1
|
Grabon W, Rheims S, Smith J, Bodennec J, Belmeguenai A, Bezin L. CB2 receptor in the CNS: from immune and neuronal modulation to behavior. Neurosci Biobehav Rev 2023; 150:105226. [PMID: 37164044 DOI: 10.1016/j.neubiorev.2023.105226] [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: 12/30/2022] [Revised: 03/20/2023] [Accepted: 05/06/2023] [Indexed: 05/12/2023]
Abstract
Despite low levels of cannabinoid receptor type 2 (CB2R) expression in the central nervous system in human and rodents, a growing body of evidence shows CB2R involvement in many processes at the behavioral level, through both immune and neuronal modulations. Recent in vitro and in vivo evidence have highlighted the complex role of CB2R under physiological and inflammatory conditions. Under neuroinflammatory states, its activation seems to protect the brain and its functions, making it a promising target in a wide range of neurological disorders. Here, we provide a complete and updated overview of CB2R function in the central nervous system of rodents, spanning from modulation of immune function in microglia but also in other cell types, to behavior and neuronal activity, in both physiological and neuroinflammatory contexts.
Collapse
Affiliation(s)
- Wanda Grabon
- Université Claude Bernard Lyon 1, CNRS, Inserm, Centre de Recherche en Neurosciences de Lyon, U10208 UMR5292, TIGER Team - F-69500 Bron, France; Epilepsy Institute IDEE, 59 boulevard Pinel - F-69500 Bron, France.
| | - Sylvain Rheims
- Université Claude Bernard Lyon 1, CNRS, Inserm, Centre de Recherche en Neurosciences de Lyon, U10208 UMR5292, TIGER Team - F-69500 Bron, France; Epilepsy Institute IDEE, 59 boulevard Pinel - F-69500 Bron, France; Department of Functional Neurology and Epileptology, Hospices Civils de Lyon - France
| | - Jonathon Smith
- Université Claude Bernard Lyon 1, CNRS, Inserm, Centre de Recherche en Neurosciences de Lyon, U10208 UMR5292, TIGER Team - F-69500 Bron, France; Epilepsy Institute IDEE, 59 boulevard Pinel - F-69500 Bron, France
| | - Jacques Bodennec
- Université Claude Bernard Lyon 1, CNRS, Inserm, Centre de Recherche en Neurosciences de Lyon, U10208 UMR5292, TIGER Team - F-69500 Bron, France; Epilepsy Institute IDEE, 59 boulevard Pinel - F-69500 Bron, France
| | - Amor Belmeguenai
- Université Claude Bernard Lyon 1, CNRS, Inserm, Centre de Recherche en Neurosciences de Lyon, U10208 UMR5292, TIGER Team - F-69500 Bron, France; Epilepsy Institute IDEE, 59 boulevard Pinel - F-69500 Bron, France
| | - Laurent Bezin
- Université Claude Bernard Lyon 1, CNRS, Inserm, Centre de Recherche en Neurosciences de Lyon, U10208 UMR5292, TIGER Team - F-69500 Bron, France.
