1
|
López-Tofiño Y, Hopkins MA, Bagues A, Boullon L, Abalo R, Llorente-Berzal Á. The Endocannabinoid System of the Nervous and Gastrointestinal Systems Changes after a Subnoxious Cisplatin Dose in Male Rats. Pharmaceuticals (Basel) 2024; 17:1256. [PMID: 39458898 PMCID: PMC11509924 DOI: 10.3390/ph17101256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Cisplatin, a common chemotherapy agent, is well known to cause severe side effects in the gastrointestinal and nervous systems due to its toxic and pro-inflammatory effects. Although pharmacological manipulation of the endocannabinoid system (ECS) can alleviate these side effects, how chemotherapy affects the ECS components in these systems remains poorly understood. Our aim was to evaluate these changes. Methods: Male Wistar rats received cisplatin (5 mg/kg, i.p.) or saline on day 0 (D0). Immediately after, serial X-rays were taken for 24 h (D0). Body weight was recorded (D0, D1, D2 and D7) and behavioural tests were performed on D4. On D7, animals were euthanized, and gastrointestinal tissue, dorsal root ganglia (DRGs) and brain areas were collected. Expression of genes related to the ECS was assessed via Rt-PCR, while LC-MS/MS was used to analyse endocannabinoid and related N-acylethanolamine levels in tissue and plasma. Results: Animals treated with cisplatin showed a reduction in body weight. Cisplatin reduced gastric emptying during D0 and decreased MAGL gene expression in the antrum at D7. Despite cisplatin not causing mechanical or heat sensitivity, we observed ECS alterations in the prefrontal cortex (PFC) and DRGs similar to those seen in other chronic pain conditions, including an increased CB1 gene expression in L4/L5 DRGs and a decreased MAGL expression in PFC. Conclusions: A single dose of cisplatin (5 mg/kg, i.p.), subnoxious, but capable of inducing acute gastrointestinal effects, caused ECS changes in both gastrointestinal and nervous systems. Modulating the ECS could alleviate or potentially prevent chemotherapy-induced toxicity.
Collapse
Affiliation(s)
- Yolanda López-Tofiño
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (Y.L.-T.); (A.B.)
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
| | - Mary A. Hopkins
- Department of Pharmacology and Therapeutics, School of Medicine, University of Galway, H91W5P7 Galway, Ireland; (L.B.); (M.A.H.)
- Galway Neuroscience Centre, University of Galway, H91W5P7 Galway, Ireland
- Centre for Pain Research, University of Galway, H91W5P7 Galway, Ireland
| | - Ana Bagues
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (Y.L.-T.); (A.B.)
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
- High Performance Research Group in Experimental Pharmacology (PHARMAKOM-URJC), University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
- Associated R+D+i Unit to the Institute of Medicinal Chemistry (IQM), Spanish National Research Council (CSIC), 28006 Madrid, Spain
| | - Laura Boullon
- Department of Pharmacology and Therapeutics, School of Medicine, University of Galway, H91W5P7 Galway, Ireland; (L.B.); (M.A.H.)
- Galway Neuroscience Centre, University of Galway, H91W5P7 Galway, Ireland
- Centre for Pain Research, University of Galway, H91W5P7 Galway, Ireland
| | - Raquel Abalo
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (Y.L.-T.); (A.B.)
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
- High Performance Research Group in Experimental Pharmacology (PHARMAKOM-URJC), University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
- Associated R+D+i Unit to the Institute of Medicinal Chemistry (IQM), Spanish National Research Council (CSIC), 28006 Madrid, Spain
- Working Group of Basic Sciences on Pain and Analgesia, Spanish Pain Society, 28046 Madrid, Spain
- Working Group of Basic Sciences on Cannabinoids, Spanish Pain Society, 28046 Madrid, Spain
| | - Álvaro Llorente-Berzal
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
- Department of Pharmacology and Therapeutics, School of Medicine, University of Galway, H91W5P7 Galway, Ireland; (L.B.); (M.A.H.)
- Galway Neuroscience Centre, University of Galway, H91W5P7 Galway, Ireland
- Centre for Pain Research, University of Galway, H91W5P7 Galway, Ireland
- Department of Physiology, School of Medicine, Autonomous University of Madrid (UAM), 28049 Madrid, Spain
| |
Collapse
|
2
|
Déciga-Campos M, Jaramillo-Morales OA, Espinosa-Juárez JV, Aguilera-Martínez ME, Ventura-Martínez R, López-Muñoz FJ. N-palmitoylethanolamide synergizes the antinociception of morphine and gabapentin in the formalin test in mice. J Pharm Pharmacol 2023; 75:1154-1162. [PMID: 36905375 DOI: 10.1093/jpp/rgad004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 01/19/2023] [Indexed: 03/12/2023]
Abstract
OBJECTIVE The antinociceptive pharmacological interaction between N-palmitoylethanolamide (PEA) and morphine (MOR), as well as gabapentin (GBP), was investigated to obtain synergistic antinociception at doses where side effects were minimal. In addition, the possible antinociceptive mechanism of PEA + MOR or PEA + GBP combinations was explored. METHODS Individual dose-response curves (DRCs) of PEA, MOR and GBP were evaluated in female mice in which intraplantar nociception was induced with 2% formalin. Isobolographic method was used to detect the pharmacological interaction in the combination of PEA + MOR or PEA + GBP. KEY FINDINGS The ED50 was calculated from the DRC; the order of potency was MOR > PEA > GBP. The isobolographic analysis was obtained at a 1:1 ratio to determine the pharmacological interaction. The experimental values of flinching (PEA + MOR, Zexp = 2.72 ± 0.2 μg/paw and PEA + GBP Zexp = 2.77 ± 0.19 μg/paw) were significantly lower than those calculated theoretically (PEA + MOR Zadd = 7.78 ± 1.07 and PEA + GBP Zadd = 24.05 ± 1.91 μg/paw), resulting in synergistic antinociception. Pretreatment with GW6471 and naloxone demonstrated that peroxisome proliferator-activated receptor alpha (PPARα) and opioid receptors are involved in both interactions. CONCLUSIONS These results suggest that MOR and GBP synergistically enhance PEA-induced antinociception through PPARα and opioid receptor mechanisms. Furthermore, the results suggest that combinations containing PEA with MOR or GBP could be of interest in aiding the treatment of inflammatory pain.
