1
|
Micheli L, Balayssac D, Busserolles J, Dalbos C, Prival L, Richard D, Quintana M, Di Cesare Mannelli L, Toti A, Ciampi C, Ghelardini C, Vlasakova K, Glaab WE, Hu Y, Loryan I, Perrault O, Slaoui M, Wuersch K, Johnson E, Frieauff W, Penraat K, Brees D, Dubost V, Theil D. The challenge to identify sensitive safety biomarkers of peripheral neurotoxicity in the rat: A collaborative effort across industry and academia (IMI NeuroDeRisk project). Toxicology 2025; 510:153998. [PMID: 39551123 DOI: 10.1016/j.tox.2024.153998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 11/19/2024]
Abstract
Peripheral nervous system (PNS) toxicity assessment in non-clinical safety studies is challenging and relies mostly on histopathological assessment. The present work aims to identify blood-based biomarkers that could detect peripheral neuropathy in rats upon exposure to neurotoxic compounds. Three anticancer agents (oxaliplatin, cisplatin, paclitaxel) and a developmental compound (NVS-1) were assessed in male rats (Wistar Han). Clinical and/or functional endpoints (i.e., electronic Von Frey, Cold Plate, and Paw Pressure tests) and blood biomarkers (i.e., neurofilament light chain (NfL), neurofilament heavy chain (NF-H), microtubule-associated protein Tau (Tau), neuron specific enolase (NSE), vascular endothelial growth factor A (VEGFA), and glial fibrillary acidic protein (GFAP)) were assessed. Drug exposure and histopathological evaluations were conducted on selected nervous tissues. Oxaliplatin, cisplatin and paclitaxel treatment resulted in a significant decrease of nociceptive thresholds. Clinical signs suggestive of PNS toxicity were observed with NVS-1. NfL was consistently increased in the NVS-1 study and correlated with moderate microscopic findings in dorsal root ganglia (DRG). Only minimal microscopic findings were observed in oxaliplatin-treated animals, whereas no treatment-related microscopic findings were observed in animals treated with cisplatin and paclitaxel. For all compounds, exposure was confirmed in the PNS tissues. Clinical and functional changes were observed with all the compounds evaluated. NfL levels in plasma proved to be the most sensitive indicator of PNS toxicities, capturing moderate nervous degeneration in DRG. A combined approach that includes both functional assessments and biomarker measurements offers a more comprehensive evaluation than histopathological analysis alone when monitoring drug-induced neurotoxicity in rat models.
Collapse
Affiliation(s)
- Laura Micheli
- University of Florence, Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba - Section of Pharmacology and Toxicology, Florence, Italy
| | - David Balayssac
- Université Clermont Auvergne, INSERM, U1107, NEURO-DOL, CHU Clermont-Ferrand, Direction de la Recherche Clinique et de l'Innovation, Clermont-Ferrand, France
| | - Jérôme Busserolles
- Université Clermont Auvergne, INSERM, U1107, NEURO-DOL, Clermont-Ferrand, France
| | - Cristelle Dalbos
- Université Clermont Auvergne, INSERM, U1107, NEURO-DOL, Clermont-Ferrand, France
| | - Laetitia Prival
- Université Clermont Auvergne, INSERM, U1107, NEURO-DOL, Clermont-Ferrand, France
| | - Damien Richard
- Université Clermont Auvergne, INSERM, U1107, NEURO-DOL, CHU Clermont-Ferrand, Laboratoire de Pharmacologie et de Toxicologie, Clermont-Ferrand, France
| | - Mercedes Quintana
- Université Clermont Auvergne, INSERM, U1240, Imagerie Moléculaire et Stratégies Théranostiques, Clermont-Ferrand, France
| | - Lorenzo Di Cesare Mannelli
- University of Florence, Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba - Section of Pharmacology and Toxicology, Florence, Italy.
| | - Alessandra Toti
- University of Florence, Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba - Section of Pharmacology and Toxicology, Florence, Italy
| | - Clara Ciampi
- University of Florence, Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba - Section of Pharmacology and Toxicology, Florence, Italy.
| | - Carla Ghelardini
- University of Florence, Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba - Section of Pharmacology and Toxicology, Florence, Italy
| | | | - Warren E Glaab
- Merck&Co., Inc., Nonclinical Drug Safety, Rahway, NJ, USA
| | - Yang Hu
- Translational Pharmacokinetics-Pharmacodynamics group, tPKPD, Department of Pharmacy, Faculty of Pharmacy, Uppsala University, Box 580, Uppsala SE-751 23, Sweden
| | - Irena Loryan
- Translational Pharmacokinetics-Pharmacodynamics group, tPKPD, Department of Pharmacy, Faculty of Pharmacy, Uppsala University, Box 580, Uppsala SE-751 23, Sweden
| | | | | | | | - Eric Johnson
- Novartis Biomedical Research, Cambridge, MA, USA
| | | | | | | | | | - Diethilde Theil
- Novartis Pharma AG, Basel, Switzerland; Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| |
Collapse
|
2
|
Gędek A, Modrzejewski S, Materna M, Szular Z, Wichniak A, Mierzejewski P, Dominiak M. Efficacy and Safety of Agomelatine in Depressed Patients with Diabetes: A Systematic Review and Meta-Analysis. Int J Mol Sci 2024; 25:12631. [PMID: 39684343 DOI: 10.3390/ijms252312631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Major depressive disorder (MDD) and diabetes mellitus (DM) remain among the most prevalent diseases and the most significant challenges faced by medicine in the 21st century. The frequent co-occurrence and bidirectional relationship between the two conditions necessitates the identification of treatment strategies that benefit both. The purpose of this study was to systematically review and meta-analyze data on the efficacy and safety of agomelatine (AGO) in the treatment of patients with depression with comorbid diabetes to explore its potential mechanism of action in both diseases and its impact on diabetic parameters. Following PRISMA guidelines, a total of 11 studies were identified, both preclinical and clinical trials. Agomelatine has shown great potential as a treatment option for patients with diabetes and comorbid depression and anxiety. In addition to improving depressive and anxiety symptoms, it is also beneficial in glycemic control. A meta-analysis demonstrated a statistically significant reduction in glycated hemoglobin (HbA1C) and fasting blood glucose (FBG) levels following AGO administration over a period of 8-16 weeks. The administration of agomelatine was found to result in a significantly greater reduction in HbA1C than that observed with the selective serotonin reuptake inhibitor (SSRI) medications (namely fluoxetine, sertraline, and paroxetine) during 12-16 weeks of therapy. Furthermore, AGO has been found to be at least as effective as SSRIs in reducing depressive symptoms and more effective than SSRIs in reducing anxiety symptoms. The safety of such treatment is similar to SSRIs; no severe adverse events were reported, and the incidence of some side effects, such as insomnia and sexual dysfunction, are even less often reported. Particularly promising is also its potential action in improving some diabetic complications reported in preclinical trials. This might be through mechanisms involving the reduction in oxidative stress, anti-inflammatory effects, and potentially noradrenergic or NMDA receptor modulation. Further clinical studies on larger sample sizes, as well as elucidating its mechanisms of action, especially in the context of diabetic complications, are needed. Research should also focus on identifying the patient subpopulations most likely to benefit from agomelatine treatment.
Collapse
Affiliation(s)
- Adam Gędek
- Department of Pharmacology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
- Third Department of Psychiatry, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| | | | | | - Zofia Szular
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Adam Wichniak
- Third Department of Psychiatry, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| | - Paweł Mierzejewski
- Department of Pharmacology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| | - Monika Dominiak
- Department of Pharmacology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| |
Collapse
|
3
|
Lucarini E, Micheli L, Rajagopalan R, Ciampi C, Branca JJ, Pacini A, Leandri M, Rajagopalan P, Ghelardini C, Di Cesare Mannelli L. Broad-spectrum neuroprotection exerted by DDD-028 in a mouse model of chemotherapy-induced neuropathy. Pain 2023; 164:2581-2595. [PMID: 37556385 PMCID: PMC10578426 DOI: 10.1097/j.pain.0000000000002963] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/28/2023] [Accepted: 05/02/2023] [Indexed: 08/11/2023]
Abstract
ABSTRACT Neurotoxicity of chemotherapeutics involves peculiar alterations in the structure and function, including abnormal nerve signal transmission, of both the peripheral and central nervous system. The lack of effective pharmacological approaches to prevent chemotherapy-induced neurotoxicity necessitates the identification of innovative therapies. Recent evidence suggests that repeated treatment with the pentacyclic pyridoindole derivative DDD-028 can exert both pain-relieving and glial modulatory effects in mice with paclitaxel-induced neuropathy. This work is aimed at assessing whether DDD-028 is a disease-modifying agent by protecting the peripheral nervous tissues from chemotherapy-induced damage. Neuropathy was induced in animals by paclitaxel injection (2.0 mg kg -1 i.p). DDD-028 (10 mg kg -1 ) and the reference drug, pregabalin (30 mg kg -1 ), were administered per os daily starting concomitantly with the first injection of paclitaxel and continuing 10 days after the end of paclitaxel treatment. The behavioural tests confirmed the antihyperalgesic efficacy of DDD-028 on paclitaxel-induced neuropathic pain. Furthermore, the electrophysiological analysis revealed the capacity of DDD-028 to restore near-normal sensory nerve conduction in paclitaxel-treated animals. Histopathology evidence indicated that DDD-028 was able to counteract effectively paclitaxel-induced peripheral neurotoxicity by protecting against the loss of intraepidermal nerve fibers, restoring physiological levels of neurofilament in nerve tissue and plasma, and preventing morphological alterations occurring in the sciatic nerves and dorsal root ganglia. Overall, DDD-028 is more effective than pregabalin in preventing chemotherapy-induced neurotoxicity. Thus, based on its potent antihyperalgesic and neuroprotective efficacy, DDD-028 seems to be a viable prophylactic medication to limit the development of neuropathies consequent to chemotherapy.