| |
Collapse
|
2
|
CB2R activation ameliorates late adolescent chronic alcohol exposure-induced anxiety-like behaviors during withdrawal by preventing morphological changes and suppressing NLRP3 inflammasome activation in prefrontal cortex microglia in mice. Brain Behav Immun 2023; 110:60-79. [PMID: 36754245 DOI: 10.1016/j.bbi.2023.02.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 01/08/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Chronic alcohol exposure (CAE) during late adolescence increases the risk of anxiety development. Alcohol-induced prefrontal cortex (PFC) microglial activation, characterized by morphological changes and increased associations with neurons, plays a critical role in the pathogenesis of anxiety. Alcohol exposure increases NLRP3 inflammasome expression, increasing cytokine secretion by activated microglia. Cannabinoid type 2 receptor (CB2R), an essential receptor of the endocannabinoid system, regulates microglial activation and neuroinflammatory reactions. We aimed to investigate the role of CB2R activation in ameliorating late adolescent CAE-induced anxiety-like behaviors and microglial activation in C57BL/6J mice. METHODS Six-week-old C57BL/6J mice were acclimated for 7 days and then were administered alcohol by gavage (4 g/kg, 25 % w/v) for 28 days. The mice were intraperitoneally injected with the specific CB2R agonist AM1241 1 h before alcohol treatment. Anxiety-like behaviors during withdrawal were assessed by open field test and elevated plus maze test 24 h after the last alcohol administration. Microglial activation, microglia-neuron interactions, and CB2R and NLRP3 inflammasome-related molecule expression in the PFC were measured using immunofluorescence, immunohistochemical, qPCR, and Western blotting assays. Microglial morphology was evaluated by Sholl analysis and the cell body-to-total cell size index. Additionally, N9 microglia were activated by LPS in vitro, and the effects of AM1241 on NLRP3 and N9 microglial activation were investigated. RESULTS After CAE, mice exhibited severe anxiety-like behaviors during withdrawal. CAE induced obvious microglia-neuron associations, and increased expression of microglial activation markers, CB2R, and NLRP3 inflammasome-related molecules in the PFC. Microglia also showed marked filament retraction and reduction and cell body enlargement after CAE. AM1241 treatment ameliorated anxiety-like behaviors in CAE model mice, and it prevented microglial morphological changes, reduced microglial activation marker expression, and suppressed the microglial NLRP3 inflammasome activation and proinflammatory cytokine secretion induced by CAE. AM1241 suppressed the LPS-induced increase in NLRP3 inflammasome-related molecules, IL-1β release, and M1 phenotype markers (iNOS and CD86) in N9 cell, which was reversed by CB2R antagonist treatment. CONCLUSIONS CAE caused anxiety-like behaviors in late adolescent mice at least partly by inducing microglial activation and increasing microglia-neuron associations in the PFC. CB2R activation ameliorated these effects by preventing morphological changes and suppressing NLRP3 inflammasome activation in PFC microglia.
Collapse
|
3
|
Microglial Cannabinoid CB 2 Receptors in Pain Modulation. Int J Mol Sci 2023; 24:ijms24032348. [PMID: 36768668 PMCID: PMC9917135 DOI: 10.3390/ijms24032348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023] Open
Abstract
Pain, especially chronic pain, can strongly affect patients' quality of life. Cannabinoids ponhave been reported to produce potent analgesic effects in different preclinical pain models, where they primarily function as agonists of Gi/o protein-coupled cannabinoid CB1 and CB2 receptors. The CB1 receptors are abundantly expressed in both the peripheral and central nervous systems. The central activation of CB1 receptors is strongly associated with psychotropic adverse effects, thus largely limiting its therapeutic potential. However, the CB2 receptors are promising targets for pain treatment without psychotropic adverse effects, as they are primarily expressed in immune cells. Additionally, as the resident immune cells in the central nervous system, microglia are increasingly recognized as critical players in chronic pain. Accumulating evidence has demonstrated that the expression of CB2 receptors is significantly increased in activated microglia in the spinal cord, which exerts protective consequences within the surrounding neural circuitry by regulating the activity and function of microglia. In this review, we focused on recent advances in understanding the role of microglial CB2 receptors in spinal nociceptive circuitry, highlighting the mechanism of CB2 receptors in modulating microglia function and its implications for CB2 receptor- selective agonist-mediated analgesia.
Collapse
|
4
|
Zhou Y, Xu Y, Yang J, Yu Z, Wang W, Yuan M, Wang Y, Bai Q, Li Z. Spinal cannabinoid receptor 2 activation alleviates neuropathic pain by regulating microglia and suppressing P2X7 receptor. Front Mol Neurosci 2023; 16:1061220. [PMID: 36969555 PMCID: PMC10030493 DOI: 10.3389/fnmol.2023.1061220] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/13/2023] [Indexed: 03/29/2023] Open
Abstract
Neuropathic pain (NP) is the chronic pain in patients resulting from injuries or diseases in the somatosensory nervous system. However, effective treatment remains limited to opioids. Currently, there is an urgent need to develop new specific pharmaceuticals with low abuse potentiality. Cannabinoid receptor 2 (CB2R) is one of the significant receptors in the endocannabinoid system. It is widely expressed in the central nervous system, especially enriched in glial cells, and plays an important role in the occurrence and development of inflammation in the nervous system. CB2R activation has a neuroprotective effect on nerve injury. In this study, we report increased and more reactive microglia (with larger cell body, shorter processes, and fewer endpoints) observed in the spinal dorsal horn of spared nerve injury (SNI) rats. Continuous intrathecal administration of CB2R agonist PM226 attenuated mechanical and cold hyperalgesia in rats and prevented the transition of microglia to the proinflammatory stage. Thus, microglia transitioned into the neuroprotective stage. Meanwhile, the proinflammatory factors TNF-α and iNOS decreased, and the levels of anti-inflammatory factors Arg-1 and IL-10 increased. The content of P2X7 receptors in the spinal dorsal horn of rats increases with time after SNI. After continuous intrathecal administration of PM226, the content of P2X7 protein decreases significantly. The administration of P2X7 inhibitor A-438079 alleviated the mechanical hyperalgesia of rats, reduced the number of microglia, and decreased the content of P2X7. These results indicate that P2X7 is involved in the neuroprotective effect caused by CB2R activation. In conclusion, this study provides new insights into the neuroprotective mechanism of CB2R activation.
