassu) leaves: Evidence for the involvement of apigenin. Life Sci 2012; 91:293-300. [DOI: 10.1016/j.lfs.2012.06.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 04/21/2012] [Accepted: 06/14/2012] [Indexed: 11/26/2022]
|
36
|
Chung C, Carteret AF, McKelvy AD, Ringkamp M, Yang F, Hartke TV, Dong X, Raja SN, Guan Y. Analgesic properties of loperamide differ following systemic and local administration to rats after spinal nerve injury. Eur J Pain 2012; 16:1021-32. [PMID: 22508374 DOI: 10.1002/j.1532-2149.2012.00148.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2012] [Indexed: 01/24/2023]
Abstract
BACKGROUND The analgesic properties and mechanisms of loperamide hydrochloride, a peripherally acting opioid receptor agonist, in neuropathic pain warrant further investigation. METHODS We examined the effects of systemic or local administration of loperamide on heat and mechanical hyperalgesia in rats after an L5 spinal nerve ligation (SNL). RESULTS (1) Systemic loperamide (0.3-10 mg/kg, subcutaneous in the back) dose dependently reversed heat hyperalgesia in SNL rats, but did not produce thermal analgesia. Systemic loperamide (3 mg/kg) did not induce thermal antinociception in naïve rats; (2) systemic loperamide-induced anti-heat hyperalgesia was blocked by pretreatment with intraperitoneal naloxone methiodide (5 mg/kg), but not by intraperitoneal naltrindole (5 mg/kg) or intrathecal naltrexone (20 μg/10 μL); (3) local administration of loperamide (150 μg), but not vehicle, into plantar or dorsal hind paw tissue induced thermal analgesia in SNL rats and thermal antinociception in naïve rats; (4) the analgesic effect of intraplantar loperamide (150 μg/15 μL) in SNL rats at 45 min, but not 10 min, post-injection was blocked by pretreatment with an intraplantar injection of naltrexone (75 μg/10 μL); (5) systemic (3.0 mg/kg) and local (150 μg) loperamide reduced the exaggerated duration of hind paw elevation to noxious pinprick stimuli in SNL rats. Intraplantar injection of loperamide also decreased the frequency of pinprick-evoked response in naïve rats. CONCLUSIONS These findings suggest that both systemic and local administration of loperamide induce an opioid receptor-dependent inhibition of heat and mechanical hyperalgesia in nerve-injured rats, but that local paw administration of loperamide also induces thermal and mechanical antinociception.
Collapse
Affiliation(s)
- C Chung
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Mizuta K, Fujita T, Kumamoto E. Inhibition by morphine and its analogs of action potentials in adult rat dorsal root ganglion neurons. J Neurosci Res 2012; 90:1830-41. [DOI: 10.1002/jnr.23059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Revised: 02/08/2012] [Accepted: 03/04/2012] [Indexed: 12/19/2022]
|
38
|
Davis CM, Cobuzzi JL, Riley AL. Assessment of the aversive effects of peripheral mu opioid receptor agonism in Fischer 344 and Lewis rats. Pharmacol Biochem Behav 2012; 101:181-6. [PMID: 22260873 DOI: 10.1016/j.pbb.2012.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Revised: 12/22/2011] [Accepted: 01/03/2012] [Indexed: 12/01/2022]
Abstract
The Fischer 344 (F344) and Lewis (LEW) inbred rat strains differ on a host of biochemical, neuroanatomical, immunological and behavioral endpoints. One behavioral difference of interest is their differential reactivity to the aversive effects of morphine as indexed by the conditioned taste aversion preparation (aversions acquired by F344 rats are significantly greater than those acquired by the LEW strain). This differential effect appears to be specific to opioids that work primarily on the mu opioid receptor. Given that morphine works systemically, it is unknown whether these differential effects in F344 and LEW animals are centrally or peripherally mediated. To address this issue, the present study investigated the ability of the peripherally acting mu preferring opioid agonist loperamide to induce differential taste aversions in F344 and LEW animals. Both F344 and LEW animals acquired dose-dependent taste aversions to the loperamide-associated solution with no difference between them. Additionally, control animals initially injected with vehicle during aversion training with loperamide and subsequently conditioned with morphine displayed the typical aversive profile to morphine (F344>LEW). Although the basis for the present data is unknown, their relation to morphine-induced taste aversions and the role of the interaction of stimulus effects of drugs that produce differential abuse liability were discussed.