Collapse
Affiliation(s)
- Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | | | - Clara Ciampi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Jacopo J.V. Branca
- Department of Experimental and Clinical Medicine, Anatomy and Histology Section, University of Florence, Florence, Italy
| | - Alessandra Pacini
- Department of Experimental and Clinical Medicine, Anatomy and Histology Section, University of Florence, Florence, Italy
| | - Massimo Leandri
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | | | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| |
Collapse
|
4
|
Arias HR, Tae HS, Micheli L, Yousuf A, Manetti D, Romanelli MN, Ghelardini C, Adams DJ, Di Cesare Mannelli L. The Antinociceptive Activity of (E)-3-(thiophen-2-yl)- N -(p-tolyl)acrylamide in Mice Is Reduced by (E)-3-(furan-2-yl)- N -methyl- N -(p-tolyl)acrylamide Through Opposing Modulatory Mechanisms at the α7 Nicotinic Acetylcholine Receptor. Anesth Analg 2023; 137:691-701. [PMID: 37058425 PMCID: PMC10408732 DOI: 10.1213/ane.0000000000006461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2023] [Indexed: 04/15/2023]
Abstract
BACKGROUND The primary objective of this study was to characterize the pharmacological and behavioral activity of 2 novel compounds, DM497 [(E)-3-(thiophen-2-yl)- N -(p-tolyl)acrylamide] and DM490 [(E)-3-(furan-2-yl)- N -methyl- N -(p-tolyl)acrylamide], structural derivatives of PAM-2, a positive allosteric modulator of the α7 nicotinic acetylcholine receptor (nAChR). METHODS A mouse model of oxaliplatin-induced neuropathic pain (2.4 mg/kg, 10 injections) was used to test the pain-relieving properties of DM497 and DM490. To assess possible mechanisms of action, the activity of these compounds was determined at heterologously expressed α7 and α9α10 nAChRs, and voltage-gated N-type calcium channel (Ca V 2.2) using electrophysiological techniques. RESULTS Cold plate tests indicated that 10 mg/kg DM497 was able to decrease neuropathic pain in mice induced by the chemotherapeutic agent oxaliplatin. In contrast, DM490 induced neither pro- nor antinociceptive activity but inhibited DM497's effect at equivalent dose (30 mg/kg). These effects are not a product of changes in motor coordination or locomotor activity. At α7 nAChRs, DM497 potentiated whereas DM490 inhibited its activity. In addition, DM490 antagonized the α9α10 nAChR with >8-fold higher potency than that for DM497. In contrast, DM497 and DM490 had minimal inhibitory activity at the Ca V 2.2 channel. Considering that DM497 did not increase the mouse exploratory activity, an indirect anxiolytic mechanism was not responsible for the observed antineuropathic effect. CONCLUSIONS The antinociceptive activity of DM497 and the concomitant inhibitory effect of DM490 are mediated by opposing modulatory mechanisms on the α7 nAChR, whereas the involvement of other possible nociception targets such as the α9α10 nAChR and Ca V 2.2 channel can be ruled out.
Collapse
Affiliation(s)
- Hugo R. Arias
- From the Department of Pharmacology and Physiology, College of Osteopathic Medicine, Oklahoma State University Center for Health Sciences, Tahlequah, Oklahoma
| | - Han-Shen Tae
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales, Australiaand Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA)
| | | | - Arsalan Yousuf
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales, Australiaand Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA)
| | - Dina Manetti
- Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Florence, Italy
| | - Maria Novella Romanelli
- Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Florence, Italy
| | | | - David J. Adams
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales, Australiaand Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA)
| | | |
Collapse
|
5
|
Cheng F, Zhang R, Sun C, Ran Q, Zhang C, Shen C, Yao Z, Wang M, Song L, Peng C. Oxaliplatin-induced peripheral neurotoxicity in colorectal cancer patients: mechanisms, pharmacokinetics and strategies. Front Pharmacol 2023; 14:1231401. [PMID: 37593174 PMCID: PMC10427877 DOI: 10.3389/fphar.2023.1231401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/18/2023] [Indexed: 08/19/2023] Open
Abstract
Oxaliplatin-based chemotherapy is a standard treatment approach for colorectal cancer (CRC). However, oxaliplatin-induced peripheral neurotoxicity (OIPN) is a severe dose-limiting clinical problem that might lead to treatment interruption. This neuropathy may be reversible after treatment discontinuation. Its complicated mechanisms are related to DNA damage, dysfunction of voltage-gated ion channels, neuroinflammation, transporters, oxidative stress, and mitochondrial dysfunction, etc. Several strategies have been proposed to diminish OIPN without compromising the efficacy of adjuvant therapy, namely, combination with chemoprotectants (such as glutathione, Ca/Mg, ibudilast, duloxetine, etc.), chronomodulated infusion, dose reduction, reintroduction of oxaliplatin and topical administration [hepatic arterial infusion chemotherapy (HAIC), pressurized intraperitoneal aerosol chemotherapy (PIPAC), and hyperthermic intraperitoneal chemotherapy (HIPEC)]. This article provides recent updates related to the potential mechanisms, therapeutic strategies in treatment of OIPN, and pharmacokinetics of several methods of oxaliplatin administration in clinical trials.
Collapse
Affiliation(s)
- Fang Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ruoqi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chen Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Ran
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cuihan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Changhong Shen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziqing Yao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Miao Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin Song
- Department of Pharmacy, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
6
|
Micheli L, Ferrara V, Akande T, Khatib M, Salawu SO, Ciampi C, Lucarini E, Di Cesare Mannelli L, Mulinacci N, Ghelardini C. Ellagitannins and triterpenoids extracts of Anogeissus leiocarpus stem bark extracts: Protective effects against osteoarthritis. Phytother Res 2023. [PMID: 36806301 DOI: 10.1002/ptr.7760] [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: 07/18/2022] [Revised: 12/27/2022] [Accepted: 01/13/2023] [Indexed: 02/22/2023]
Abstract
Osteoarthritis (OA) is a complex joint disease characterized by persistent pain. Unfortunately, current pharmacological therapies are unsatisfactory and characterized by side effects, reason why new strategies are needed. We tested the efficacy of different classes of compounds, ellagitannins and olean-type triterpenoids, contained in Anogeissus leiocarpus extract (Combretaceae family) in comparison to ellagitannins of Castanea sativa extract in a rat model of osteoarthritis induced by the intra-articular injection of sodium monoiodoacetate (MIA). The decoction of stem bark of A. leiocarpus AL-DEC-TOT (300 mg/kg; 4.8% triterpenoids; 11.0% tannins), the butanol extract AL-BuOH-EXT (120 mg/kg; triterpenoids 20.9%; tannins 6.4%) and its correlated aqueous residue AL-Res-H2 O (300 mg/kg; triterpenoids 0.7%; tannins 8.7%) and the decoction of C. sativa, CS-DEC-TOT, (240 mg/kg; triterpenoids 0.65%; tannins 10.8%) were orally administered for two weeks starting from the day of the damage. Behavioural tests highlighted that all stem bark extracts of A. leiocarpus counteracted hypersensitivity development, reduced spontaneous pain, and improved motor skills. Histologically, AL-DEC-TOT, AL-BuOH-EXT and AL-Res-H2 O were effective in preventing joint alterations. In conclusion, all the extracts were effective demonstrating that both olean-type triterpenoid and ellagitannin fractions have anti-hypersensitivity and restorative properties running the stem bark extracts of A. leiocarpus as a candidate in the treatment of OA.
Collapse
Affiliation(s)
- Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Valentina Ferrara
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Titilayo Akande
- Department of Biochemistry-Functional Foods and Nutraceuticals Unit, School of Sciences, Federal University of Technology Akure, Akure, Nigeria
| | - Mohamad Khatib
- Department of Neuroscience, Psychology, Drug and Child Health-NEUROFARBA-Pharmaceutical and Nutraceutical Section, University of Florence and Multidisciplinary Centre for Research on food Sciences (M.C.R.F.S-Ce.R.A.), University of Florence, Florence, Italy
| | - Sule Ola Salawu
- Department of Biochemistry-Functional Foods and Nutraceuticals Unit, School of Sciences, Federal University of Technology Akure, Akure, Nigeria
| | - Clara Ciampi
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Nadia Mulinacci
- Department of Neuroscience, Psychology, Drug and Child Health-NEUROFARBA-Pharmaceutical and Nutraceutical Section, University of Florence and Multidisciplinary Centre for Research on food Sciences (M.C.R.F.S-Ce.R.A.), University of Florence, Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| |
Collapse
|
7
|
Micheli L, Maggini V, Ciampi C, Gallo E, Bogani P, Fani R, Pistelli L, Ghelardini C, Di Cesare Mannelli L, De Leo M, Firenzuoli F. Echinacea purpurea against neuropathic pain: Alkamides versus polyphenols efficacy. Phytother Res 2022; 37:1911-1923. [PMID: 36578266 DOI: 10.1002/ptr.7709] [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: 09/13/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/30/2022]
Abstract
Chemotherapy-induced neuropathy represents the main dose-limiting toxicity of several anticancer drugs, such as oxaliplatin, leading to chronic pain and an impairment of the quality of life. Echinacea purpurea n-hexane extract (EP4 -RE ; rich in alkamides) and butanolic extract (EP4 -RBU ; rich in polyphenols) have been characterized and tested in an in vivo model of oxaliplatin-induced neuropathic pain, addressing the endocannabinoid system with alkamides and counteracting the redox imbalance with polyphenols. Thermal hypersensitivity was evaluated by the Cold Plate test. EP4 -RE showed a dose-dependent anti-hyperalgesic profile. The extract was more effective than its main constituent, dodeca-2 E,4 E,8Z,10 E/Z-tetraenoic acid isobutylamide (18 mg kg-1 , twofold to equimolar EP4 -RE 30 mg kg-1 ), suggesting a synergy with other extract constituents. Administration of cannabinoid type 2 (CB2) receptor-selective antagonist completely blocked the anti-allodynic effect of EP4 -RE , differently from the antagonism of CB1 receptors. EP4 -RBU (30 mg kg-1 ) exhibited anti-neuropathic properties too. The effect was mainly exerted by chicoric acid, which administered alone (123 μg kg-1 , equimolar to EP4 -RBU 30 mg kg-1 ) completely reverted oxaliplatin-induced allodynia. A synergy between different polyphenols in the extract had not been highlighted. Echinacea extracts have therapeutic potential in the treatment of neuropathic pain, through both alkamides CB2-selective activity and polyphenols protective properties.