Collapse
Affiliation(s)
- Myrna Déciga-Campos
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México, México
| | - Osmar Antonio Jaramillo-Morales
- Departamento de Enfermería y Obstetricia, División de Ciencias de la Vida, Campus Irapuato-Salamanca, Universidad de Guanajuato, Irapuato, Guanajuato, México
| | | | - María Elena Aguilera-Martínez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México, México
| | - Rosa Ventura-Martínez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Delegación Coyoacán, México, México
| | | |
Collapse
|
3
|
Effects of Intra-BLA Administration of PPAR Antagonists on Formalin-Evoked Nociceptive Behaviour, Fear-Conditioned Analgesia, and Conditioned Fear in the Presence or Absence of Nociceptive Tone in Rats. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27062021. [PMID: 35335382 PMCID: PMC8949000 DOI: 10.3390/molecules27062021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 11/16/2022]
Abstract
There is evidence for the involvement of peroxisome proliferator-activated receptors (PPARs) in pain, cognition, and anxiety. However, their role in pain–fear interactions is unknown. The amygdala plays a key role in pain, conditioned fear, and fear-conditioned analgesia (FCA). We investigated the effects of intra-basolateral amygdala (BLA) administration of PPARα, PPARβ/δ, and PPARγ antagonists on nociceptive behaviour, FCA, and conditioned fear in the presence or absence of nociceptive tone. Male Sprague-Dawley (SD) rats received footshock (FC) or no footshock (NFC) in a conditioning arena. Twenty-three and a half hours later, rats received an intraplantar injection of formalin or saline and, 15 min later, intra-BLA microinjections of vehicle, PPARα (GW6471) PPARβ/δ (GSK0660), or PPARγ (GW9662) antagonists before arena re-exposure. Pain and fear-related behaviour were assessed, and neurotransmitters/endocannabinoids measured post-mortem. Intra-BLA administration of PPARα or PPARγ antagonists potentiated freezing in the presence of nociceptive tone. Blockade of all PPAR subtypes in the BLA increased freezing and BLA dopamine levels in NFC rats in the absence of nociceptive tone. Administration of intra-BLA PPARα and PPARγ antagonists increased levels of dopamine in the BLA compared with the vehicle-treated counterparts. In conclusion, PPARα and PPARγ in the BLA play a role in the expression or extinction of conditioned fear in the presence or absence of nociceptive tone.
Collapse
|
4
|
Khoei HA, Rahimi-Madiseh M, Dehkordi KA, Mohammadabadi MSM, Mohammadi S, Sadeghian R. Physospermum cornubienseL. alleviates nociceptive and neuropathic pain: Evidences and possible mechanisms. JOURNAL OF ETHNOPHARMACOLOGY 2022:114957. [PMID: 34995691 DOI: 10.1016/j.jep.2021.114957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In Iranian/Persian folkloric medicine, Physospermum cornubiense (Shokaran Baghi in Persian) is used for the treatment of pain and inflammation. OBJECTIVE This modern examination included Swiss mice to investigate the anti-neuropathic and anti-nociceptive effects of Physospermum cornubiense essential oil (PCEO). MATERIALS AND METHODS To determine PCEO 's anti-nociceptive function in formalin-induced paw licking (FML) paradigm, researchers looked at the arginine-nitric oxide and potassium channels pathway in addition to involvements of more specific examples of receptors such as adrenergic, opioid, cannabinoid, peroxisome proliferator-activated (PPA), and transient receptor potential vanilloid. The CVC or cervical spinal cord contusion exemplar has also been used to induce neuropathic pain. RESULTS PCEO (450mg/kg) relative to control mice in the phase_ II of FML exemplar provided strong antinociception (p < 0.001). Furthermore, pre-treatments with arginine, glibenclamide, methylene blue, L-NAME, SNP, GW6471, naloxonazine, and GW9662 (p < 0.05) returned the PCEO antinociceptive response in the FML (inflammatory phase) model. Orally limonene administration significantly diminished (p < 0.001) acute pain in inflammatory phase of FML test. Moreover, the von Frey test indicated that both PCEO and limonene could return neuropathic pain (mechanical allodynia) in CVC mice. CONCLUSION The results obtained from this study, together with literature, give evidence of properties of PCEO for therapy of antinociceptive and neuropathic pain.
Collapse
Affiliation(s)
- Hossein Amini Khoei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Rahimi-Madiseh
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Korosh Ashrafi Dehkordi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | - Saeed Mohammadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Reihaneh Sadeghian
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
5
|
Pharmacological Blockade of PPARα Exacerbates Inflammatory Pain-Related Impairment of Spatial Memory in Rats. Biomedicines 2021; 9:biomedicines9060610. [PMID: 34072060 PMCID: PMC8227714 DOI: 10.3390/biomedicines9060610] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/17/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-dependent transcription factors that exist in three isoforms: PPARα, PPARβ/δ and PPARγ. Studies suggest that the PPAR signalling system may modulate pain, anxiety and cognition. The aim of the present study was to investigate whether endogenous signalling via PPARs differentially modulates innate anxiety responses and mnemonic function in the presence and absence of inflammatory pain. We examined the effects of intraperitoneal administration of GW6471 (PPARα antagonist), GSK0660 (PPARβ/δ antagonist), GW9662 (PPARγ antagonist), and N-palmitoylethanolamide (PEA) on rat behaviour in the elevated plus maze (EPM), open field (OF), light-dark box (LDB), and novel object recognition (NOR) tests in the presence or absence of chronic inflammatory pain. Complete Freund’s Adjuvant (CFA)-injected rats exhibited impaired recognition and spatial mnemonic performance in the NOR test and pharmacological blockade of PPARα further impaired spatial memory in CFA-treated rats. N-oleoylethanolamide (OEA) levels were higher in the dorsal hippocampus in CFA-injected animals compared to their counterparts. The results suggest a modulatory effect of CFA-induced chronic inflammatory pain on cognitive processing, but not on innate anxiety-related responses. Increased OEA-PPARα signalling may act as a compensatory mechanism to preserve spatial memory function following CFA injection.