Collapse
Affiliation(s)
- Catherine M Davis
- Psychopharmacology Laboratory, Department of Psychology, American University, Washington, DC 20016, United States
| | | | | |
Collapse
|
39
|
Stein C, Machelska H. Modulation of peripheral sensory neurons by the immune system: implications for pain therapy. Pharmacol Rev 2011; 63:860-81. [PMID: 21969325 DOI: 10.1124/pr.110.003145] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
The concept that the immune system can communicate with peripheral sensory neurons to modulate pain is based mostly on documented interactions between opioid ligands and receptors. Such findings may have broad implications for the development of safer pain medication. Innovative strategies take into account that analgesics should be particularly active in pathological states rather than producing a general suppression of the central nervous system, as with conventional morphine- or cannabinoid-like drugs. Inflammation of peripheral tissue leads to increased functionality of opioid receptors on peripheral sensory neurons and to local production of endogenous opioid peptides. In addition, endocannabinoids were detected in leukocytes, but their role in pain modulation has yet to be addressed. Future aims include the development of peripherally restricted opioid agonists, selective targeting of opioid-containing immune cells to sites of painful injury, and the augmentation of peripheral ligand and receptor synthesis (e.g., by gene therapy). Similar approaches may be pursued for cannabinoids. The ultimate goal is to avoid detrimental side effects of currently available analgesics such as respiratory depression, cognitive impairment, addiction, gastrointestinal bleeding, and thromboembolic complications.
Collapse
Affiliation(s)
- Christoph Stein
- Department of Anesthesiology and Critical Care Medicine, Freie Universität Berlin, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany.
| | | |
Collapse
|
40
|
Mitch CH, Quimby SJ, Diaz N, Pedregal C, de la Torre MG, Jimenez A, Shi Q, Canada EJ, Kahl SD, Statnick MA, McKinzie DL, Benesh DR, Rash KS, Barth VN. Discovery of aminobenzyloxyarylamides as κ opioid receptor selective antagonists: application to preclinical development of a κ opioid receptor antagonist receptor occupancy tracer. J Med Chem 2011; 54:8000-12. [PMID: 21958337 DOI: 10.1021/jm200789r] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Arylphenylpyrrolidinylmethylphenoxybenzamides were found to have high affinity and selectivity for κ opioid receptors. On the basis of receptor binding assays in Chinese hamster ovary (CHO) cells expressing cloned human opioid receptors, (S)-3-fluoro-4-(4-((2-(3-fluorophenyl)pyrrolidin-1-yl)methyl)phenoxy)benzamide (25) had a K(i) = 0.565 nM for κ opioid receptor binding while having a K(i) = 35.8 nM for μ opioid receptors and a K(i) = 211 nM for δ opioid receptor binding. Compound 25 was also a potent antagonist of κ opioid receptors when tested in vitro using a [(35)S]-guanosine 5'O-[3-thiotriphosphate] ([(35)S]GTP-γ-S) functional assay in CHO cells expressing cloned human opioid receptors. Compounds were also evaluated for potential use as receptor occupancy tracers. Tracer evaluation was done in vivo, using liquid chromatography-tandem mass spectrometry (LC/MS/MS) methods, precluding the need for radiolabeling. (S)-3-Chloro-4-(4-((2-(pyridine-3-yl)pyrrolidin-1-yl)methyl)phenoxy)benzamide (18) was found to have favorable properties for a tracer for receptor occupancy, including good specific versus nonspecific binding and good brain uptake.