Collapse
Affiliation(s)
- Laura Micheli
- Department of Neurosciences, Psychology, Drug Research and Child Health-Neurofarba-Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Valentina Maggini
- Research and Innovation Center in Phytotherapy and Integrated Medicine (CERFIT), Referring Center for Phytotherapy, Tuscany Region, Careggi University Hospital, Florence, Italy
| | - Clara Ciampi
- Department of Neurosciences, Psychology, Drug Research and Child Health-Neurofarba-Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Eugenia Gallo
- Research and Innovation Center in Phytotherapy and Integrated Medicine (CERFIT), Referring Center for Phytotherapy, Tuscany Region, Careggi University Hospital, Florence, Italy
| | - Patrizia Bogani
- Department of Biology, University of Florence, Sesto Fiorentino, Italy
| | - Renato Fani
- Department of Biology, University of Florence, Sesto Fiorentino, Italy
| | - Luisa Pistelli
- Department of Pharmacy, University of Pisa, Pisa, Italy.,CISUP, Centre for Instrumentation Sharing Pisa University, Pisa, Italy
| | - Carla Ghelardini
- Department of Neurosciences, Psychology, Drug Research and Child Health-Neurofarba-Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neurosciences, Psychology, Drug Research and Child Health-Neurofarba-Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Marinella De Leo
- Department of Pharmacy, University of Pisa, Pisa, Italy.,CISUP, Centre for Instrumentation Sharing Pisa University, Pisa, Italy
| | - Fabio Firenzuoli
- Research and Innovation Center in Phytotherapy and Integrated Medicine (CERFIT), Referring Center for Phytotherapy, Tuscany Region, Careggi University Hospital, Florence, Italy
| |
Collapse
|
8
|
Bonifacino T, Micheli L, Torazza C, Ghelardini C, Farina C, Bonanno G, Milanese M, Di Cesare Mannelli L, Scherz MW. Pharmacological Profile of MP-101, a Novel Non-racemic Mixture of R- and S-dimiracetam with Increased Potency in Rat Models of Cognition, Depression and Neuropathic Pain. Cells 2022; 11:cells11244027. [PMID: 36552791 PMCID: PMC9776800 DOI: 10.3390/cells11244027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
The racemic mixture dimiracetam negatively modulates NMDA-induced glutamate release in rat spinal cord synaptosomal preparations and is orally effective in models of neuropathic pain. In this study, we compared the effects of dimiracetam, its R- or S-enantiomers, and the R:S 3:1 non-racemic mixture (MP-101). In vitro, dimiracetam was more potent than its R- or S-enantiomers in reducing the NMDA-induced [3H]D-aspartate release in rat spinal cord synaptosomes. Similarly, acute oral administration of dimiracetam was more effective than a single enantiomer in the sodium monoiodoacetate (MIA) paradigm of painful osteoarthritis. Then, we compared the in vitro effects of a broad range of non-racemic enantiomeric mixtures on the NMDA-induced [3H]D-aspartate release. Dimiracetam was a more potent blocker than each isolated enantiomer but the R:S 3:1 non-racemic mixture (MP-101) was even more potent than dimiracetam, with an IC50 in the picomolar range. In the chronic oxaliplatin-induced neuropathic pain model, MP-101 showed a significantly improved anti-neuropathic profile, and its effect continued one week after treatment suspension. MP-101 also performed better than dimiracetam in animal models of cognition and depression. Based on the benign safety and tolerability profile previously observed with racemic dimiracetam, MP-101 appears to be a novel, promising clinical candidate for the prevention and treatment of several neuropathic and neurological disorders.
Collapse
Affiliation(s)
- Tiziana Bonifacino
- Department of Pharmacy (DIFAR), Pharmacology and Toxicology Unit, University of Genoa, 16148 Genoa, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - Carola Torazza
- Department of Pharmacy (DIFAR), Pharmacology and Toxicology Unit, University of Genoa, 16148 Genoa, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - Carlo Farina
- Metys Pharmaceuticals c/o Novaremed AG, 4051 Basel, Switzerland
| | - Giambattista Bonanno
- Department of Pharmacy (DIFAR), Pharmacology and Toxicology Unit, University of Genoa, 16148 Genoa, Italy
- IRCCS, Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Marco Milanese
- Department of Pharmacy (DIFAR), Pharmacology and Toxicology Unit, University of Genoa, 16148 Genoa, Italy
- IRCCS, Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Correspondence: (M.M.); (L.D.C.M.)
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
- Correspondence: (M.M.); (L.D.C.M.)
| | | |
Collapse
|
9
|
In Silico Drug Repurposing Framework Predicts Repaglinide, Agomelatine and Protokylol as TRPV1 Modulators with Analgesic Activity. Pharmaceutics 2022; 14:pharmaceutics14122563. [PMID: 36559057 PMCID: PMC9781017 DOI: 10.3390/pharmaceutics14122563] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
Pain is one of the most common symptoms experienced by patients. The use of current analgesics is limited by low efficacy and important side effects. Transient receptor potential vanilloid-1 (TRPV1) is a non-selective cation channel, activated by capsaicin, heat, low pH or pro-inflammatory agents. Since TRPV1 is a potential target for the development of novel analgesics due to its distribution and function, we aimed to develop an in silico drug repositioning framework to predict potential TRPV1 ligands among approved drugs as candidates for treating various types of pain. Structures of known TRPV1 agonists and antagonists were retrieved from ChEMBL databases and three datasets were established: agonists, antagonists and inactive molecules (pIC50 or pEC50 < 5 M). Structures of candidates for repurposing were retrieved from the DrugBank database. The curated active/inactive datasets were used to build and validate ligand-based predictive models using Bemis−Murcko structural scaffolds, plain ring systems, flexophore similarities and molecular descriptors. Further, molecular docking studies were performed on both active and inactive conformations of the TRPV1 channel to predict the binding affinities of repurposing candidates. Variables obtained from calculated scaffold-based activity scores, molecular descriptors criteria and molecular docking were used to build a multi-class neural network as an integrated machine learning algorithm to predict TRPV1 antagonists and agonists. The proposed predictive model had a higher accuracy for classifying TRPV1 agonists than antagonists, the ROC AUC values being 0.980 for predicting agonists, 0.972 for antagonists and 0.952 for inactive molecules. After screening the approved drugs with the validated algorithm, repaglinide (antidiabetic) and agomelatine (antidepressant) emerged as potential TRPV1 antagonists, and protokylol (bronchodilator) as an agonist. Further studies are required to confirm the predicted activity on TRPV1 and to assess the candidates’ efficacy in alleviating pain.
Collapse
|
10
|
Micheli L, Toti A, Lucarini E, Ferrara V, Ciampi C, Olivero G, Pittaluga A, Mattoli L, Pelucchini C, Burico M, Lucci J, Carrino D, Pacini A, Pallanti S, Di Cesare Mannelli L, Ghelardini C. Efficacy of a vegetal mixture composed of Zingiber officinale, Echinacea purpurea, and Centella asiatica in a mouse model of neuroinflammation: In vivo and ex vivo analysis. Front Nutr 2022; 9:887378. [PMID: 36118773 PMCID: PMC9472218 DOI: 10.3389/fnut.2022.887378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 07/25/2022] [Indexed: 12/04/2022] Open
Abstract
Experimental evidence suggests that neuroinflammation is a key pathological event of many diseases affecting the nervous system. It has been well recognized that these devastating illnesses (e.g., Alzheimer’s, Parkinson’s, depression, and chronic pain) are multifactorial, involving many pathogenic mechanisms, reason why pharmacological treatments are unsatisfactory. The purpose of this study was to evaluate the efficacy of a vegetal mixture capable of offering a multiple approach required to manage the multifactoriality of neuroinflammation. A mixture composed of Zingiber officinale (150 mg kg−1), Echinacea purpurea (20 mg kg−1), and Centella asiatica (200 mg kg−1) was tested in a mouse model of systemic neuroinflammation induced by lipopolysaccharide (LPS, 1 mg kg−1). Repeated treatment with the vegetal mixture was able to completely counteract thermal and mechanical allodynia as reported by the Cold plate and von Frey tests, respectively, and to reduce the motor impairments as demonstrated by the Rota rod test. Moreover, the mixture was capable of neutralizing the memory loss in the Passive avoidance test and reducing depressive-like behavior in the Porsolt test, while no efficacy was shown in decreasing anhedonia as demonstrated by the Sucrose preference test. Finally, LPS stimulation caused a significant increase in the activation of glial cells, of the central complement proteins and of inflammatory cytokines in selected regions of the central nervous system (CNS), which were rebalanced in animals treated with the vegetal mixture. In conclusion, the vegetal mixture tested thwarted the plethora of symptoms evoked by LPS, thus being a potential candidate for future investigations in the context of neuroinflammation.
Collapse
Affiliation(s)
- Laura Micheli
- Neurofarba—Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- *Correspondence: Laura Micheli,
| | - Alessandra Toti
- Neurofarba—Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Elena Lucarini
- Neurofarba—Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Valentina Ferrara
- Neurofarba—Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Clara Ciampi
- Neurofarba—Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Guendalina Olivero
- Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Anna Pittaluga
- Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Luisa Mattoli
- Innovation and Medical Science Division, Aboca SpA Società Agricola, Sansepolcro, Italy
| | - Caroline Pelucchini
- Innovation and Medical Science Division, Aboca SpA Società Agricola, Sansepolcro, Italy
| | - Michela Burico
- Innovation and Medical Science Division, Aboca SpA Società Agricola, Sansepolcro, Italy
| | - Jacopo Lucci
- Innovation and Medical Science Division, Aboca SpA Società Agricola, Sansepolcro, Italy
| | - Donatello Carrino
- Anatomy and Histology Section, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandra Pacini
- Anatomy and Histology Section, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Stefano Pallanti
- Psychiatry Section, Department of Neurofarba, University of Florence, Florence, Italy
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, NY, United States
- Institute of Neuroscience, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Neurofarba—Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Carla Ghelardini
- Neurofarba—Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| |
Collapse
|
11
|
Inhibition of Monoacylglycerol Lipase by NSD1819 as an Effective Strategy for the Endocannabinoid System Modulation against Neuroinflammation-Related Disorders. Int J Mol Sci 2022; 23:ijms23158428. [PMID: 35955562 PMCID: PMC9369272 DOI: 10.3390/ijms23158428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
Neuroinflammation is a key pathological event shared by different diseases affecting the nervous system. Since the underlying mechanism of neuroinflammation is a complex and multifaceted process, current pharmacological treatments are unsatisfactory—a reason why new therapeutic approaches are mandatory. In this context, the endocannabinoid system has proven to possess neuroprotective and immunomodulatory actions under neuroinflammatory status, and its modulation could represent a valuable approach to address different inflammatory processes. To this aim, we evaluated the efficacy of a repeated treatment with NSD1819, a potent β-lactam-based monoacylglycerol lipase inhibitor in a mouse model of neuroinflammation induced by lipopolysaccharide (LPS) injection. Mice were intraperitoneally injected with LPS 1 mg/kg for five consecutive days to induce systemic inflammation. Concurrently, NSD1819 (3 mg/kg) was daily per os administered from day 1 until the end of the experiment (day 11). Starting from day 8, behavioral measurements were performed to evaluate the effect of the treatment on cognitive impairments, allodynia, motor alterations, anhedonia, and depressive-like behaviors evoked by LPS. Histologically, glial analysis of the spinal cord was also performed. The administration of NSD1819 was able to completely counteract thermal and mechanical allodynia as highlighted by the Cold plate and von Frey tests, respectively, and to reduce motor impairments as demonstrated by the Rota rod test. Moreover, the compound was capable of neutralizing the memory loss in the Passive avoidance test, and reducing depressive-like behavior in the Porsolt test. Finally, LPS stimulation caused a significant glial cells activation in the dorsal horn of the lumbar spinal cord that was significantly recovered by NSD1819 repeated treatment. In conclusion, NSD1819 was able to thwart the plethora of symptoms evoked by LPS, thus representing a promising candidate for future applications in the context of neuroinflammation and related diseases.