Collapse
Affiliation(s)
- Yifan Zhou
- Department of Anesthesiology and Perioperative Medicine, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yaowei Xu
- Institute of Neuroscience, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jingjie Yang
- Department of Anesthesiology and Perioperative Medicine, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhixiang Yu
- Department of Anesthesiology and Perioperative Medicine, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenting Wang
- Department of Anesthesiology and Perioperative Medicine, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meng Yuan
- Department of Anesthesiology and Perioperative Medicine, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiming Wang
- Department of Anesthesiology and Perioperative Medicine, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qian Bai
- Department of Anesthesiology and Perioperative Medicine, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Qian Bai,
| | - Zhisong Li
- Department of Anesthesiology and Perioperative Medicine, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Zhisong Li,
| |
Collapse
|
5
|
Gonçalves MR, da Conceição MS, Jesus CHA, Gasparin AT, Rosa ES, da Cunha JM. Spinal cannabinoid CB1 or CB2 receptors activation attenuates mechanical allodynia in streptozotocin-induced diabetic rats. Behav Pharmacol 2022; 33:158-164. [PMID: 32804775 DOI: 10.1097/fbp.0000000000000580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Diabetes is a chronic disease associated with a high number of complications such as peripheral neuropathy, which causes sensorial disturbances and may lead to the development of diabetic neuropathic pain (DNP). The current treatment for DNP is just palliative and the drugs may cause severe adverse effects, leading to discontinuation of treatment. Thus, new therapeutic targets need to be urgently investigated. Studies have shown that cannabinoids have promising effects in the treatment of several pathological conditions, including chronic pain. Thus, we aimed to investigate the acute effect of the intrathecal injection of CB1 or CB2 cannabinoid receptor agonists N-(2-chloroethyl)-5Z, 8Z, 11Z, 14Z-eicosatetraenamide (ACEA) or JWH 133, respectively (10, 30 or 100 μg/rat) on the mechanical allodynia associated with experimental diabetes induced by streptozotocin (60 mg/kg; intraperitoneal) in rats. Cannabinoid receptor antagonists CB1 AM251 or CB2 AM630 (1 mg/kg) were given before treatment with respective agonists to confirm the involvement of cannabinoid CB1 or CB2 receptors. Rats with diabetes exhibited a significant reduction on the paw mechanical threshold 2 weeks after diabetes induction, having the maximum effect observed 4 weeks after the streptozotocin injection. This mechanical allodynia was significantly improved by intrathecal treatment with ACEA or JWH 133 (only at the higher dose of 100 μg). Pre-treatment with AM251 or AM630 significantly reverted the anti-allodynic effect of the ACEA or JWH 133, respectively. Considering the clinical challenge that the treatment of DPN represents, this study showed for the first time, that the intrathecal cannabinoid receptors agonists may represent an alternative for the treatment of DNP.