Collapse
Affiliation(s)
- Charles H Mitch
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, Indiana 46285-0150, United States.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
de Freitas Pires A, Assreuy AMS, Lopes ÉAB, Celedônio NR, Soares CEA, Rodrigues NVFC, Sousa PL, Benevides RG, Nagano CS, Cavada BS, Leal-Cardoso JH, Coelho-de-Souza AN, Santos CF. Opioid-like antinociceptive effects of oral administration of a lectin purified from the seeds of Canavalia brasiliensis. Fundam Clin Pharmacol 2011; 27:201-9. [PMID: 21895762 DOI: 10.1111/j.1472-8206.2011.00987.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The objective of this study was to evaluate the antinociceptive effects of a lectin from Canavalia brasiliensis (ConBr) when administered orally to murine models of chemical and thermal nociception. ConBr up to 100 mg/kg produced significant and dose-dependent antinociceptive effects: 81% reduction in abdominal writhing induced by 0.6% acetic acid; 26 and 52% reduction in early- and late-stage paw licking, respectively, induced by 2.5% formalin; and 155% increase in reaction latency (heightened thermal pain threshold). In all models, the antinociceptive effect was reversed by the lectin-binding carbohydrate α-d-methyl-mannoside and by the nonselective opioid antagonist naloxone. The antinociceptive effect observed in the formalin test was inhibited by the δ-selective antagonist naltrindole and the κ-selective antagonist nor-binaltorphimine but not by the μ-selective antagonist cyprodime. In conclusion, when administered orally to Swiss mice, the ConBr lectin displayed antinociceptive activity, both peripheral and central, mediated by the opioid system and involving δ-and κ-receptors and the lectin domain.
Collapse
Affiliation(s)
- Alana de Freitas Pires
- Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Av. Paranjana 1700, 60.740-000, Fortaleza, Ceará, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
|
43
|
Farias WRL, Lima PCWC, Rodrigues NVFC, Siqueira RCL, Amorim RMF, Pereira MG, Assreuy AMS. A Novel Antinociceptive Sulphated Polysaccharide of the Brown Marine Alga Spatoglossum Schroederi. Nat Prod Commun 2011. [DOI: 10.1177/1934578x1100600626] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Sulfated polysaccharides (SP) of brown algae (Phaeophyta) are composed mainly of α- L-fucose, being classified as fucans, with recognized role in inflammation but not in nociception, which was already described for SP obtained from red algae. Here the SP of the brown marine alga S. schroederi (named Ss-SP) was isolated and assayed for the antinociceptive effect. Ss-SP was isolated by DEAE-cellulose, analyzed by agarose gel electrophoresis and evaluated in nociception models (Formalin, Hot plate, Von Frey) using Swiss mice (20-25g). Anion exchange chromatography provided four major fractions being F1 (Ss-SP) that of highest metachromatic activity and sugar content. Ss-SP inhibited both phases of the formalin test. In the first phase the paw licking (55.2±8.07s) was reduced by 45% (30.5±6.51s) and 40% (32.85±8.66s) at 0.1 and 1 μg/kg, respectively. In the second phase, Ss-SP was also inhibitory about 39%, but only at 1 mg/kg (83.0±15.70s) compared to formalin (136.8±10.27s). This inhibitory effect suggests a mixed mechanism similar to morphine, which was not confirmed in the hot plate test, a model of pain associated with central neurotransmission. However, Ss-SP reduced the animal reaction in response to stimulation withVon Frey filament at the 2nd and 3rd h (20.8±6.86% versus carrageenan: 47.9±5.83%; 33.3±7.71% versus carrageenan: 62.5±9.83%). Accordingly, the paw edema induced by carrageenan (0.08±0.01g) was potently reduced in 45.35% by Ss-SP pre-treatment (0.02±0.003g), corroborating the anti-inflammatory activity demonstrated for brown seaweed polysaccharides. In conclusion our data revealed for the first time the antinociceptive effect of Ss-SP which could be used as a new source of analgesic substances.