Collapse
|
12
|
Inhibitors of Mitochondrial Human Carbonic Anhydrases VA and VB as a Therapeutic Strategy against Paclitaxel-Induced Neuropathic Pain in Mice. Int J Mol Sci 2022; 23:ijms23116229. [PMID: 35682907 PMCID: PMC9181376 DOI: 10.3390/ijms23116229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 11/16/2022] Open
Abstract
Neuropathy development is a major dose-limiting side effect of anticancer treatments that significantly reduces patient's quality of life. The inadequate pharmacological approaches for neuropathic pain management warrant the identification of novel therapeutic targets. Mitochondrial dysfunctions that lead to reactive oxygen species (ROS) increase, cytosolic Ca2+ imbalance, and lactate acidosis are implicated in neuropathic pain pathogenesis. It has been observed that in these deregulations, a pivotal role is played by the mitochondrial carbonic anhydrases (CA) VA and VB isoforms. Hence, preclinical studies should be conducted to assess the efficacy of two novel selenides bearing benzenesulfonamide moieties, named 5b and 5d, and able to inhibit CA VA and VB against paclitaxel-induced neurotoxicity in mice. Acute treatment with 5b and 5d (30-100 mg/kg, per os - p.o.) determined a dose-dependent and long-lasting anti-hyperalgesic effect in the Cold plate test. Further, repeated daily treatment for 15 days with 100 mg/kg of both compounds (starting the first day of paclitaxel injection) significantly prevented neuropathic pain development without the onset of tolerance to the anti-hyperalgesic effect. In both experiments, acetazolamide (AAZ, 100 mg/kg, p.o.) used as the reference drug was partially active. Moreover, ex vivo analysis demonstrated the efficacy of 5b and 5d repeated treatments in reducing the maladaptive plasticity that occurs to glia cells in the lumbar portion of the spinal cord and in improving mitochondrial functions in the brain and spinal cord that were strongly impaired by paclitaxel-repeated treatment. In this regard, 5b and 5d ameliorated the metabolic activity, as observed by the increase in citrate synthase activity, and preserved an optimal mitochondrial membrane potential (ΔΨ) value, which appeared depolarized in brains from paclitaxel-treated animals. In conclusion, 5b and 5d have therapeutic and protective effects against paclitaxel-induced neuropathy without tolerance development. Moreover, 5b and 5d reduced glial cell activation and mitochondrial dysfunction in the central nervous system, being a promising candidate for the management of neuropathic pain and neurotoxicity evoked by chemotherapeutic drugs.
Collapse
|
13
|
Neuronal alarmin IL-1α evokes astrocyte-mediated protective signals: Effectiveness in chemotherapy-induced neuropathic pain. Neurobiol Dis 2022; 168:105716. [PMID: 35367629 DOI: 10.1016/j.nbd.2022.105716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 03/03/2022] [Accepted: 03/28/2022] [Indexed: 12/24/2022] Open
Abstract
The distinction between glial painful and protective pathways is unclear and the possibility to finely modulate the system is lacking. Focusing on painful neuropathies, we studied the role of interleukin 1α (IL-1α), an alarmin belonging to the larger family of damage-associated molecular patterns endogenously secreted to restore homeostasis. The treatment of rat primary neurons with increasing dose of the neurotoxic anticancer drug oxaliplatin (0.3-100μM, 48 h) induced the release of IL-1α. The knockdown of the alarmin in neurons leads to their higher mortality when co-cultured with astrocytes. This toxicity was related to increased extracellular ATP and decreased release of transforming growth factor β1, mostly produced by astrocytes. In a rat model of neuropathy induced by oxaliplatin, the intrathecal treatment with IL-1α was able to reduce mechanical and thermal hypersensitivity both after acute injection and continuous infusion. Ex vivo analysis on spinal purified astrocyte processes (gliosomes) and nerve terminals (synaptosomes) revealed the property of IL-1α to reduce the endogenous glutamate release induced by oxaliplatin. This protective effect paralleled with an increased number of GFAP-positive cells in the spinal cord, suggesting the ability of IL-1α to evoke a positive, conservative astrocyte phenotype. Endogenous IL-1α induces protective signals in the cross-talk between neurons and astrocytes. Exogenously administered in rats, IL-1α prevents neuropathic pain in the presence of spinal glutamate decrease and astrocyte activation.
Collapse
|
14
|
Lopes LT, Canto-de-Souza L, Baptista-de-Souza D, de Souza RR, Nunes-de-Souza RL, Canto-de-Souza A. The interplay between 5-HT 2C and 5-HT 3A receptors in the dorsal periaqueductal gray mediates anxiety-like behavior in mice. Behav Brain Res 2022; 417:113588. [PMID: 34547341 DOI: 10.1016/j.bbr.2021.113588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/17/2021] [Accepted: 09/15/2021] [Indexed: 11/17/2022]
Abstract
The monoamine neurotransmitter serotonin (5-HT) modulates anxiety by its activity on 5-HT2C receptors (5-HT2CR) expressed in the dorsal periaqueductal gray (dPAG). Here, we investigated the presence of 5-HT3A receptors (5-HT3AR) in the dPAG, and the interplay between 5-HT2CR and 5-HT3AR in the dPAG in mediating anxiety-like behavior in mice. We found that 5-HT3AR is expressed in the dPAG and the blockade of these receptors using intra-dPAG infusion of ondansetron (5-HT3AR antagonist; 3.0 nmol) induced an anxiogenic-like effect. The activation of 5-HT3ABR by the infusion of mCPBG [1-(m-Chlorophenyl)-biguanide; 5-HT3R agonist] did not alter anxiety-like behaviors. In addition, blockade of 5-HT3AR (1.0 nmol) prevented the anxiolytic-like effect induced by the infusion of the 5-HT2CR agonist mCPP (1-(3-chlorophenyl) piperazine; 0.03 nmol). None of the treatment effects on anxiety-like behaviors altered the locomotor activity levels. The present results suggest that the anxiolytic-like effect exerted by serotonin activity on 5-HT2CR in the dPAG is modulated by 5-HT3AR expressed in same region.
Collapse
Affiliation(s)
- Luana Tenorio Lopes
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, Alberta, Canada.
| | - Lucas Canto-de-Souza
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, Univ. Estadual Paulista, UNESP, Araraquara, SP 14801-902, Brazil; Neuroscience and Behavioral Institute, Av. do Café, 2.450, 14050-220 Ribeirão Preto, SP, Brazil.
| | - Daniela Baptista-de-Souza
- Psychobiology Group/Department of Psychology/CECH-UFSCar, São Carlos, SP 13565-905, Brazil; Laboratory of Pharmacology, School of Pharmaceutical Sciences, Univ. Estadual Paulista, UNESP, Araraquara, SP 14801-902, Brazil; Neuroscience and Behavioral Institute, Av. do Café, 2.450, 14050-220 Ribeirão Preto, SP, Brazil.
| | - Rimenez Rodrigues de Souza
- The University of Texas at Dallas, School of Behavior and Brain Sciences, 800 West Campbell Road, Richardson, TX 75080-3021, United States; The University of Texas at Dallas, Texas Biomedical Device Center, 800 West Campbell Road, Richardson, TX 75080-3021, United States.
| | - Ricardo L Nunes-de-Souza
- Psychobiology Group/Department of Psychology/CECH-UFSCar, São Carlos, SP 13565-905, Brazil; Joint Graduate Program in Physiological Sciences UFSCar/UNESP, Rod. Washington Luís, Km 235, São Carlos, SP 13565-905, Brazil; Laboratory of Pharmacology, School of Pharmaceutical Sciences, Univ. Estadual Paulista, UNESP, Araraquara, SP 14801-902, Brazil; Neuroscience and Behavioral Institute, Av. do Café, 2.450, 14050-220 Ribeirão Preto, SP, Brazil.
| | - Azair Canto-de-Souza
- Psychobiology Group/Department of Psychology/CECH-UFSCar, São Carlos, SP 13565-905, Brazil; Joint Graduate Program in Physiological Sciences UFSCar/UNESP, Rod. Washington Luís, Km 235, São Carlos, SP 13565-905, Brazil; Graduate Program in Psychology UFSCar, Rod. Washington Luís, Km 235, São Carlos, SP 13565-905, Brazil; Neuroscience and Behavioral Institute, Av. do Café, 2.450, 14050-220 Ribeirão Preto, SP, Brazil.