Collapse
Affiliation(s)
- Maryna Rodrigues Gonçalves
- Department of Pharmacology, Biological Science Sector, Federal University of Parana, Curitiba, Parana, Brazil
| | | | | | | | | | | |
Collapse
|
6
|
van den Hoogen NJ, Harding EK, Davidson CED, Trang T. Cannabinoids in Chronic Pain: Therapeutic Potential Through Microglia Modulation. Front Neural Circuits 2022; 15:816747. [PMID: 35069129 PMCID: PMC8777271 DOI: 10.3389/fncir.2021.816747] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/17/2021] [Indexed: 12/16/2022] Open
Abstract
Chronic pain is a complex sensory, cognitive, and emotional experience that imposes a great personal, psychological, and socioeconomic burden on patients. An estimated 1.5 billion people worldwide are afflicted with chronic pain, which is often difficult to treat and may be resistant to the potent pain-relieving effects of opioid analgesics. Attention has therefore focused on advancing new pain therapies directed at the cannabinoid system because of its key role in pain modulation. Endocannabinoids and exogenous cannabinoids exert their actions primarily through Gi/o-protein coupled cannabinoid CB1 and CB2 receptors expressed throughout the nervous system. CB1 receptors are found at key nodes along the pain pathway and their activity gates both the sensory and affective components of pain. CB2 receptors are typically expressed at low levels on microglia, astrocytes, and peripheral immune cells. In chronic pain states, there is a marked increase in CB2 expression which modulates the activity of these central and peripheral immune cells with important consequences for the surrounding pain circuitry. Growing evidence indicate that interventions targeting CB1 or CB2 receptors improve pain outcomes in a variety of preclinical pain models. In this mini-review, we will highlight recent advances in understanding how cannabinoids modulate microglia function and its implications for cannabinoid-mediated analgesia, focusing on microglia-neuron interactions within the spinal nociceptive circuitry.
Collapse
Affiliation(s)
- Nynke J. van den Hoogen
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Erika K. Harding
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Chloé E. D. Davidson
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Tuan Trang
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- *Correspondence: Tuan Trang
| |
Collapse
|
7
|
Endocannabinoid System Attenuates Oxaliplatin-Induced Peripheral Sensory Neuropathy Through the Activation of CB1 Receptors. Neurotox Res 2021; 39:1782-1799. [PMID: 34792764 DOI: 10.1007/s12640-021-00442-x] [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: 04/07/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 10/19/2022]
Abstract
Oxaliplatin-induced neurotoxicity is expressed as a dose-limiting peripheral sensory neuropathy (PSN). Cannabinoid substances have been investigated for the analgesic effect. This study aimed to investigate the role of cannabinoid receptors in oxaliplatin-associated PSN. Swiss male mice received nine oxaliplatin injections (2 mg/kg, i.v.). Mechanical and thermal nociceptive tests were performed for 56 days. CB1, CB2, and c-Fos expression were assessed in dorsal root ganglia (DRG), spinal cord (SC), trigeminal ganglia (TG), spinal trigeminal nucleus caudalis (Sp5C), and periaqueductal gray (PAG). Iba-1 expression was assessed in DRG and ATF3 in TG. Cannabidiol (10 mg/kg, p.o.) or a CB1/CB2 non-selective agonist (WIN 55,212-2; 0.5 mg/kg, s.c.) or AM251 (CB1 antagonist) or AM630 (CB2 antagonist) (3 mg/kg, i.p.) were injected before oxaliplatin. Oxaliplatin increased CB1 in DRG, SC, TG, Sp5C, and ventrolateral PAG, with no interference in CB2 expression. Cannabidiol increased CB1 in DRG, reduced mechanical hyperalgesia and c-Fos expression in DRG and SC. Additionally, WIN 55,212-2 increased CB1 in DRG, reduced mechanical hyperalgesia, cold allodynia and c-Fos expression in DRG and SC. CB1 blockage hastened the cold allodynia response, but the CB2 antagonist failed to modulate the oxaliplatin-induced nociceptive behavior. Oxaliplatin also increased Iba-1 in DRG, suggesting immune response modulation which was reduced by cannabidiol and enhanced by AM630. The modulation of the endocannabinoid system, through the CB1 receptor, attenuates the oxaliplatin-associated PNS. The activation of the endocannabinoid system could be considered as a therapeutic target for controlling oxaliplatin-associated neuropathy.