Collapse
Affiliation(s)
- Wladimir R. L. Farias
- Departamento de Engenharia de Pesca, Universidade Federal do Ceará, Av. Mister Hull s/n Bloco 827, 60.356-000, Fortaleza-CE-Brasil
| | - Paula Cristina W. C. Lima
- Departamento de Engenharia de Pesca, Universidade Federal do Ceará, Av. Mister Hull s/n Bloco 827, 60.356-000, Fortaleza-CE-Brasil
| | - Natália Velloso F. C. Rodrigues
- Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Av. Paranjana 1700, 60.740-000, Fortaleza-CE-Brasil
| | - Rômmulo Celly L. Siqueira
- Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Av. Paranjana 1700, 60.740-000, Fortaleza-CE-Brasil
| | - Renata M. F. Amorim
- Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Av. Paranjana 1700, 60.740-000, Fortaleza-CE-Brasil
| | - Maria G. Pereira
- Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Av. Paranjana 1700, 60.740-000, Fortaleza-CE-Brasil
| | - Ana Maria S. Assreuy
- Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Av. Paranjana 1700, 60.740-000, Fortaleza-CE-Brasil
| |
Collapse
|
44
|
|
45
|
Gavra T, Libersat F. Involvement of the opioid system in the hypokinetic state induced in cockroaches by a parasitoid wasp. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2010; 197:279-91. [DOI: 10.1007/s00359-010-0610-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 11/01/2010] [Accepted: 11/04/2010] [Indexed: 11/24/2022]
|
46
|
Opioid receptors and opioid peptide-producing leukocytes in inflammatory pain--basic and therapeutic aspects. Brain Behav Immun 2010; 24:683-94. [PMID: 19879349 DOI: 10.1016/j.bbi.2009.10.013] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2009] [Revised: 10/22/2009] [Accepted: 10/24/2009] [Indexed: 12/12/2022] Open
Abstract
This review summarizes recent findings on neuro-immune mechanisms underlying opioid-mediated inhibition of pain. The focus is on events occurring in peripheral injured tissues that lead to the sensitization and excitation of primary afferent neurons, and on the modulation of such mechanisms by immune cell-derived opioid peptides. Primary afferent neurons are of particular interest from a therapeutic perspective because they are the initial generators of impulses relaying nociceptive information towards the spinal cord and the brain. Thus, if one finds ways to inhibit the sensitization and/or excitation of peripheral sensory neurons, subsequent central events such as wind-up, sensitization and plasticity may be prevented. This is in part achieved by endogenously released immune cell-derived opioid peptides within inflamed tissue. In addition, exogenous opioid receptor ligands that selectively modulate primary afferent function and do not cross the blood-brain barrier, avoid centrally mediated untoward side effects of conventional analgesics (e.g., opioids, anticonvulsants). This article discusses peripheral opioid receptors and their signaling pathways, opioid peptide-producing/secreting inflammatory cells and arising therapeutic perspectives.
Collapse
|
47
|
Gomes NDM, de Rezende CM, Fontes SP, Matheus ME, Pinto ADC, Fernandes PD. Characterization of the antinociceptive and anti-inflammatory activities of fractions obtained from Copaifera multijuga Hayne. JOURNAL OF ETHNOPHARMACOLOGY 2010; 128:177-183. [PMID: 20064592 DOI: 10.1016/j.jep.2010.01.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 12/22/2009] [Accepted: 01/03/2010] [Indexed: 05/28/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Copaifera multijuga Hayne (Leguminosae) is a tree that produces an oleoresin, which is extensively commercialized in Brazil as capsules or crude oil for the treatment of several disorders. Ethnopharmacological studies show a diversity of indications such as anti-inflammatory and epidermal wound cicatrization. AIM OF THE STUDY In the present work three fractions obtained from Copaifera multijuga oleoresin (hexane (HF), chloroform (CF), and methanol (MF) from a KOH impregnated silica gel column chromatography, representing the three main classes of compounds in the Copaifera genus (hydrocarbon sesquiterpenes, oxygenated sesquiterpenes and acidic diterpenes), were evaluated using antinociceptive and anti-inflammatory models. MATERIALS AND METHODS HF, CF, and MF (doses ranging between 1 and 150 mg/kg, depending on the model used), Copaifera multijuga oleoresin (CMO, 100mg/kg, p.o.) and the reference drug morphine (5mg/kg, p.o.) were evaluated using models for analgesia (acetic acid-induced contortions and tail flick) or inflammation (rat paw oedema and increase in vascular permeability). To elucidate the mechanism of action from the fractions, animals were pre-treated with naloxone (opioid receptor antagonist, 5mg/kg, i.p.). RESULTS Fractions significantly inhibited (in a concentration-dependant way) the number of contortions induced by acetic acid and the second phase of formalin-induced licking response. Similar results were observed in the tail flick model. The central antinociceptive effect for HF and CF at the doses of 50 and 100mg/kg was higher than the one observed for morphine (1mg/kg). Administration of naloxone inhibited the antinociceptive effect of fractions indicating that HF, CF, and MF may be acting on opioid receptors. All three fractions also inhibited rat paw oedema and the increase in vascular permeability induced by several phlogistic agents (carrageenan, histamine, and serotonin). CONCLUSIONS Our results indicate that fractions obtained from Copaifera multijuga Hayne demonstrate an antinociceptive effect probably mediated by opioid receptors, and anti-inflammatory activity through inhibition of histaminergic and serotoninergic pathways.