Collapse
|
6
|
Okine BN, Mc Laughlin G, Gaspar JC, Harhen B, Roche M, Finn DP. Antinociceptive Effects of the GPR55 Antagonist CID16020046 Injected into the Rat Anterior Cingulate Cortex. Neuroscience 2020; 443:19-29. [PMID: 32673629 DOI: 10.1016/j.neuroscience.2020.07.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 07/04/2020] [Accepted: 07/07/2020] [Indexed: 10/23/2022]
Abstract
The G-protein coupled receptor, GPR55, modulates nociceptive processing. Given the expression of GPR55 in the anterior cingulate cortex (ACC), a key brain region involved in the cognitive and affective dimensions of pain, the present study tested the hypothesis that GPR55 signalling in the ACC facilitates inflammatory pain behaviour in rats. The expression of GPR55 in the ACC was confirmed by both western blotting and immunostaining, with evidence for neuronal localisation. Microinjection of the selective GPR55 antagonist CID16020046 into the ACC of adult male Sprague-Dawley rats significantly reduced second phase formalin-evoked nociceptive behaviour compared with vehicle-treated controls. CID16020046 administration was associated with a reduction in phosphorylation of extracellular signal-regulated kinase (ERK), a downstream target of GPR55 activation, in the ACC. Intra-ACC administration of CID16020046 prevented the formalin-induced increases in expression of mRNA coding for the immediate early gene and marker of neuronal activity, c-Fos, in the ipsilateral dorsal horn of the spinal cord. Intra-plantar injection of formalin reduced tissue levels of the endogenous GPR55 ligand 2-arachidonoyl-sn-glycero-3-phosphoinositol (2-AGPI) in the ACC, likely reflecting its increased release/utilisation. These data suggest that endogenous activation of GPR55 signalling and increased ERK phosphorylation in the ACC facilitates inflammatory pain via top-down modulation of descending pain control.
Collapse
Affiliation(s)
- Bright N Okine
- Pharmacology and Therapeutics, NCBES, National University of Ireland, Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland, Galway, University Road, Galway, Ireland
| | - Gemma Mc Laughlin
- Pharmacology and Therapeutics, NCBES, National University of Ireland, Galway, University Road, Galway, Ireland
| | - Jessica C Gaspar
- Pharmacology and Therapeutics, NCBES, National University of Ireland, Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland, Galway, University Road, Galway, Ireland
| | - Brendan Harhen
- Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland, Galway, University Road, Galway, Ireland
| | - Michelle Roche
- Physiology, School of Medicine, NCBES, National University of Ireland, Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland, Galway, University Road, Galway, Ireland
| | - David P Finn
- Pharmacology and Therapeutics, NCBES, National University of Ireland, Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland, Galway, University Road, Galway, Ireland.
| |
Collapse
|
7
|
Thomas A, Okine BN, Finn DP, Masocha W. Peripheral deficiency and antiallodynic effects of 2-arachidonoyl glycerol in a mouse model of paclitaxel-induced neuropathic pain. Biomed Pharmacother 2020; 129:110456. [PMID: 32603895 DOI: 10.1016/j.biopha.2020.110456] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/16/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Modulation of the endocannabinoid system has been shown to alleviate neuropathic pain. The aim of this study was to evaluate if treatment with paclitaxel, a chemotherapeutic agent that induces neuropathic pain, affects endocannabinoid levels at a time when mice develop paclitaxel-induced mechanical allodynia. We also evaluated the peripheral antiallodynic activity of the endocannabinoid 2-arachidonoyl glycerol (2-AG) and an inhibitor of monoacylglycerol lipase (MAGL), an enzyme responsible for 2-AG hydrolysis. METHODS Female BALB/c mice were treated intraperitoneally with paclitaxel to induce mechanical allodynia. Levels of the endocannabinoids, N-arachidonoylethanolamine (anandamide, AEA), 2-AG, and the N-acylethanolamines (NAEs), N-palmitoylethanolamide (PEA) and N-oleoylethanolamide (OEA), which are structurally-related to AEA, in the brain, spinal cord and paw skin were measured using LC-MS/MS. Protein expression of MAGL in the paw skin was measured using Wes™. The effects of subcutaneous (s.c.) injection of 2-AG and JZL184 (a MAGL inhibitor) into the right hind paw of mice with paclitaxel-induced mechanical allodynia were assessed using the dynamic plantar aesthesiometer. The effects of pretreatment, s.c., into the right hind paw, with cannabinoid type 1 (CB1) receptor antagonist AM251 and CB2 receptor antagonist AM630 on the antiallodynic effects of 2-AG were also evaluated. RESULTS The levels of 2-AG were reduced only in the paw skin of paclitaxel-treated mice, whilst the levels of AEA, PEA and OEA were not significantly altered. There was no change in the expression of MAGL in the paw skin. Administration of 2-AG and JZL184 produced antiallodynic effects against paclitaxel-induced mechanical allodynia in the injected right paw, but did not affect the uninjected left paw. The antiallodynic activity of 2-AG was antagonized by both AM251 and AM630. CONCLUSION These results indicate that during paclitaxel-induced mechanical allodynia there is a deficiency of 2-AG in the periphery, but not in the CNS. Increasing 2-AG in the paw by local administration of 2-AG or a MAGL inhibitor, alleviates mechanical allodynia in a CB1 and CB2 receptor-dependent manner.
Collapse
Affiliation(s)
- Amal Thomas
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Kuwait University, Kuwait
| | - Bright N Okine
- Pharmacology and Therapeutics, School of Medicine, NCBES Centre for Pain Research and Galway Neuroscience Centre, National University of Ireland Galway, University Road, Galway, Ireland
| | - David P Finn
- Pharmacology and Therapeutics, School of Medicine, NCBES Centre for Pain Research and Galway Neuroscience Centre, National University of Ireland Galway, University Road, Galway, Ireland
| | - Willias Masocha
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Kuwait University, Kuwait.
| |
Collapse
|
8
|
Gaspar JC, Okine BN, Llorente-Berzal A, Roche M, Finn DP. Pharmacological Blockade of PPAR Isoforms Increases Conditioned Fear Responding in the Presence of Nociceptive Tone. Molecules 2020; 25:molecules25041007. [PMID: 32102354 PMCID: PMC7070536 DOI: 10.3390/molecules25041007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 12/29/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors with three isoforms (PPARα, PPARβ/δ, PPARγ) and can regulate pain, anxiety, and cognition. However, their role in conditioned fear and pain-fear interactions has not yet been investigated. Here, we investigated the effects of systemically administered PPAR antagonists on formalin-evoked nociceptive behaviour, fear-conditioned analgesia (FCA), and conditioned fear in the presence of nociceptive tone in rats. Twenty-three and a half hours following fear conditioning to context, male Sprague-Dawley rats received an intraplantar injection of formalin and intraperitoneal administration of vehicle, PPARα (GW6471), PPARβ/δ (GSK0660) or PPARγ (GW9662) antagonists, and 30 min later were re-exposed to the conditioning arena for 15 min. The PPAR antagonists did not alter nociceptive behaviour or fear-conditioned analgesia. The PPARα and PPARβ/δ antagonists prolonged context-induced freezing in the presence of nociceptive tone without affecting its initial expression. The PPARγ antagonist potentiated freezing over the entire trial. In conclusion, pharmacological blockade of PPARα and PPARβ/δ in the presence of formalin-evoked nociceptive tone, impaired short-term, within-trial fear-extinction in rats without affecting pain response, while blockade of PPARγ potentiated conditioned fear responding. These results suggest that endogenous signalling through these three PPAR isoforms may reduce the expression of conditioned fear in the presence of nociceptive tone.