Collapse
|
8
|
Duffy SS, Hayes JP, Fiore NT, Moalem-Taylor G. The cannabinoid system and microglia in health and disease. Neuropharmacology 2021; 190:108555. [PMID: 33845074 DOI: 10.1016/j.neuropharm.2021.108555] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/13/2022]
Abstract
Recent years have yielded significant advances in our understanding of microglia, the immune cells of the central nervous system (CNS). Microglia are key players in CNS development, immune surveillance, and the maintenance of proper neuronal function throughout life. In the healthy brain, homeostatic microglia have a unique molecular signature. In neurological diseases, microglia become activated and adopt distinct transcriptomic signatures, including disease-associated microglia (DAM) implicated in neurodegenerative disorders. Homeostatic microglia synthesise the endogenous cannabinoids 2-arachidonoylglycerol and anandamide and express the cannabinoid receptors CB1 and CB2 at constitutively low levels. Upon activation, microglia significantly increase their synthesis of endocannabinoids and upregulate their expression of CB2 receptors, which promote a protective microglial phenotype by enhancing their production of neuroprotective factors and reducing their production of pro-inflammatory factors. Here, we summarise the effects of the microglial cannabinoid system in the CNS demyelinating disease multiple sclerosis, the neurodegenerative diseases Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis, chronic inflammatory and neuropathic pain, and psychiatric disorders including depression, anxiety and schizophrenia. We discuss the therapeutic potential of cannabinoids in regulating microglial activity and highlight the need to further investigate their specific microglia-dependent immunomodulatory effects.
Collapse
Affiliation(s)
- Samuel S Duffy
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, UNSW Sydney, NSW, 2052, Australia
| | - Jessica P Hayes
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, UNSW Sydney, NSW, 2052, Australia
| | - Nathan T Fiore
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, UNSW Sydney, NSW, 2052, Australia
| | - Gila Moalem-Taylor
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, UNSW Sydney, NSW, 2052, Australia.
| |
Collapse
|
9
|
Hsu JM, Shiue SJ, Yang KD, Shiue HS, Hung YW, Pannuru P, Poongodi R, Lin HY, Cheng JK. Locally Applied Stem Cell Exosome-Scaffold Attenuates Nerve Injury-Induced Pain in Rats. J Pain Res 2020; 13:3257-3268. [PMID: 33304105 PMCID: PMC7723240 DOI: 10.2147/jpr.s286771] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 11/13/2020] [Indexed: 12/18/2022] Open
Abstract
Purpose Nerve injury-induced pain is difficult to treat. In this study, we developed an alginate scaffold with human umbilical cord mesenchymal stem cell exosomes (EX-SC) to treat nerve injury-induced pain. Materials and Methods The scaffold was prepared and characterized for its physical traits and biocompatibility. In vitro studies of PC12 and HEK293 cells were used to evaluate the neuroprotective and neurotrophic effects of exosomes. Right L5/6 spinal nerve ligation (SNL) was performed in Sprague-Dawley rats to induce mechanical allodynia and thermal hyperalgesia, evaluated by von Frey hair and radiant heat tests. The EX-SC was wrapped around ligated L5/6 spinal nerves for treatment. Western blotting and immunofluorescence staining were used to evaluate neuron/glial activation, cytokines and neurotrophic factor of affected dorsal root ganglion (DRG). Results In cell culture assay, the exosomes induce neurite outgrowth of PC12 cells and protect PC12 and HEK293 cells against formaldehyde acid treatment. On post-ligation day 21, rats receiving EX-SC had significantly higher median (interquartile range) withdrawal threshold and latency [14.1 (13.7-15.5) g, 14.2 (13.7-15.3) s] than saline-SC-treated rats [2.1 (1.7-3.0) g, 2.0 (1.8-2.4) s, P=0.02 and 0.002]. The EX-SC also attenuated SNL-induced up-regulation of c-Fos, GFAP, Iba1, TNF-α and IL-1β, while enhancing the level of IL-10 and GDNF, in the ipsilateral L5/6 DRG. After implantation for 21 days, the EX-SC enhanced the expression of myelin basic protein and IL-10 in injured L5/6 axons. Conclusion We demonstrate the EX-SC possesses antinociceptive, anti-inflammation and pro-neurotrophic effects in the SNL pain model. It could be a promising therapeutic alternative for nerve injury-induced pain.