Collapse
Affiliation(s)
- Niele de Matos Gomes
- Laboratório de Farmacologia da Inflamação e do Oxido Nítrico, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | | | | | | | | | | |
Collapse
|
48
|
Buprenorphine inhibits bradykinin-induced release of calcitonin gene-related peptide from rat trigeminal neurons via both mu-opioid and nociceptin/orphanin peptide receptors. Eur J Pharmacol 2009; 609:45-50. [PMID: 19285495 DOI: 10.1016/j.ejphar.2009.03.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Revised: 03/04/2009] [Accepted: 03/05/2009] [Indexed: 12/14/2022]
Abstract
In this study we used the dual opioid and nociceptin/orphanin peptide (NOP) agonist buprenorphine to investigate the relative contributions of opioid and NOP systems in regulating bradykinin-stimulated calcitonin-gene related peptide (CGRP) release from primary cultures of neonatal rat trigeminal neurons. We found that: bradykinin stimulates CGRP secretion either by a direct effect or after applying so-called "bradykinin-priming" protocol. In both cases, buprenorphine was able to inhibit bradykinin-stimulated CGRP secretion; however, inhibition was mediated by NOP receptors when buprenorphine was added to the incubation medium along with bradykinin, whereas it appeared to be mediated by mu-opioid receptors in bradykinin priming experiments. Bradykinin treatments also caused an increase in neuronal prostaglandin production; prostanoids appeared to be involved in the stimulatory effects of bradykinin as well as in buprenorphine inhibition, through apparently unrelated mechanisms.
Collapse
|
49
|
He L, Kim J, Ou C, McFadden W, van Rijn RM, Whistler JL. Methadone antinociception is dependent on peripheral opioid receptors. THE JOURNAL OF PAIN 2009; 10:369-79. [PMID: 19327642 PMCID: PMC2952445 DOI: 10.1016/j.jpain.2008.09.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/13/2008] [Revised: 09/06/2008] [Accepted: 09/12/2008] [Indexed: 01/30/2023]
Abstract
UNLABELLED Morphine and methadone are both high-affinity, potent mu-opioid peptide (MOP) receptor analgesics. In this report, we compared the antinociceptive potencies of these 2 drugs when administered subcutaneously (s.c.), intrathecally (i.t.), or intracerebroventricularly (i.c.v.) in both rat and mouse, using the tail-flick assay. We found that both morphine and methadone were potently antinociceptive when the drugs were administered s.c., showing comparable AD50 values in both species. However, the antinociception produced by methadone, when it was administered centrally, was much weaker than that produced by centrally administered morphine. Specifically, the AD50 value for methadone antinociception was more than 30-fold higher at both the i.t. and i.c.v. sites in mouse and not measurable in rat. Naloxone methiodide (NLX-M), a peripherally restricted antagonist, was used to further examine the relative contribution of central versus peripheral sites to morphine and methadone antinociception. NLX-M, when administered s.c., blocked the antinociceptive effect of either systemically or centrally administered methadone but had little effect on the antinociception produced by centrally administered morphine. Furthermore, centrally administered NLX-M significantly blocked antinociception produced by centrally administered morphine but not that produced by centrally administered methadone. Together, these results suggest that methadone antinociception is significantly dependent on an action of the drug at peripheral sites and could provide novel insight into the neural mechanisms that distinguish morphine versus methadone antinociception. PERSPECTIVE Methadone is often used as an alternative for pain management. The present study shows that a peripheral action plays a crucial role in methadone antinociception. This finding could have significant clinical relevance for the use of methadone versus morphine for the treatment of certain types of pain.