Collapse
Affiliation(s)
- Jessica C. Gaspar
- Pharmacology and Therapeutics Department, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland; (J.C.G.); (B.N.O.); (A.L.-B.)
- Galway Neuroscience Centre, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland;
- Centre for Pain Research, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland
| | - Bright N. Okine
- Pharmacology and Therapeutics Department, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland; (J.C.G.); (B.N.O.); (A.L.-B.)
- Galway Neuroscience Centre, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland;
- Centre for Pain Research, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland
| | - Alvaro Llorente-Berzal
- Pharmacology and Therapeutics Department, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland; (J.C.G.); (B.N.O.); (A.L.-B.)
- Galway Neuroscience Centre, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland;
- Centre for Pain Research, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland
| | - Michelle Roche
- Galway Neuroscience Centre, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland;
- Centre for Pain Research, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland
- Physiology Department, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland
| | - David P. Finn
- Pharmacology and Therapeutics Department, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland; (J.C.G.); (B.N.O.); (A.L.-B.)
- Galway Neuroscience Centre, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland;
- Physiology Department, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland
- Correspondence: ; Tel.: +353-(0)91-495-280; Fax: +353-(0)91-495-586
| |
Collapse
|
9
|
Yu X, Abdul M, Fan BQ, Zhang L, Lin X, Wu Y, Fu H, Lin Q, Meng H. The release of exosomes in the medial prefrontal cortex and nucleus accumbens brain regions of chronic constriction injury (CCI) model mice could elevate the pain sensation. Neurosci Lett 2020; 723:134774. [PMID: 31981720 DOI: 10.1016/j.neulet.2020.134774] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/17/2020] [Accepted: 01/19/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Brain function relies on the capacity of neurons to locally modulate each other at the level of synapses. Therefore, the exosomal pathway may constitute a well-designed mechanism for local and systemic interneuronal transfer of information within functional brain networks. Exosomes bind to and are endocytosed by neurons of different brain regions to play a definite role. The medial prefrontal cortex (mPFC) and nucleus accumbens (NAc) brain regions are known to involve in pain modulation. Our study observes the roles of exosomal activity in these two dominant regions of the pain-related pathway, and there influence on the analgesic effects in CCI mice. METHODS We induced pain exosomes in the mPFC and NAc in the mice of chronic constriction injury of the sciatic nerve model to produce neuropathic pain, and assessed changes that might affect analgesic behaviors. These changes were measured through a combination of behavioral, surgical, and other cellular testings. RESULTS Our study found that pain expression was elevated in mice given exogenous exosomes isolated from CCI mice, especially at the 2 h and 4 h time interval, in mice given exosomes at the mPFC and NAc, respectively. We also found that inhibiting formation of pain exosomes through GW4869 within the mPFC and NAc can elevate the pain threshold. CONCLUSION Results from our study supported the idea that the release of mPFC and NAc exosomes of CCI model has elevated the pain sensations in the subjected mice. This study will further help in designing new clinical trials, and will revolutionize the drug-induced anesthetic responses.
Collapse
Affiliation(s)
- Xiaolu Yu
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| | - Mannan Abdul
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| | - Bing-Qian Fan
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| | - Lilu Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| | - Xing Lin
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| | - Yan Wu
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| | - Hui Fu
- Department of Neurology, Zibo Municipal First Hospital, Zibo, Shandong 255200, China.
| | - Qisi Lin
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Hao Meng
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China.
| |
Collapse
|
10
|
Okine BN, Gaspar JC, Finn DP. PPARs and pain. Br J Pharmacol 2018; 176:1421-1442. [PMID: 29679493 DOI: 10.1111/bph.14339] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/19/2018] [Accepted: 03/26/2018] [Indexed: 02/06/2023] Open
Abstract
Chronic pain is a common cause of disability worldwide and remains a global health and socio-economic challenge. Current analgesics are either ineffective in a significant proportion of patients with chronic pain or associated with significant adverse side effects. The PPARs, a family of nuclear hormone transcription factors, have emerged as important modulators of pain in preclinical studies and therefore a potential therapeutic target for the treatment of pain. Modulation of nociceptive processing by PPARs is likely to involve both transcription-dependent and transcription-independent mechanisms. This review presents a comprehensive overview of preclinical studies investigating the contribution of PPAR signalling to nociceptive processing in animal models of inflammatory and neuropathic pain. We examine current evidence from anatomical, molecular and pharmacological studies demonstrating a role for PPARs in pain control. We also discuss the limited evidence available from relevant clinical studies and identify areas that warrant further research. LINKED ARTICLES: This article is part of a themed section on 8th European Workshop on Cannabinoid Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.10/issuetoc.