| |
Collapse
|
15
|
Micheli L, Parisio C, Lucarini E, Vona A, Toti A, Pacini A, Mello T, Boccella S, Ricciardi F, Maione S, Graziani G, Lacal PM, Failli P, Ghelardini C, Di Cesare Mannelli L. VEGF-A/VEGFR-1 signalling and chemotherapy-induced neuropathic pain: therapeutic potential of a novel anti-VEGFR-1 monoclonal antibody. J Exp Clin Cancer Res 2021; 40:320. [PMID: 34649573 PMCID: PMC8515680 DOI: 10.1186/s13046-021-02127-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/04/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Neuropathic pain is a clinically relevant adverse effect of several anticancer drugs that markedly impairs patients' quality of life and frequently leads to dose reduction or therapy discontinuation. The poor knowledge about the mechanisms involved in neuropathy development and pain chronicization, and the lack of effective therapies, make treatment of chemotherapy-induced neuropathic pain an unmet medical need. In this context, the vascular endothelial growth factor A (VEGF-A) has emerged as a candidate neuropathy hallmark and its decrease has been related to pain relief. In the present study, we have investigated the role of VEGF-A and its receptors, VEGFR-1 and VEGFR-2, in pain signalling and in chemotherapy-induced neuropathy establishment as well as the therapeutic potential of receptor blockade in the management of pain. METHODS Behavioural and electrophysiological analyses were performed in an in vivo murine model, by using selective receptor agonists, blocking monoclonal antibodies or siRNA-mediated silencing of VEGF-A and VEGFRs. Expression of VEGF-A and VEGFR-1 in astrocytes and neurons was detected by immunofluorescence staining and confocal microscopy analysis. RESULTS In mice, the intrathecal infusion of VEGF-A (VEGF165 isoforms) induced a dose-dependent noxious hypersensitivity and this effect was mediated by VEGFR-1. Consistently, electrophysiological studies indicated that VEGF-A strongly stimulated the spinal nociceptive neurons activity through VEGFR-1. In the dorsal horn of the spinal cord of animals affected by oxaliplatin-induced neuropathy, VEGF-A expression was increased in astrocytes while VEGFR-1 was mainly detected in neurons, suggesting a VEGF-A/VEGFR-1-mediated astrocyte-neuron cross-talk in neuropathic pain pathophysiology. Accordingly, the selective knockdown of astrocytic VEGF-A by intraspinal injection of shRNAmir blocked the development of oxaliplatin-induced neuropathic hyperalgesia and allodynia. Interestingly, both intrathecal and systemic administration of the novel anti-VEGFR-1 monoclonal antibody D16F7, endowed with anti-angiogenic and antitumor properties, reverted oxaliplatin-induced neuropathic pain. Besides, D16F7 effectively relieved hypersensitivity induced by other neurotoxic chemotherapeutic agents, such as paclitaxel and vincristine. CONCLUSIONS These data strongly support the role of the VEGF-A/VEGFR-1 system in mediating chemotherapy-induced neuropathic pain at the central nervous system level. Thus, treatment with the anti-VEGFR-1 mAb D16F7, besides exerting antitumor activity, might result in the additional advantage of attenuating neuropathic pain when combined with neurotoxic anticancer agents.
Collapse
Affiliation(s)
- Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Carmen Parisio
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Alessia Vona
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Alessandra Toti
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Alessandra Pacini
- Department of Experimental and Clinical Medicine - DMSC - Anatomy and Histology Section, University of Florence, L.go Brambilla 3, 50134, Florence, Italy
| | - Tommaso Mello
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Serena Boccella
- Department of Experimental Medicine, Section of Pharmacology, University of Campania "L. Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| | - Flavia Ricciardi
- Department of Experimental Medicine, Section of Pharmacology, University of Campania "L. Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Section of Pharmacology, University of Campania "L. Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
- I.R.C.S.S., Neuromed, 86077, Pozzilli, Italy
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.
- IDI-IRCCS, Via Monti di Creta 104, 00167, Rome, Italy.
| | | | - Paola Failli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy.
| |
Collapse
|
16
|
The Anti-Arthritic Efficacy of Khellin Loaded in Ascorbyl Decanoate Nanovesicles after an Intra-Articular Administration. Pharmaceutics 2021; 13:pharmaceutics13081275. [PMID: 34452236 PMCID: PMC8399539 DOI: 10.3390/pharmaceutics13081275] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis is the most widespread joint-affecting disease. The management of persistent pain remains inadequate and demands new therapeutic strategies. In this study, we explored the pain relieving and protective properties of a single intra-articular (i.a.) injection of khellin loaded in nanovesicles (K-Ves) based on ascorbyl decanoate plus phosphatidylcholine in a rat model of osteoarthritis (OA) induced by monosodium iodoacetate (MIA) treatment. The developed nanovesicles (approximately 136 nm) had a narrow size distribution (PdI 0.26), a good recovery (about 80%) and a worthy encapsulation efficiency (about 70%) with a ζ-potential of about −40 mV. The stability of K-Ves was assessed in simulated synovial fluid. Seven days after the articular damage with MIA, both K-Ves and a suspension of khellin (K, 50 μL) were i.a. injected. K-Ves significantly counteracted MIA-induced hypersensitivity to mechanical noxious (paw pressure test) and non-noxious stimuli (von Frey test) and significantly reduced the postural unbalance related to spontaneous pain (incapacitance test) and the motor alterations (beam balance test) 7 and 14 days after the i.a. injection. K was partially active only on day 7 after the treatment. The histology emphasized the improvement of several morphological factors in MIA plus K-Ves-treated animals. In conclusion, K-Ves could be successfully used for the local treatment of osteoarthritis.
Collapse
|
17
|
Lee JH, Kim W. Involvement of Serotonergic System in Oxaliplatin-Induced Neuropathic Pain. Biomedicines 2021; 9:970. [PMID: 34440174 PMCID: PMC8394518 DOI: 10.3390/biomedicines9080970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/27/2022] Open
Abstract
Oxaliplatin is a chemotherapeutic agent widely used against colorectal and breast cancers; however, it can also induce peripheral neuropathy that can rapidly occur even after a single infusion in up to 80-90% of treated patients. Numerous efforts have been made to understand the underlying mechanism and find an effective therapeutic agent that could diminish pain without damaging its anti-tumor effect. However, its mechanism is not yet clearly understood. The serotonergic system, as part of the descending pain inhibitory system, has been reported to be involved in different types of pain. The malfunction of serotonin (5-hydroxytryptamine; 5-HT) or its receptors has been associated with the development and maintenance of pain. However, its role in oxaliplatin-induced neuropathy has not been clearly elucidated. In this review, 16 in vivo studies focused on the role of the serotonergic system in oxaliplatin-induced neuropathic pain were analyzed. Five studies analyzed the involvement of 5-HT, while fourteen studies observed the role of its receptors in oxaliplatin-induced allodynia. The results show that 5-HT is not involved in the development of oxaliplatin-induced allodynia, but increasing the activity of the 5-HT1A, 5-HT2A, and 5-HT3 receptors and decreasing the action of 5-HT2C and 5-HT6 receptors may help inhibit pain.
Collapse
Affiliation(s)
| | - Woojin Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea;
| |
Collapse
|
18
|
Rodrigues Tavares LR, Pelarin V, Baptista-de-Souza D, Pereira Ferrari D, Nunes-de-Souza RL, Canto-de-Souza A. 5-HT 3 receptor within the amygdaloid complex modulates pain hypersensitivity induced by empathy model of cohabitation with a partner in chronic pain condition in mice. Soc Neurosci 2021; 16:534-548. [PMID: 34253155 DOI: 10.1080/17470919.2021.1954083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cohabitation with a partner undergoing chronic pain induces pain hypersensitivity. Among a lot of other neurochemical pathways, the serotonin (5-HT) role, specifically the 5-HT3 receptor (5-HT3R), in the amygdala has never been evaluated in this model. Here we studied the effects of the amygdala's chemical inhibition, its neuronal activation pattern, and 5-HT, 5-HIAA, and 5-HT turnover within the amygdala. Furthermore, the systemic and intra-amygdala 5-HT3R activation and blockade in mice that cohabited with a conspecific subjected to chronic constriction injury were investigated. Male Swiss mice were housed in partners for 28 days. The dyads were divided into two groups on the 14th day: cagemate nerve constriction (CNC) and cagemate sham (CS). On the 24th day, cagemates underwent a stereotaxic surgery (when necessary) and, on the 28th day, they were evaluated on the writhing test. The amygdala inactivation promotes pain-hypersensitivity behaviors in groups and dyads; cohabitation with a partner with chronic pain did not change FosB-labeled cells in the amygdala's nucleus and increases 5-HT turnover in cagemates. Systemic and intra-amygdala 5-HT3R activation attenuated and enhanced the number of writhes, respectively. In contrast, 5-HT3R blockade reduced hypersensitivity pain response. Results suggest the involvement of amygdala serotonergic signaling via 5-HT3R in empathy-like behavior.