Collapse
Affiliation(s)
- Jong-Ming Hsu
- Department of Urology, Mackay Memorial Hospital, Taipei 10449, Taiwan.,Mackay Junior College of Medicine, Nursing, and Management, Taipei 11260, Taiwan.,Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan
| | - Sheng-Jie Shiue
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan
| | - Kuender D Yang
- Institute of Biomedical Science, Mackay Medical College, New Taipei City 252, Taiwan.,Department of Pediatrics, Mackay Memorial Hospital, Taipei 10449, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| | - Han-Shiang Shiue
- Institute of Neuroscience, National Yang-Ming University, Taipei 11221, Taiwan
| | - Yi-Wei Hung
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan
| | - Pavani Pannuru
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan
| | - Raju Poongodi
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan
| | - Hsin-Yi Lin
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 106, Taiwan.,Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of Technology, Taipei 106, Taiwan
| | - Jen-Kun Cheng
- Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan.,Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan.,Department of Anesthesiology, Mackay Memorial Hospital, Taipei 10449, Taiwan
| |
Collapse
|
10
|
Binda K, Real C, Ferreira A, Britto L, Chacur M. Antinociceptive effects of treadmill exercise in a rat model of Parkinson's disease: The role of cannabinoid and opioid receptors. Brain Res 2020; 1727:146521. [DOI: 10.1016/j.brainres.2019.146521] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 09/19/2019] [Accepted: 10/21/2019] [Indexed: 12/17/2022]
|
11
|
Abstract
Nerve injury-induced neuropathic pain is difficult to treat. In this study, we used exosomes derived from human umbilical cord mesenchymal stem cell (UCMSC) as a cell-free therapy for nerve injury-induced pain in rats. Isolated UCMSC exosomes range in size from 30 to 160 nm and contain CD63, HSP60, and CD81 exosome markers. After L5/6 spinal nerve ligation surgery, single intrathecal injection of exosomes reversed nerve ligation-induced mechanical and thermal hypersensitivities of right hindpaw of rats at initial and well-developed pain stages. Moreover, continuous intrathecal infusion of exosomes achieved excellent preventive and reversal effects for nerve ligation-induced pain. In immunofluorescent study, lots of Exo-green-labelled exosomes could be found majorly in the ipsilateral L5 spinal dorsal horn, dorsal root ganglion, and peripheral axons, suggesting the homing ability of UCMSC exosomes. They also appeared in the central terminals or cell bodies of IB4, CGRP, and NF200 sensory neurons. In addition, exosome treatment suppressed nerve ligation-induced upregulation of c-Fos, CNPase, GFAP, and Iba1. All these data suggest that the analgesic effects of exosomes may involve their actions on neuron and glial cells. Exosomes also inhibited the level of TNF-α and IL-1β, while enhanced the level of IL-10, brain-derived neurotrophic factor, and glial cell line-derived neurotrophic factor in the ipsilateral L5/6 dorsal root ganglion of nerve-ligated rats, indicating anti-inflammatory and proneurotrophic abilities. Protein analysis revealed the content of vascular endothelial growth factor C, angiopoietin-2, and fibroblast growth factor-2 in the exosomes. In summary, intrathecal infusion of exosomes from UCMSCs may be considered as a novel therapeutic approach for nerve injury-induced pain.
Collapse
|
12
|
Guerrero-Alba R, Barragán-Iglesias P, González-Hernández A, Valdez-Moráles EE, Granados-Soto V, Condés-Lara M, Rodríguez MG, Marichal-Cancino BA. Some Prospective Alternatives for Treating Pain: The Endocannabinoid System and Its Putative Receptors GPR18 and GPR55. Front Pharmacol 2019; 9:1496. [PMID: 30670965 PMCID: PMC6331465 DOI: 10.3389/fphar.2018.01496] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/07/2018] [Indexed: 12/12/2022] Open
Abstract
Background: Marijuana extracts (cannabinoids) have been used for several millennia for pain treatment. Regarding the site of action, cannabinoids are highly promiscuous molecules, but only two cannabinoid receptors (CB1 and CB2) have been deeply studied and classified. Thus, therapeutic actions, side effects and pharmacological targets for cannabinoids have been explained based on the pharmacology of cannabinoid CB1/CB2 receptors. However, the accumulation of confusing and sometimes contradictory results suggests the existence of other cannabinoid receptors. Different orphan proteins (e.g., GPR18, GPR55, GPR119, etc.) have been proposed as putative cannabinoid receptors. According to their expression, GPR18 and GPR55 could be involved in sensory transmission and pain integration. Methods: This article reviews select relevant information about the potential role of GPR18 and GPR55 in the pathophysiology of pain. Results: This work summarized novel data supporting that, besides cannabinoid CB1 and CB2 receptors, GPR18 and GPR55 may be useful for pain treatment. Conclusion: There is evidence to support an antinociceptive role for GPR18 and GPR55.