Collapse
Affiliation(s)
- Bright N Okine
- Pharmacology and Therapeutics, National University of Ireland Galway, Galway, Ireland.,Galway Neuroscience Centre, National University of Ireland Galway, Galway, Ireland.,Centre for Pain Research, NCBES, National University of Ireland Galway, Galway, Ireland
| | - Jessica C Gaspar
- Pharmacology and Therapeutics, National University of Ireland Galway, Galway, Ireland.,Galway Neuroscience Centre, National University of Ireland Galway, Galway, Ireland.,Centre for Pain Research, NCBES, National University of Ireland Galway, Galway, Ireland
| | - David P Finn
- Pharmacology and Therapeutics, National University of Ireland Galway, Galway, Ireland.,Galway Neuroscience Centre, National University of Ireland Galway, Galway, Ireland.,Centre for Pain Research, NCBES, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
11
|
N-palmitoylethanolamide in the anterior cingulate cortex attenuates inflammatory pain behaviour indirectly via a CB1 receptor-mediated mechanism. Pain 2017; 157:2687-2696. [PMID: 27649266 DOI: 10.1097/j.pain.0000000000000687] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The neural substrates and mechanisms mediating the antinociceptive effects of the endogenous bioactive lipid, N-palmitoylethanolamide (PEA), require further investigation. We investigated the effects of exogenous PEA administration into the anterior cingulate cortex (ACC), an important brain region linked with cognitive and affective modulation of pain, on formalin-evoked nociceptive behaviour in rats. Potential involvement of peroxisome proliferator-activated receptor isoforms (PPAR) α and γ or endocannabinoid-mediated entourage effects at cannabinoid1 (CB1) receptors or transient receptor potential subfamily V member 1 (TRPV1) in mediating the effects of PEA was also investigated. Intra-ACC administration of PEA significantly attenuated the first and early second phases of formalin-evoked nociceptive behaviour. This effect was attenuated by the CB1 receptor antagonist AM251, but not by the PPARα antagonist GW6471, the PPARγ antagonist GW9662, or the TRPV1 antagonist 5'-iodo resiniferatoxin. All antagonists, administered alone, significantly reduced formalin-evoked nociceptive behaviour, suggesting facilitatory/permissive roles for these receptors in the ACC in inflammatory pain. Post-mortem tissue analysis revealed a strong trend for increased levels of the endocannabinoid anandamide in the ACC of rats that received intra-ACC PEA. Expression of c-Fos, a marker of neuronal activity, was significantly reduced in the basolateral nucleus of the amygdala, but not in the central nucleus of the amygdala, the rostral ventromedial medulla or the dorsal horn of the spinal cord. In conclusion, these data indicate that PEA in the ACC can reduce inflammatory pain-related behaviour, possibly via AEA-induced activation of CB1 receptors and associated modulation of neuronal activity in the basolateral amygdala.
Collapse
|
12
|
Peng X, Studholme K, Kanjiya MP, Luk J, Bogdan D, Elmes MW, Carbonetti G, Tong S, Gary Teng YH, Rizzo RC, Li H, Deutsch DG, Ojima I, Rebecchi MJ, Puopolo M, Kaczocha M. Fatty-acid-binding protein inhibition produces analgesic effects through peripheral and central mechanisms. Mol Pain 2017; 13:1744806917697007. [PMID: 28326944 PMCID: PMC5407663 DOI: 10.1177/1744806917697007] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Background Fatty-acid-binding proteins (FABPs) are intracellular carriers for endocannabinoids, N-acylethanolamines, and related lipids. Previous work indicates that systemically administered FABP5 inhibitors produce analgesia in models of inflammatory pain. It is currently not known whether FABP inhibitors exert their effects through peripheral or central mechanisms. Here, we examined FABP5 distribution in dorsal root ganglia and spinal cord and examined the analgesic effects of peripherally and centrally administered FABP5 inhibitors. Results Immunofluorescence revealed robust expression of FABP5 in lumbar dorsal root ganglia. FABP5 was distributed in peptidergic calcitonin gene-related peptide-expressing dorsal root ganglia and non-peptidergic isolectin B4-expressing dorsal root ganglia. In addition, the majority of dorsal root ganglia expressing FABP5 also expressed transient receptor potential vanilloid 1 (TRPV1) and peripherin, a marker of nociceptive fibers. Intraplantar administration of FABP5 inhibitors reduced thermal and mechanical hyperalgesia in the complete Freund’s adjuvant model of chronic inflammatory pain. In contrast to its robust expression in dorsal root ganglia, FABP5 was sparsely distributed in the lumbar spinal cord and intrathecal administration of FABP inhibitor did not confer analgesic effects. Administration of FABP inhibitor via the intracerebroventricular (i.c.v.) route reduced thermal hyperalgesia. Antagonists of peroxisome proliferator-activated receptor alpha blocked the analgesic effects of peripherally and i.c.v. administered FABP inhibitor while antagonism of cannabinoid receptor 1 blocked the effects of peripheral FABP inhibition and a TRPV1 antagonist blocked the effects of i.c.v. administered inhibitor. Although FABP5 and TRPV1 were co-expressed in the periaqueductal gray region of the brain, which is known to modulate pain, knockdown of FABP5 in the periaqueductal gray using adeno-associated viruses and pharmacological FABP5 inhibition did not produce analgesic effects. Conclusions This study demonstrates that FABP5 is highly expressed in nociceptive dorsal root ganglia neurons and FABP inhibitors exert peripheral and supraspinal analgesic effects. This indicates that peripherally restricted FABP inhibitors may serve as a new class of analgesic and anti-inflammatory agents.
Collapse
Affiliation(s)
- Xiaoxue Peng
- 1 Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Keith Studholme
- 1 Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Martha P Kanjiya
- 1 Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Jennifer Luk
- 1 Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Diane Bogdan
- 1 Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Matthew W Elmes
- 2 Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Gregory Carbonetti
- 2 Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Simon Tong
- 3 Department of Chemistry, Stony Brook University, Stony Brook, NY, USA.,4 Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| | - Yu-Han Gary Teng
- 3 Department of Chemistry, Stony Brook University, Stony Brook, NY, USA.,4 Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| | - Robert C Rizzo
- 4 Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA.,5 Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA
| | - Huilin Li
- 2 Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA.,4 Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| | - Dale G Deutsch
- 2 Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA.,4 Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| | - Iwao Ojima
- 3 Department of Chemistry, Stony Brook University, Stony Brook, NY, USA.,4 Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| | - Mario J Rebecchi
- 1 Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Michelino Puopolo
- 1 Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Martin Kaczocha
- 1 Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA.,2 Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA.,4 Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
13
|
Starowicz K, Finn DP. Cannabinoids and Pain: Sites and Mechanisms of Action. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 80:437-475. [PMID: 28826543 DOI: 10.1016/bs.apha.2017.05.003] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The endocannabinoid system, consisting of the cannabinoid1 receptor (CB1R) and cannabinoid2 receptor (CB2R), endogenous cannabinoid ligands (endocannabinoids), and metabolizing enzymes, is present throughout the pain pathways. Endocannabinoids, phytocannabinoids, and synthetic cannabinoid receptor agonists have antinociceptive effects in animal models of acute, inflammatory, and neuropathic pain. CB1R and CB2R located at peripheral, spinal, or supraspinal sites are important targets mediating these antinociceptive effects. The mechanisms underlying the analgesic effects of cannabinoids likely include inhibition of presynaptic neurotransmitter and neuropeptide release, modulation of postsynaptic neuronal excitability, activation of the descending inhibitory pain pathway, and reductions in neuroinflammatory signaling. Strategies to dissociate the psychoactive effects of cannabinoids from their analgesic effects have focused on peripherally restricted CB1R agonists, CB2R agonists, inhibitors of endocannabinoid catabolism or uptake, and modulation of other non-CB1R/non-CB2R targets of cannabinoids including TRPV1, GPR55, and PPARs. The large body of preclinical evidence in support of cannabinoids as potential analgesic agents is supported by clinical studies demonstrating their efficacy across a variety of pain disorders.