Collapse
Affiliation(s)
- Lígia Renata Rodrigues Tavares
- Psychobiology Group, Department of Psychology/CECH, Universidade Federal de São Carlos - UFSCar, São Carlos, Brazil.,Joint Graduate Program in Physiological Sciences UFSCar/UNESP, São Carlos, Brazil
| | - Vinícius Pelarin
- Psychobiology Group, Department of Psychology/CECH, Universidade Federal de São Carlos - UFSCar, São Carlos, Brazil.,Joint Graduate Program in Physiological Sciences UFSCar/UNESP, São Carlos, Brazil
| | - Daniela Baptista-de-Souza
- Psychobiology Group, Department of Psychology/CECH, Universidade Federal de São Carlos - UFSCar, São Carlos, Brazil.,Lab. Pharmacology, School of Pharmaceutical Sciences, Univ. Estadual Paulista - UNESP, Araraquara, Brazil.,Neuroscience and Behavior Institute - IneC, Ribeirão Preto, Brazil
| | - Daniele Pereira Ferrari
- Psychobiology Group, Department of Psychology/CECH, Universidade Federal de São Carlos - UFSCar, São Carlos, Brazil
| | - Ricardo Luiz Nunes-de-Souza
- Joint Graduate Program in Physiological Sciences UFSCar/UNESP, São Carlos, Brazil.,Lab. Pharmacology, School of Pharmaceutical Sciences, Univ. Estadual Paulista - UNESP, Araraquara, Brazil.,Neuroscience and Behavior Institute - IneC, Ribeirão Preto, Brazil
| | - Azair Canto-de-Souza
- Psychobiology Group, Department of Psychology/CECH, Universidade Federal de São Carlos - UFSCar, São Carlos, Brazil.,Joint Graduate Program in Physiological Sciences UFSCar/UNESP, São Carlos, Brazil.,Neuroscience and Behavior Institute - IneC, Ribeirão Preto, Brazil.,Program in Psychology UFSCar, São Carlos, Brazil
| |
Collapse
|
19
|
Micheli L, Rajagopalan R, Lucarini E, Toti A, Parisio C, Carrino D, Pacini A, Ghelardini C, Rajagopalan P, Di Cesare Mannelli L. Pain Relieving and Neuroprotective Effects of Non-opioid Compound, DDD-028, in the Rat Model of Paclitaxel-Induced Neuropathy. Neurotherapeutics 2021; 18:2008-2020. [PMID: 34312766 PMCID: PMC8608957 DOI: 10.1007/s13311-021-01069-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 02/04/2023] Open
Abstract
Chemotherapy-induced neuropathy (CIN) is a major dose-limiting side effect of anticancer therapy that can compel therapy discontinuation. Inadequate analgesic efficacy of current pharmacological approaches requires the identification of innovative therapeutics and, hence, the purpose of this study is to conduct a preclinical evaluation of the efficacy of DDD-028, a versatile pentacyclic pyridoindole derivative, against paclitaxel-induced neuropathic pain. In two separate experiments, DDD-028 was administered per os acutely (1-25 mg kg-1) or repeatedly (10 mg kg-1) in paclitaxel-treated rats. The response to mechanical noxious stimulus (paw pressure) as well as to non-noxious mechanical (von Frey) and thermal (cold plate) stimuli was investigated. Acute administration of DDD-028 induced a dose-dependent anti-neuropathic pain effect in all tests performed. Further, repeated daily treatment for 18 consecutive days (starting the first day of paclitaxel administration) significantly reduced the development of pain over time without the development of tolerance to the anti-hyperalgesic effect. Ex vivo analysis showed that DDD-028 was able to reduce oxidative damage of dorsal root ganglia as evidenced by the increase in the level of carbonylated proteins and the decrease in catalase activity. In the lumbar spinal cord, periaqueductal gray matter, thalamus, and somatosensory cortex 1, DDD-28 significantly prevented the activation of microglia and astrocytes. The pharmacodynamic study revealed that the pain-relieving effects of DDD-028 were fully blocked by both the non-selective nicotinic receptor (nAChR) antagonist mecamylamine and by the selective α7 nAChR antagonist methyllycaconitine. In conclusion, DDD-028 was active in reducing paclitaxel-induced neuropathic pain after single or repeated administrations without tolerance development and displaying a double symptomatic and neuroprotective profile. DDD-028 could represent a valuable candidate for the treatment of CIN.
Collapse
Affiliation(s)
- Laura Micheli
- Department of Neuroscience, NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, Psychology, Drug Research and Child HealthViale Pieraccini 6, 50139, Florence, Italy
| | | | - Elena Lucarini
- Department of Neuroscience, NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, Psychology, Drug Research and Child HealthViale Pieraccini 6, 50139, Florence, Italy
| | - Alessandra Toti
- Department of Neuroscience, NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, Psychology, Drug Research and Child HealthViale Pieraccini 6, 50139, Florence, Italy
| | - Carmen Parisio
- Department of Neuroscience, NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, Psychology, Drug Research and Child HealthViale Pieraccini 6, 50139, Florence, Italy
| | - Donatello Carrino
- Department of Experimental and Clinical Medicine, Anatomy and Histology Section, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| | - Alessandra Pacini
- Department of Experimental and Clinical Medicine, Anatomy and Histology Section, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, Psychology, Drug Research and Child HealthViale Pieraccini 6, 50139, Florence, Italy
| | | | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, Psychology, Drug Research and Child HealthViale Pieraccini 6, 50139, Florence, Italy.
| |
Collapse
|
20
|
Therapeutic Agents for Oxaliplatin-Induced Peripheral Neuropathy; Experimental and Clinical Evidence. Int J Mol Sci 2021; 22:1393. [PMID: 33573316 PMCID: PMC7866815 DOI: 10.3390/ijms22031393&set/a 813269399+839900579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Oxaliplatin is an essential drug in the chemotherapy of colorectal, gastric, and pancreatic cancers, but it frequently causes peripheral neuropathy as a dose-limiting factor. So far, animal models of oxaliplatin-induced peripheral neuropathy have been established. The mechanisms of development of neuropathy induced by oxaliplatin have been elucidated, and many drugs and agents have been proven to have neuroprotective effects in basic studies. In addition, some of these drugs have been validated in clinical studies for their inhibitory effects on neuropathy. In this review, we summarize the basic and clinical evidence for the therapeutic effects of oxaliplatin. In basic research, there are many reports of neuropathy inhibitors that target oxidative stress, inflammatory response, sodium channel, transient receptor potential (TRP) channel, glutamate nervous system, and monoamine nervous system. Alternatively, very few drugs have clearly demonstrated the efficacy for oxaliplatin-induced peripheral neuropathy in clinical trials. It is important to activate translational research in order to translate basic research into clinical research.
Collapse
|
21
|
Therapeutic Agents for Oxaliplatin-Induced Peripheral Neuropathy; Experimental and Clinical Evidence. Int J Mol Sci 2021. [DOI: 10.3390/ijms22031393
expr 945913974 + 948698388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Oxaliplatin is an essential drug in the chemotherapy of colorectal, gastric, and pancreatic cancers, but it frequently causes peripheral neuropathy as a dose-limiting factor. So far, animal models of oxaliplatin-induced peripheral neuropathy have been established. The mechanisms of development of neuropathy induced by oxaliplatin have been elucidated, and many drugs and agents have been proven to have neuroprotective effects in basic studies. In addition, some of these drugs have been validated in clinical studies for their inhibitory effects on neuropathy. In this review, we summarize the basic and clinical evidence for the therapeutic effects of oxaliplatin. In basic research, there are many reports of neuropathy inhibitors that target oxidative stress, inflammatory response, sodium channel, transient receptor potential (TRP) channel, glutamate nervous system, and monoamine nervous system. Alternatively, very few drugs have clearly demonstrated the efficacy for oxaliplatin-induced peripheral neuropathy in clinical trials. It is important to activate translational research in order to translate basic research into clinical research.
Collapse
|
22
|
Kawashiri T, Mine K, Kobayashi D, Inoue M, Ushio S, Uchida M, Egashira N, Shimazoe T. Therapeutic Agents for Oxaliplatin-Induced Peripheral Neuropathy; Experimental and Clinical Evidence. Int J Mol Sci 2021; 22:ijms22031393. [PMID: 33573316 PMCID: PMC7866815 DOI: 10.3390/ijms22031393] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/20/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Oxaliplatin is an essential drug in the chemotherapy of colorectal, gastric, and pancreatic cancers, but it frequently causes peripheral neuropathy as a dose-limiting factor. So far, animal models of oxaliplatin-induced peripheral neuropathy have been established. The mechanisms of development of neuropathy induced by oxaliplatin have been elucidated, and many drugs and agents have been proven to have neuroprotective effects in basic studies. In addition, some of these drugs have been validated in clinical studies for their inhibitory effects on neuropathy. In this review, we summarize the basic and clinical evidence for the therapeutic effects of oxaliplatin. In basic research, there are many reports of neuropathy inhibitors that target oxidative stress, inflammatory response, sodium channel, transient receptor potential (TRP) channel, glutamate nervous system, and monoamine nervous system. Alternatively, very few drugs have clearly demonstrated the efficacy for oxaliplatin-induced peripheral neuropathy in clinical trials. It is important to activate translational research in order to translate basic research into clinical research.
Collapse
Affiliation(s)
- Takehiro Kawashiri
- Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.M.); (D.K.); (M.I.); (T.S.)
- Correspondence: ; Tel.: +81-92-642-6573
| | - Keisuke Mine
- Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.M.); (D.K.); (M.I.); (T.S.)
| | - Daisuke Kobayashi
- Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.M.); (D.K.); (M.I.); (T.S.)
| | - Mizuki Inoue
- Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.M.); (D.K.); (M.I.); (T.S.)
| | - Soichiro Ushio
- Department of Pharmacy, Okayama University Hospital, Okayama 700-8558, Japan;
| | - Mayako Uchida
- Education and Research Center for Clinical Pharmacy, Osaka University of Pharmaceutical Sciences, Osaka 569-1094, Japan;
| | - Nobuaki Egashira
- Department of Pharmacy, Kyushu University Hospital, Fukuoka 812-8582, Japan;
| | - Takao Shimazoe
- Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.M.); (D.K.); (M.I.); (T.S.)
| |
Collapse
|
23
|
Micov AM, Tomić MA, Todorović MB, Vuković MJ, Pecikoza UB, Jasnic NI, Djordjevic JD, Stepanović-Petrović RM. Vortioxetine reduces pain hypersensitivity and associated depression-like behavior in mice with oxaliplatin-induced neuropathy. Prog Neuropsychopharmacol Biol Psychiatry 2020; 103:109975. [PMID: 32464241 DOI: 10.1016/j.pnpbp.2020.109975] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/11/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023]
Abstract
Chronic pain and depression commonly occur together so dual-acting agents might be particularly useful. The population of patients with chemotherapy-induced neuropathy is increasing in parallel with the increase of population of cancer survivors and there is a compelling need for satisfactory treatment of symptoms of neuropathy and concomitant depression. We examined the effects of vortioxetine, a novel antidepressant with unique mechanism of action, on pain hypersensitivity and depression-like behavior in oxaliplatin-induced neuropathy model in mice (OIPN). Vortioxetine (1-10 mg/kg, p.o.) significantly and dose-dependently reduced mechanical allodynia in von Frey test and cold allodynia in acetone test in OIPN mice, in both repeated prophylactic and acute therapeutic treatment regimens. It also reduced depression-like behavior in the forced swimming test in OIPN mice, in both treatment paradigms. Its antiallodynic and antidepressive-like effects were comparable to those exerted by duloxetine (1-15 mg/kg, p.o.). The antiallodynic and antidepressive-like effects of repeatedly administered vortioxetine might be related to the increased content of 5-hydroxytryptamine (5-HT) and noradrenaline (NA), detected in the brainstem of treated OIPN mice. These results indicate that vortioxetine could be potentially useful in prevention and treatment of chemotherapy-induced neuropathy, for the relief of pain and concomitant depressive symptoms. It should be further tested to this regard in clinical settings.