Collapse
Affiliation(s)
- Raquel Guerrero-Alba
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Paulino Barragán-Iglesias
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States
| | - Abimael González-Hernández
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Santiago de Querétaro, Mexico
| | - Eduardo E Valdez-Moráles
- Cátedras CONACYT, Departamento de Cirugía, Centro de Ciencias Biomédicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, Mexico City, Mexico
| | - Miguel Condés-Lara
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Santiago de Querétaro, Mexico
| | - Martín G Rodríguez
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Bruno A Marichal-Cancino
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| |
Collapse
|
13
|
Gamble LJ, Boesch JM, Frye CW, Schwark WS, Mann S, Wolfe L, Brown H, Berthelsen ES, Wakshlag JJ. Pharmacokinetics, Safety, and Clinical Efficacy of Cannabidiol Treatment in Osteoarthritic Dogs. Front Vet Sci 2018; 5:165. [PMID: 30083539 PMCID: PMC6065210 DOI: 10.3389/fvets.2018.00165] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 07/02/2018] [Indexed: 11/13/2022] Open
Abstract
Objectives: The objectives of this study were to determine basic oral pharmacokinetics, and assess safety and analgesic efficacy of a cannabidiol (CBD) based oil in dogs with osteoarthritis (OA). Methods: Single-dose pharmacokinetics was performed using two different doses of CBD enriched (2 and 8 mg/kg) oil. Thereafter, a randomized placebo-controlled, veterinarian, and owner blinded, cross-over study was conducted. Dogs received each of two treatments: CBD oil (2 mg/kg) or placebo oil every 12 h. Each treatment lasted for 4 weeks with a 2-week washout period. Baseline veterinary assessment and owner questionnaires were completed before initiating each treatment and at weeks 2 and 4. Hematology, serum chemistry and physical examinations were performed at each visit. A mixed model analysis, analyzing the change from enrollment baseline for all other time points was utilized for all variables of interest, with a p ≤ 0.05 defined as significant. Results: Pharmacokinetics revealed an elimination half-life of 4.2 h at both doses and no observable side effects. Clinically, canine brief pain inventory and Hudson activity scores showed a significant decrease in pain and increase in activity (p < 0.01) with CBD oil. Veterinary assessment showed decreased pain during CBD treatment (p < 0.02). No side effects were reported by owners, however, serum chemistry showed an increase in alkaline phosphatase during CBD treatment (p < 0.01). Clinical significance: This pharmacokinetic and clinical study suggests that 2 mg/kg of CBD twice daily can help increase comfort and activity in dogs with OA.
Collapse
Affiliation(s)
- Lauri-Jo Gamble
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Jordyn M Boesch
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Christopher W Frye
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Wayne S Schwark
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Sabine Mann
- Department of Population Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Lisa Wolfe
- Proteomic and Metabolomic Facility, Colorado State University, Fort Collins, CO, United States
| | - Holly Brown
- Metzger Animal Hospital, State College, PA, United States
| | - Erin S Berthelsen
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Joseph J Wakshlag
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| |
Collapse
|
14
|
Bufalin attenuates cancer-induced pain and bone destruction in a model of bone cancer. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:1211-1219. [PMID: 28840279 DOI: 10.1007/s00210-017-1419-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/10/2017] [Indexed: 01/01/2023]
Abstract
Bufalin is a natural anti-inflammatory small molecule. Given the close relationship between inflammation and cancer, many scholars have studied the effect of bufalin on cancer in vitro, but in vivo research is still lacking. A murine bone cancer model was used in this study. We conducted pain sensitive test on mice with bone cancer, by nocifensive behavior, mechanical allodynia, and thermal hyperalgesia. Serum levels of bone loss markers with bufalin treatment were measured by ELISA. Expressions of osteoprotegerin (OPG) and receptor activator of NF-κB ligand (RANKL) were analyzed in bufalin-treated mice by real-time PCR and Western blot. Cannabinoid 2 receptor (CB2) inverse agonist AM630 was administrated to mice with bone cancer together with bufalin. Bufalin relieved cancer-induced pain and bone destruction in the murine bone cancer model. Serum levels of bone loss markers after bufalin treatment were reduced. Bufalin upregulated OPG and downregulated RANKL. The CB2 receptor inverse agonist, AM630, reduced the pain relief of bufalin treatment in the mouse bone cancer model. This study demonstrates that bufalin relieves cancer-induced pain and bone destruction, which is mediated through the CB2 receptor.
Collapse
|