Collapse
Affiliation(s)
- Katarzyna Starowicz
- Institute of Pharmacology, Polish Academy of Sciences, Laboratory of Pain Pathophysiology, Krakow, Poland
| | - David P Finn
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland, Galway, Ireland.
| |
Collapse
|
14
|
Okine BN, Gaspar JC, Madasu MK, Olango WM, Harhen B, Roche M, Finn DP. Characterisation of peroxisome proliferator-activated receptor signalling in the midbrain periaqueductal grey of rats genetically prone to heightened stress, negative affect and hyperalgesia. Brain Res 2016; 1657:185-192. [PMID: 27916440 DOI: 10.1016/j.brainres.2016.11.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/12/2016] [Accepted: 11/19/2016] [Indexed: 10/20/2022]
Abstract
The stress-hyperresponsive Wistar-Kyoto (WKY) rat strain exhibits a hyperalgesic phenotype and is a useful genetic model for studying stress-pain interactions. Peroxisome proliferator-activated receptor (PPAR) signalling in the midbrain periaqueductal grey (PAG) modulates pain. This study characterised PPAR signalling in the PAG of WKY rats exposed to the formalin test of inflammatory pain, versus Sprague-Dawley (SD) controls. Formalin injection reduced levels of the endogenous PPAR ligands N-palmitoylethanolamide (PEA) and N-oleoylethanolamide (OEA) in the lateral(l) PAG of SD rats, but not WKY rats which exhibited higher levels of these analytes compared with formalin-injected SD counterparts. Levels of mRNA coding for fatty acid amide hydrolase (FAAH; catabolises PEA and OEA) were lower in the lPAG of WKY versus SD rats. PPARγ mRNA and protein levels in the lPAG were higher in saline-treated WKY rats, with PPARγ protein levels reduced by formalin treatment in WKY rats only. In the dorsolateral(dl) or ventrolateral(vl) PAG, there were no effects of formalin injection on PEA or OEA levels but there were some differences in levels of these analytes between saline-treated WKY and SD rats and some formalin-evoked alterations in levels of PPARα, PPARγ or FAAH mRNA in WKY and/or SD rats. Pharmacological blockade of PPARγ in the lPAG enhanced formalin-evoked nociceptive behaviour in WKY, but not SD, rats. These data indicate differences in the PPAR signalling system in the PAG of WKY versus SD rats and suggest that enhanced PEA/OEA-mediated tone at PPARγ in the lPAG may represent an adaptive mechanism to lower hyperalgesia in WKY rats.
Collapse
Affiliation(s)
- Bright N Okine
- Pharmacology and Therapeutics, Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland
| | - Jessica C Gaspar
- Pharmacology and Therapeutics, Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland
| | - Manish K Madasu
- Pharmacology and Therapeutics, Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland
| | - Weredeselam M Olango
- Pharmacology and Therapeutics, Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland
| | - Brendan Harhen
- Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland
| | - Michelle Roche
- Physiology, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland
| | - David P Finn
- Pharmacology and Therapeutics, Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland.
| |
Collapse
|
15
|
Donvito G, Wilkerson JL, Damaj MI, Lichtman AH. Palmitoylethanolamide Reverses Paclitaxel-Induced Allodynia in Mice. J Pharmacol Exp Ther 2016; 359:310-318. [PMID: 27608657 PMCID: PMC5074488 DOI: 10.1124/jpet.116.236182] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 09/07/2016] [Indexed: 12/29/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) represents a serious complication associated with antineoplastic drugs. Although there are no medications available that effectively prevent CIPN, many classes of drugs have been used to treat this condition, including anticonvulsants, serotonin and noradrenaline reuptake inhibitors, and opioids. However, these therapeutic options yielded inconclusive results in CIPN clinical trials and produced assorted side effects with their prolonged use. Thus, there is an urgent need to develop efficacious and safe treatments for CIPN. In this report, we tested whether the endogenous lipid palmitoylethanolamide (PEA) alone or in combination with the anticonvulsant gabapentin would reduce allodynia in a mouse paclitaxel model of CIPN. Gabapentin and PEA reversed paclitaxel-induced allodynia with respective ED50 doses (95% confidence interval) of 67.4 (61.52-73.94) and 9.2 (8.39-10.16) mg/kg. Isobolographic analysis of these drugs in combination revealed synergistic antiallodynic effects. The PPAR-α antagonist receptor antagonist GW6471 [N-((2S)-2-(((1Z)-1-methyl-3-oxo-3-(4-(trifluoromethyl)phenyl)prop-1-enyl)amino)-3-(4-(2-(5-methyl-2-phenyl-1,3-oxazol-4-yl)ethoxy)phenyl)propyl)propanamide] completely blocked the antinociceptive effects of PEA. In addition, PEA administered via intraplantar injection into a paw, intrathecal injection, and intracerebroventricular injection reversed paclitaxel-induced allodynia, suggesting that it may act at multiple sites in the neuroaxis and periphery. Finally, repeated administration of PEA (30 mg/kg, 7 days) preserved the antiallodynic effects with no evidence of tolerance. These findings taken together suggest that PEA possesses potential to treat peripheral neuropathy in cancer patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Giulia Donvito
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia (G.D.; J.L.W.; M.I.D.; A.H.L.); and Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy (G.D.)
| | - Jenny L Wilkerson
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia (G.D.; J.L.W.; M.I.D.; A.H.L.); and Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy (G.D.)
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia (G.D.; J.L.W.; M.I.D.; A.H.L.); and Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy (G.D.)