Collapse
Affiliation(s)
- Ana M Micov
- University of Belgrade - Faculty of Pharmacy, Department of Pharmacology, Belgrade, Serbia
| | - Maja A Tomić
- University of Belgrade - Faculty of Pharmacy, Department of Pharmacology, Belgrade, Serbia.
| | - Marija B Todorović
- University of Belgrade - Faculty of Pharmacy, Department of Pharmacology, Belgrade, Serbia
| | - Milja J Vuković
- University of Belgrade - Faculty of Pharmacy, Department of Pharmacology, Belgrade, Serbia
| | - Uroš B Pecikoza
- University of Belgrade - Faculty of Pharmacy, Department of Pharmacology, Belgrade, Serbia
| | - Nebojsa I Jasnic
- University of Belgrade - Faculty of Biology, Institute of Physiology and Biochemistry "Ivan Djaja", Belgrade, Serbia
| | - Jelena D Djordjevic
- University of Belgrade - Faculty of Biology, Institute of Physiology and Biochemistry "Ivan Djaja", Belgrade, Serbia
| | | |
Collapse
|
24
|
Bouchenaki H, Danigo A, Sturtz F, Hajj R, Magy L, Demiot C. An overview of ongoing clinical trials assessing pharmacological therapeutic strategies to manage chemotherapy-induced peripheral neuropathy, based on preclinical studies in rodent models. Fundam Clin Pharmacol 2020; 35:506-523. [PMID: 33107619 DOI: 10.1111/fcp.12617] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 12/22/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a major dose-limiting side effect induced by a variety of chemotherapeutic agents. Symptoms are mainly sensory: pain, tingling, numbness, and temperature sensitivity. They may require the tapering of chemotherapy regimens or even their cessation; thus, the prevention/treatment of CIPN is critical to increase effectiveness of cancer treatment. However, CIPN management is mainly based on conventional neuropathic pain treatments, with poor clinical efficacy. Therefore, significant effort is made to identify new pharmacological targets to prevent/treat CIPN. Animal modeling is a key component in predicting human response to drugs and in understanding the pathophysiological mechanisms underlying CIPN. In fact, studies performed in rodents highlighted several pharmacological targets to treat/prevent CIPN. This review provides updated information about ongoing clinical trials testing drugs for the management of CIPN and presents some of their proof-of-concept studies conducted in rodent models. The presented drugs target oxidative stress, renin-angiotensin system, glutamatergic neurotransmission, sphingolipid metabolism, neuronal uptake transporters, nicotinamide adenine dinucleotide metabolism, endocannabinoid system, transient receptor potential channels, and serotoninergic receptors. As some clinical trials focus on the effect of the drugs on pain, others evaluate their efficacy by assessing general neuropathy. Moreover, based on studies conducted in rodent models, it remains unclear if some of the tested drugs act in an antinociceptive fashion or have neuroprotective properties. Thus, further investigations are needed to understand their mechanism of action, as well as a global standardization of the methods used to assess efficacy of new therapeutic strategies in the treatment of CIPN.
Collapse
Affiliation(s)
- Hichem Bouchenaki
- EA 6309 - Myelin Maintenance & Peripheral Neuropathy, Faculties of Medicine and Pharmacy, University of Limoges, Limoges, France.,Pharnext SA, Issy-les-Moulineaux, France
| | - Aurore Danigo
- EA 6309 - Myelin Maintenance & Peripheral Neuropathy, Faculties of Medicine and Pharmacy, University of Limoges, Limoges, France
| | - Franck Sturtz
- EA 6309 - Myelin Maintenance & Peripheral Neuropathy, Faculties of Medicine and Pharmacy, University of Limoges, Limoges, France
| | | | - Laurent Magy
- Department of Neurology, Reference Center for Rare Peripheral Neuropathies, University Hospital of Limoges, Limoges, France
| | - Claire Demiot
- EA 6309 - Myelin Maintenance & Peripheral Neuropathy, Faculties of Medicine and Pharmacy, University of Limoges, Limoges, France
| |
Collapse
|
25
|
Liu X, Wang G, Ai G, Xu X, Niu X, Zhang M. Selective Ablation of Descending Serotonin from the Rostral Ventromedial Medulla Unmasks Its Pro-Nociceptive Role in Chemotherapy-Induced Painful Neuropathy. J Pain Res 2020; 13:3081-3094. [PMID: 33262643 PMCID: PMC7700091 DOI: 10.2147/jpr.s275254] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/22/2020] [Indexed: 12/22/2022] Open
Abstract
Purpose Chemotherapy-induced painful neuropathy (CIPN) is a severe adverse effect of many anti-neoplastic drugs that is difficult to manage. Serotonin (5-hydroxytryptamine, 5-HT) is an important neurotransmitter in the rostral ventromedial medulla (RVM), which modulates descending spinal nociceptive transmission. However, the influence of the descending 5-HT from the RVM on CIPN is poorly understood. We investigated the role of 5-HT released from descending RVM neurons in a paclitaxel-induced CIPN rat model. Methods CIPN rat model was produced by intraperitoneally injecting of paclitaxel. Pain behavioral assessments included mechanical allodynia and heat hyperalgesia. 5-HT content was analyzed by high-performance liquid chromatography (HPLC). Western blot and immunohistochemistry were used to determine tryptophan hydroxylase (Tph) and c-Fos expression. The inhibitors p-chlorophenylalanine (PCPA) and SB203580 were administrated by stereotaxical RVM microinjection. Ondansetron was injected through intrathecal catheterization. Results The results demonstrated that Tph, the rate-limiting enzyme in 5-HT synthesis, was significantly upregulated in the RVM, and that spinal 5-HT release was increased in CIPN rats. Intra-RVM microinjection of Tph inhibitor PCPA significantly attenuated mechanical and thermal pain behavior through Tph downregulation and decreased spinal 5-HT. Intra-RVM administration of p38 mitogen-activated protein kinase (p38 MAPK) inhibitor SB203580 alleviated paclitaxel-induced pain in a similar manner to PCPA. Intrathecal injection of ondansetron, a 5-HT3 receptor antagonist, partially reversed paclitaxel-induced pain, indicating that 5-HT3 receptors were involved in descending serotoninergic modulation of spinal pain processing. Conclusion The results suggest that activation of the p38 MAPK pathway in the RVM leads to increased RVM Tph expression and descending serotoninergic projection to the spinal dorsal horn and contributes to the persistence of CIPN via spinal 5-HT3 receptors.
Collapse
Affiliation(s)
- Xijiang Liu
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, People's Republic of China
| | - Gongming Wang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, People's Republic of China
| | - Geyi Ai
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, People's Republic of China
| | - Xiqiang Xu
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, People's Republic of China
| | - Xinhuan Niu
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, People's Republic of China
| | - Mengyuan Zhang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, People's Republic of China
| |
Collapse
|
26
|
The active second-generation proteasome inhibitor oprozomib reverts the oxaliplatin-induced neuropathy symptoms. Biochem Pharmacol 2020; 182:114255. [PMID: 33010214 DOI: 10.1016/j.bcp.2020.114255] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/28/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
Oxaliplatin-induced neuropathy (OXAIN) is a major adverse effect of this antineoplastic drug, widely used in the treatment of colorectal cancer. Although its molecular mechanisms remain poorly understood, recent evidence suggest that maladaptive neuroplasticity and oxidative stress may participate to the development of this neuropathy. Given the role played on protein remodeling by ubiquitin-proteasome system (UPS) in response to oxidative stress and in neuropathic pain, we investigated whether oxaliplatin might cause alterations in the UPS-mediated degradation pathway, in order to identify new pharmacological tools useful in OXAIN. In a rat model of OXAIN (2.4 mg kg-1 i.p., daily for 10 days), a significant increase in chymotrypsin-(β5) like activity of the constitutive proteasome 26S was observed in the thalamus (TH) and somatosensory cortex (SSCx). In addition, the selective up-regulation of β5 and LMP7 (β5i) subunit gene expression was assessed in the SSCx. Furthermore, this study revealed that oprozomib, a selective β5 subunit proteasome inhibitor, is able to normalize the spinal prodynorphin gene expression upregulation induced by oxaliplatin, as well as to revert mechanical allodynia and thermal hyperalgesia observed in oxaliplatin-treated rats. These results underline the relevant role of UPS in the OXAIN and suggest new pharmacological targets to counteract this severe adverse effect. This preclinical study reveals the involvement of the proteasome in the oxaliplatin-induced neuropathy and adds useful information to better understand the molecular mechanism underlying this pain condition. Moreover, although further evidence is required, these findings suggest that oprozomib could be a therapeutic option to counteract chemotherapy-induced neuropathy.
Collapse
|
27
|
Micheli L, Di Cesare Mannelli L, Del Bello F, Giannella M, Piergentili A, Quaglia W, Carrino D, Pacini A, Ghelardini C. The Use of the Selective Imidazoline I 1 Receptor Agonist Carbophenyline as a Strategy for Neuropathic Pain Relief: Preclinical Evaluation in a Mouse Model of Oxaliplatin-Induced Neurotoxicity. Neurotherapeutics 2020; 17:1005-1015. [PMID: 32572830 PMCID: PMC7609613 DOI: 10.1007/s13311-020-00873-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Anti-cancer therapy based on the repeated administration of oxaliplatin is limited by the development of a disabling neuropathic syndrome with detrimental effects on the patient's quality of life. The lack of effective pharmacological approaches calls for the identification of innovative therapeutic strategies based on new targets. We focused our attention on the imidazoline I1 receptor (I1-R) and in particular on the selective I1-R agonist 2-(1-([1,1'-biphenyl]-2-yl)propan-2-yl)-4,5-dihydro-1H-imidazole) (carbophenyline). The purpose of this work was the preclinical evaluation of the efficacy of carbophenyline on oxaliplatin-induced neuropathic pain in mice. Carbophenyline, acutely per os administered (0.1-10 mg kg-1), induced a dose-dependent anti-hyperalgesic effect that was completely blocked by the pre-treatment with the I1-R antagonist 3 or the I1/α2 receptor antagonist efaroxan, confirming the I1-R-dependent mechanism. Conversely, pre-treatment with the I2-R antagonist BU224 did not block the anti-nociceptive effect evoked by carbophenyline. Repeated oral administrations of carbophenyline (1 mg kg-1) for 14 days, starting from the first day of oxaliplatin injection, counteracted the development of neuropathic pain in all behavioral tests (cold plate, Von Frey, and paw pressure tests) carried out 24 h after the last carbophenyline treatment on days 7 and 14. In the dorsal horn of the spinal cord, carbophenyline significantly decreased the oxaliplatin-induced astrocyte activation detected by immunofluorescence staining by the specific labelling with GFAP antibody. In conclusion, carbophenyline showed anti-neuropathic properties both after acute and chronic treatment with preventive effect against oxaliplatin-induced astrocyte activation in the spinal cord. Therefore, I1-R agonists emerge as a new class of candidates for the management of oxaliplatin-induced neuropathic pain.