Collapse
Affiliation(s)
- Li He
- Ernest Gallo Clinic and Research Center and the Department of Neurology, University of California, San Francisco, 5858 Horton Street, Suite 200, Emeryville, CA 94608 USA
| | - Joseph Kim
- Ernest Gallo Clinic and Research Center and the Department of Neurology, University of California, San Francisco, 5858 Horton Street, Suite 200, Emeryville, CA 94608 USA
| | - Chrissi Ou
- Ernest Gallo Clinic and Research Center and the Department of Neurology, University of California, San Francisco, 5858 Horton Street, Suite 200, Emeryville, CA 94608 USA
| | - Whitney McFadden
- Ernest Gallo Clinic and Research Center and the Department of Neurology, University of California, San Francisco, 5858 Horton Street, Suite 200, Emeryville, CA 94608 USA
| | - Richard M. van Rijn
- Ernest Gallo Clinic and Research Center and the Department of Neurology, University of California, San Francisco, 5858 Horton Street, Suite 200, Emeryville, CA 94608 USA
| | - Jennifer L. Whistler
- Ernest Gallo Clinic and Research Center and the Department of Neurology, University of California, San Francisco, 5858 Horton Street, Suite 200, Emeryville, CA 94608 USA
| |
Collapse
|
50
|
Stein C, Clark JD, Oh U, Vasko MR, Wilcox GL, Overland AC, Vanderah TW, Spencer RH. Peripheral mechanisms of pain and analgesia. BRAIN RESEARCH REVIEWS 2009; 60:90-113. [PMID: 19150465 PMCID: PMC2730351 DOI: 10.1016/j.brainresrev.2008.12.017] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Accepted: 12/29/2008] [Indexed: 12/23/2022]
Abstract
This review summarizes recent findings on peripheral mechanisms underlying the generation and inhibition of pain. The focus is on events occurring in peripheral injured tissues that lead to the sensitization and excitation of primary afferent neurons, and on the modulation of such mechanisms. Primary afferent neurons are of particular interest from a therapeutic perspective because they are the initial generator of noxious impulses traveling towards relay stations in the spinal cord and the brain. Thus, if one finds ways to inhibit the sensitization and/or excitation of peripheral sensory neurons, subsequent central events such as wind-up, sensitization and plasticity may be prevented. Most importantly, if agents are found that selectively modulate primary afferent function and do not cross the blood-brain-barrier, centrally mediated untoward side effects of conventional analgesics (e.g. opioids, anticonvulsants) may be avoided. This article begins with the peripheral actions of opioids, turns to a discussion of the effects of adrenergic co-adjuvants, and then moves on to a discussion of pro-inflammatory mechanisms focusing on TRP channels and nerve growth factor, their signaling pathways and arising therapeutic perspectives.
Collapse
Affiliation(s)
- Christoph Stein
- Department of Anesthesiology and Critical Care Medicine, Charité Campus Benjamin Franklin, Freie Universität Berlin, Germany
| | - J. David Clark
- Department of Anesthesia, Stanford University School of Medicine, USA
| | - Uhtaek Oh
- Sensory Research Center, CRI, Seoul National University, Korea
| | - Michael R. Vasko
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, USA
| | - George L. Wilcox
- Department of Neuroscience, University of Minnesota, Minneapolis, USA
| | - Aaron C. Overland
- Department of Neuroscience, University of Minnesota, Minneapolis, USA
| | - Todd W. Vanderah
- Department of Pharmacology, University of Arizona Health Sciences Center, Tucson, USA
| | | |
Collapse
|