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia (G.D.; J.L.W.; M.I.D.; A.H.L.); and Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy (G.D.)
| |
Collapse
|
16
|
Geng KW, He T, Wang RR, Li CL, Luo WJ, Wu FF, Wang Y, Li Z, Lu YF, Guan SM, Chen J. Ethanol Increases Mechanical Pain Sensitivity in Rats via Activation of GABAA Receptors in Medial Prefrontal Cortex. Neurosci Bull 2016; 32:433-44. [PMID: 27628528 DOI: 10.1007/s12264-016-0063-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/14/2016] [Indexed: 12/23/2022] Open
Abstract
Ethanol is widely known for its ability to cause dramatic changes in emotion, social cognition, and behavior following systemic administration in humans. Human neuroimaging studies suggest that alcohol dependence and chronic pain may share common mechanisms through amygdala-medial prefrontal cortex (mPFC) interactions. However, whether acute administration of ethanol in the mPFC can modulate pain perception is unknown. Here we showed that bilateral microinjections of ethanol into the prelimbic and infralimbic areas of the mPFC lowered the bilateral mechanical pain threshold for 48 h without influencing thermal pain sensitivity in adult rats. However, bilateral microinjections of artificial cerebrospinal fluid into the mPFC or bilateral microinjections of ethanol into the dorsolateral PFC (also termed as motor cortex area 1 in Paxinos and Watson's atlas of The Rat Brain. Elsevier Academic Press, Amsterdam, 2005) failed to do so, suggesting regional selectivity of the effects of ethanol. Moreover, bilateral microinjections of ethanol did not change the expression of either pro-apoptotic (caspase-3 and Bax) or anti-apoptotic (Bcl-2) proteins, suggesting that the dose was safe and validating the method used in the current study. To determine whether γ-aminobutyric acid A (GABAA) receptors are involved in mediating the ethanol effects, muscimol, a selective GABAA receptor agonist, or bicuculline, a selective GABAA receptor antagonist, was administered alone or co-administered with ethanol through the same route into the bilateral mPFC. The results showed that muscimol mimicked the effects of ethanol while bicuculline completely reversed the effects of ethanol and muscimol. In conclusion, ethanol increases mechanical pain sensitivity through activation of GABAA receptors in the mPFC of rats.
Collapse
Affiliation(s)
- Kai-Wen Geng
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Ting He
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Rui-Rui Wang
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Chun-Li Li
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Wen-Jun Luo
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Fang-Fang Wu
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Yan Wang
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Zhen Li
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Yun-Fei Lu
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Su-Min Guan
- School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Jun Chen
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China.
- Beijing Institute for Brain Disorders, Beijing, 100069, China.
| |
Collapse
|
17
|
Okine BN, Spicer C, Millns P, Bennett A, Chapman V. Systemic administration of WY-14643, a selective synthetic agonist of peroxisome proliferator activator receptor-alpha, alters spinal neuronal firing in a rodent model of neuropathic pain. Scand J Pain 2015; 9:42-48. [DOI: 10.1016/j.sjpain.2015.06.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 06/14/2015] [Indexed: 12/21/2022]
Abstract
Abstract
Background and aims
The clinical management of chronic neuropathic pain remains a global health challenge. Current treatments are either ineffective, or associated with unwanted side-effects. The development of effective, safe therapies requires the identification of novel therapeutic targets using clinically relevant animal models of neuropathic pain.
Peroxisome proliferator activated receptor alpha (PPARα), is a member of the nuclear hormone family of transcription factors, which is widely distributed in the peripheral and central nervous systems. Pharmacological studies report antinociceptive effects of PPARα agonists following systemic administration in rodent models of neuropathic pain, however the neuronal mechanisms and sites of action mediating these effects are unclear.
The aim of this study was to investigate the effects of systemic administration of the synthetic PPARα agonist, WY-14643 on mechanically-evoked responses of spinal cord dorsal horn wide dynamic range (WDR) neurones in the spinal nerve ligated (SNL) model of neuropathic pain in rats. In addition, comparative molecular analysis of mRNA coding for PPARα and PPARα protein expression in the spinal cord of sham-operated and neuropathic rats was performed.
Methods
Lumbar L5–L6 spinal nerve ligation was performed in male Sprague–Dawley rats (110–130 g) under isoflurane anaesthesia. Sham controls underwent similar surgical conditions, but without ligation of the L5–L6 spinal nerves. Hindpaw withdrawal thresholds were measured on the day of surgery -day 0, and on days- 2, 4, 7, 10 and 14 post-surgery. At day 14 extracellular single-unit recordings of spinal (WDR) dorsal horn neurons were performed in both sham and SNL neuropathic rats under anaesthesia. The effects of intraperitoneal (i.p.) administration of WY-14643 (15 and 30 mg/kg) or vehicle on evoked responses of WDR neurons to punctate mechanical stimulation of the peripheral receptive field of varying bending force (8–60 g) were recorded. In a separate cohort of SNL and sham neuropathic rats, the expression of mRNA coding for PPARα and protein expression in the ipsilateral and contralateral spinal cord was determined using quantitative real time polymerase chain reaction (qRT-PCR) and western blotting techniques respectively.
Results
WY-14643 (15 and 30mg/kg i.p.) rapidly attenuated mechanically evoked (8, 10 and 15g) responses of spinal WDR neurones in SNL, but not sham-operated rats. Molecular analysis revealed significantly increased PPARα protein, but not mRNA, expression in the ipsilateral spinal cord of SNL, compared to the contralateral side in SNL rats. There were no changes in PPARα mRNA or protein expression in the sham controls.
Conclusion
The observation that levels of PPARα protein were increased in ipsilateral spinal cord of neuropathic rats supports a contribution of spinal sites of action mediating the effects of systemic WY-14643. Our data suggests that the inhibitory effects of a PPARα agonist on spinal neuronal responses may account, at least in part, for their analgesic effects of in neuropathic pain.
Implication
Selective activation of PPARα in the spinal cord may be therapeutically relevant for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Bright N. Okine
- School of Life Sciences , University of Nottingham Medical School , Queens Medical Centre, NG7 2UH , Nottingham , UK
- Pharmacology and Therapeutics, School of Medicine , National University of Ireland Galway , University Road , Galway , Ireland
| | - Clare Spicer
- School of Life Sciences , University of Nottingham Medical School , Queens Medical Centre, NG7 2UH , Nottingham , UK
| | - Paul Millns
- School of Life Sciences , University of Nottingham Medical School , Queens Medical Centre, NG7 2UH , Nottingham , UK
| | - Andrew Bennett
- School of Life Sciences , University of Nottingham Medical School , Queens Medical Centre, NG7 2UH , Nottingham , UK
| | - Victoria Chapman
- School of Life Sciences , University of Nottingham Medical School , Queens Medical Centre, NG7 2UH , Nottingham , UK
| |
Collapse
|
18
|
Capasso R, Orlando P, Pagano E, Aveta T, Buono L, Borrelli F, Di Marzo V, Izzo AA. Palmitoylethanolamide normalizes intestinal motility in a model of post-inflammatory accelerated transit: involvement of CB₁ receptors and TRPV1 channels. Br J Pharmacol 2015; 171:4026-37. [PMID: 24818658 DOI: 10.1111/bph.12759] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 04/15/2014] [Accepted: 04/23/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Palmitoylethanolamide (PEA), a naturally occurring acylethanolamide chemically related to the endocannabinoid anandamide, interacts with targets that have been identified in peripheral nerves controlling gastrointestinal motility, such as cannabinoid CB1 and CB2 receptors, TRPV1 channels and PPARα. Here, we investigated the effect of PEA in a mouse model of functional accelerated transit which persists after the resolution of colonic inflammation (post-inflammatory irritable bowel syndrome). EXPERIMENTAL APPROACH Intestinal inflammation was induced by intracolonic administration of oil of mustard (OM). Mice were tested for motility and biochemical and molecular biology changes 4 weeks later. PEA, oleoylethanolamide and endocannabinoid levels were measured by liquid chromatography-mass spectrometry and receptor and enzyme mRNA expression by qRT-PCR. KEY RESULTS OM induced transient colitis and a functional post-inflammatory increase in upper gastrointestinal transit, associated with increased intestinal anandamide (but not 2-arachidonoylglycerol, PEA or oleoylethanolamide) levels and down-regulation of mRNA for TRPV1 channels. Exogenous PEA inhibited the OM-induced increase in transit and tended to increase anandamide levels. Palmitic acid had a weaker effect on transit. Inhibition of transit by PEA was blocked by rimonabant (CB1 receptor antagonist), further increased by 5'-iodoresiniferatoxin (TRPV1 antagonist) and not significantly modified by the PPARα antagonist GW6471. CONCLUSIONS AND IMPLICATIONS Intestinal endocannabinoids and TRPV1 channel were dysregulated in a functional model of accelerated transit exhibiting aspects of post-inflammatory irritable bowel syndrome. PEA counteracted the accelerated transit, the effect being mediated by CB1 receptors (possibly via increased anandamide levels) and modulated by TRPV1 channels.
Collapse
Affiliation(s)
- Raffaele Capasso
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Differential upregulation of the cannabinoid CB₂ receptor in neurotoxic and inflammation-driven rat models of Parkinson's disease. Exp Neurol 2015; 269:133-41. [PMID: 25895887 DOI: 10.1016/j.expneurol.2015.04.007] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 04/08/2015] [Accepted: 04/11/2015] [Indexed: 11/24/2022]
Abstract
The cannabinoid CB2 receptor has recently emerged as a potential anti-inflammatory target to break the self-sustaining cycle of neuroinflammation and neurodegeneration that is associated with neurodegenerative diseases. However, in order to facilitate the development of cannabinoid drugs for neurodegenerative disease, the changes that occur in the endocannabinoid system in response to different neurodegenerative triggers needs to be elucidated. Therefore, the aim of this study was to investigate and compare the changes that occur in the endocannabinoid system in neurotoxic and inflammation-driven models of Parkinson's disease. To do so, male Sprague Dawley rats were given unilateral, intra-striatal injections of the dopaminergic neurotoxin, 6-hydroxydopamine, or the bacterial inflammagen, lipopolysaccharide (LPS). Animals underwent behavioural testing for motor dysfunction on Days 7, 14 and 28 post-surgery, and were sacrificed on Days 1, 4, 14 and 28. Changes in the endocannabinoid system were investigated by qRT-PCR, liquid chromatography-mass spectrometry and immunohistochemistry. After injection of 6-hydroxydopamine or LPS into the rat striatum, we found that expression of the CB2 receptor was significantly elevated in both models, and that this increase correlated significantly with an increase in microglial activation. Interestingly, the increase in CB2 receptor expression in the inflammation-driven model was significantly more pronounced than that in the neurotoxic model. Moreover, endocannabinoid levels were also elevated in the LPS model but not the 6-hydroxydopamine model. Thus, this study has shown that the endocannabinoid system is dysregulated in animal models of Parkinson's disease, and has also revealed significant differences in the level of dysregulation between the models themselves. This study indicates that targeting the CB2 receptor may represent a viable target for anti-inflammatory disease modification in Parkinson's disease.
Collapse
|
20
|
Corcoran L, Roche M, Finn DP. The Role of the Brain's Endocannabinoid System in Pain and Its Modulation by Stress. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 125:203-55. [DOI: 10.1016/bs.irn.2015.10.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
21
|
Palmitoylethanolamide in CNS health and disease. Pharmacol Res 2014; 86:32-41. [PMID: 24844438 DOI: 10.1016/j.phrs.2014.05.006] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 05/07/2014] [Accepted: 05/08/2014] [Indexed: 01/08/2023]
Abstract
The existence of acylethanolamides (AEs) in the mammalian brain has been known for decades. Among AEs, palmitoylethanolamide (PEA) is abundant in the central nervous system (CNS) and conspicuously produced by neurons and glial cells. Antihyperalgesic and neuroprotective properties of PEA have been mainly related to the reduction of neuronal firing and to control of inflammation. Growing evidence suggest that PEA may be neuroprotective during CNS neurodegenerative diseases. Advances in the understanding of the physiology and pharmacology of PEA have potentiated its interest as useful biological tool for disease management. Several rapid non-genomic and delayed genomic mechanisms of action have been identified for PEA as peroxisome proliferator-activated receptor (PPAR)-α dependent. First, an early molecular control, through Ca(+2)-activated intermediate- and/or big-conductance K(+) channels opening, drives to rapid neuronal hyperpolarization. This is reinforced by the increase of the inward Cl(-) currents due to the modulation of the gamma aminobutyric acid A receptor and by the desensitization of the transient receptor potential channel type V1. Moreover, the gene transcription-mediated mechanism sustains the long-term anti-inflammatory effects, by reducing pro-inflammatory enzyme expression and increasing neurosteroid synthesis. Overall, the integration of these different modes of action allows PEA to exert an immediate and prolonged efficacious control in neuron signaling either on inflammatory process or neuronal excitability, maintaining cellular homeostasis. In this review, we will discuss the effect of PEA on metabolism, behavior, inflammation and pain perception, related to the control of central functions and the emerging evidence demonstrating its therapeutic efficacy in several neurodegenerative diseases.
Collapse
|
22
|
Downer EJ, Finn DP. Cannabinoids: clearing the smoke on pain, inflammation and neurodegeneration. Br J Pharmacol 2014; 171:1341-4. [DOI: 10.1111/bph.12642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Affiliation(s)
- E J Downer
- Department of Anatomy and Neuroscience; Western Gateway Building; University College Cork; Cork Ireland
| | - D P Finn
- Pharmacology and Therapeutics; School of Medicine; Galway Neuroscience Centre and Centre for Pain Research; NCBES; National University of Ireland Galway; University Road Galway Ireland
| |
Collapse
|