Collapse
Affiliation(s)
- Laura Micheli
- Dept. of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale Gaetano Pieraccini 6, 50139, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Dept. of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale Gaetano Pieraccini 6, 50139, Florence, Italy.
| | - Fabio Del Bello
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032, Camerino, Italy
| | - Mario Giannella
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032, Camerino, Italy
| | - Alessandro Piergentili
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032, Camerino, Italy
| | - Wilma Quaglia
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032, Camerino, Italy
| | - Donatello Carrino
- Department of Experimental and Clinical Medicine, Anatomy and Histology Section, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| | - Alessandra Pacini
- Department of Experimental and Clinical Medicine, Anatomy and Histology Section, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| | - Carla Ghelardini
- Dept. of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale Gaetano Pieraccini 6, 50139, Florence, Italy
| |
Collapse
|
28
|
Intra-Articular Route for the System of Molecules 14G1862 from Centella Asiatica: Pain Relieving and Protective Effects in a Rat Model of Osteoarthritis. Nutrients 2020; 12:nu12061618. [PMID: 32486519 PMCID: PMC7352185 DOI: 10.3390/nu12061618] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/15/2020] [Accepted: 05/27/2020] [Indexed: 12/20/2022] Open
Abstract
Current pharmacological therapies for the management of chronic articular diseases are far from being satisfactory, so new strategies need to be investigated. We tested the intra-articular pain relieving properties of a system of molecules from a characterized Centella asiatica extract (14G1862) in a rat model of osteoarthritis induced by monoiodoacetate (MIA). 14G1862 (0.2–2 mg mL−1) was intra-articularly (i.a.) injected 7 days after MIA, behavioural and histological evaluations were performed 14, 30 and 60 days after treatments. Moreover, the effect of 14G1862 on nitrate production and iNOS expression in RAW 264.7 macrophages stimulated with LPS was assessed. In vitro, 14G1862 treatment attenuated LPS-induced NO production and iNOS expression in a comparable manner to celecoxib. In vivo, 14G1862 significantly reduced mechanical allodynia and hyperalgesia, spontaneous pain and motor alterations starting on day 14 up to day 60. The efficacy was higher or comparable to that evoked by triamcinolone acetonide (100 μg i.a.) used as reference drug. Histological evaluation highlighted the improvement of several morphological parameters in MIA + 14G1862-treated animals with particularly benefic effects on joint space and fibrin deposition. In conclusion, i.a. treatment with Centella asiatica is a candidate to be a novel effective approach for osteoarthritis therapy.
Collapse
|
29
|
Nocentini A, Alterio V, Bua S, Micheli L, Esposito D, Buonanno M, Bartolucci G, Osman SM, ALOthman ZA, Cirilli R, Pierini M, Monti SM, Di Cesare Mannelli L, Gratteri P, Ghelardini C, De Simone G, Supuran CT. Phenyl(thio)phosphon(amid)ate Benzenesulfonamides as Potent and Selective Inhibitors of Human Carbonic Anhydrases II and VII Counteract Allodynia in a Mouse Model of Oxaliplatin-Induced Neuropathy. J Med Chem 2020; 63:5185-5200. [PMID: 32364386 PMCID: PMC8007106 DOI: 10.1021/acs.jmedchem.9b02135] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Indexed: 12/13/2022]
Abstract
Human carbonic anhydrase (CA; EC 4.2.1.1) isoforms II and VII are implicated in neuronal excitation, seizures, and neuropathic pain (NP). Their selective inhibition over off-target CAs is expected to produce an anti-NP action devoid of side effects due to promiscuous CA modulation. Here, a drug design strategy based on the observation of (dis)similarities between the target CA active sites was planned with benzenesulfonamide derivatives and, for the first time, a phosphorus-based linker. Potent and selective CA II/VII inhibitors were identified among the synthesized phenyl(thio)phosphon(amid)ates 3-22. X-ray crystallography depicted the binding mode of phosphonic acid 3 to both CAs II and VII. The most promising derivatives, after evaluation of their stability in acidic media, were tested in a mouse model of oxaliplatin-induced neuropathy. The most potent compound racemic mixture was subjected to HPLC enantioseparation, and the identification of the eutomer, the (S)-enantiomer, allowed to halve the dose totally relieving allodynia in mice.
Collapse
Affiliation(s)
- Alessio Nocentini
- Department
of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Vincenzo Alterio
- Istituto
di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Silvia Bua
- Department
of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Laura Micheli
- Department
of NEUROFARBA, Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Davide Esposito
- Istituto
di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Martina Buonanno
- Istituto
di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Gianluca Bartolucci
- Department
of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Sameh M. Osman
- Chemistry
Department, College of Science, King Saud
University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Zeid A. ALOthman
- Chemistry
Department, College of Science, King Saud
University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Roberto Cirilli
- Centro
nazionale per il controllo e la valutazione dei farmaci, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Marco Pierini
- Dipartimento
di Chimica e Tecnologie del Farmaco, Sapienza
University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Simona Maria Monti
- Istituto
di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Lorenzo Di Cesare Mannelli
- Department
of NEUROFARBA, Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Paola Gratteri
- Department
of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Carla Ghelardini
- Department
of NEUROFARBA, Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Giuseppina De Simone
- Istituto
di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Claudiu T. Supuran
- Department
of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
30
|
Liu QQ, Yao XX, Gao SH, Li R, Li BJ, Yang W, Cui RJ. Role of 5-HT receptors in neuropathic pain: potential therapeutic implications. Pharmacol Res 2020; 159:104949. [PMID: 32464329 DOI: 10.1016/j.phrs.2020.104949] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 10/24/2022]
Abstract
5-HT plays a crucial role in the progress and adjustment of pain both centrally and peripherally. The therapeutic action of the 5-HT receptors` agonist and antagonist in neuropathic pain have been widely reported in many studies. However, the specific roles of 5-HT subtype receptors have not been reviewed comprehensively. Therefore, we summarized the recent findings on multiple subtypes of 5-HT receptors in both central and peripheral nervous system in neuropathic pain, particularly, 5-HT1, 5-HT2, 5-HT3 and 5-HT7 receptors. In addition, 5-HT4, 5-HT5 and 5-HT6 receptors were also reviewed. Most of studies focused on the function of 5-HT subtype receptors in spinal level compared to brain areas. Based on these evidences, the pain process can be facilitated or inhibited that depending on the specific subtypes and the distribution of 5-HT receptors. Therefore, this review may provide potential therapeutic implications in treatment of neuropathic pain.
Collapse
Affiliation(s)
- Qian Qian Liu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China; Hand Surgery Department, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiao Xiao Yao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Shuo Hui Gao
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Rui Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China; Hand Surgery Department, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Bing Jin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ran Ji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
31
|
Baptista-de-Souza D, Tavares LRR, Furuya-da-Cunha EM, Carneiro de Oliveira PE, Canto-de-Souza L, Nunes-de-Souza RL, Canto-de-Souza A. Chronic Fluoxetine Impairs the Effects of 5-HT 1A and 5-HT 2C Receptors Activation in the PAG and Amygdala on Antinociception Induced by Aversive Situation in Mice. Front Pharmacol 2020; 11:260. [PMID: 32218734 PMCID: PMC7078365 DOI: 10.3389/fphar.2020.00260] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/24/2020] [Indexed: 11/24/2022] Open
Abstract
Growing evidence suggests an important role of fluoxetine with serotonin 5-HT1A and 5-HT2C receptors in the modulation of emotion and nociception in brain areas such as the amygdala and periaqueductal gray (PAG). Acute fluoxetine impairs 5-HT2C (but not 5-HT1A) receptor activation in the amygdaloid complex. Given that fluoxetine produces its clinical therapeutic effects only when given chronically, this study investigated the effects of chronic treatment with fluoxetine on the effects produced by 5-HT1A or 5-HT2C receptors activation in the amygdala or PAG on fear-induced antinociception. We recorded the effects of chronic fluoxetine on serotonin and its metabolite 5-hydroxyindoleacetic acid (5-HIAA) levels as well as serotonin turnover; 5-HT1A and 5-HT2C receptor protein levels in the amygdala and PAG. Also, we evaluated the effects of chronic fluoxetine combined with intra-amygdala or intra-PAG injection of MK-212 (a 5-HT2C agonist; 0.63 nmol) or 8-OH-DPAT (a 5-HT1A agonist; 10 nmol) on the antinociceptive response in mice confined in the open arm of the elevated plus-maze (EPM). Nociception was assessed with the writhing test induced by intraperitoneal injection of 0.6% acetic acid. Results showed that fluoxetine (20 mg/kg, s.c.) enhanced the open-arm induced antinociception (OAA) and reduced the number of writhes in mice confined in the enclosed arm, featuring an analgesic effect. In addition, fluoxetine increased the expression of 5-HT2C receptors and 5-HT levels whereas reduced its turnover in the amygdala. While fluoxetine did not change 5-HT and 5-HIAA levels, and its turnover in the PAG, it up-regulated 5HT1A and 5-HT2C receptors in this midbrain area. Chronic fluoxetine (5.0 mg/Kg, an intrinsically inactive dose on nociception) antagonized the enhancement of OAA produced by intra-amygdala or intra-PAG injection of MK-212. Fluoxetine also impaired the attenuation of OAA induced by intra-amygdala injection of 8-OH-DPAT and totally prevented OAA in mice that received intra-PAG 8-OH-DPAT. These results suggest that (i) 5-HT may facilitate nociception and intensify OAA, acting at amygdala 5-HT1A and 5-HT2C receptors, respectively, and (ii) fluoxetine modulates the OAA through activation of 5-HT2C receptors within the PAG. These findings indicate that chronic fluoxetine impairs the effects of 5-HT1A and 5-HT2C receptors activation in the amygdala and PAG on fear-induced antinociception in mice.
Collapse
Affiliation(s)
- Daniela Baptista-de-Souza
- Department of Psychology, Federal University of São Carlos-UFSCar, São Carlos, Brazil.,Joint Graduate Program in Physiological Sciences, UFSCar/UNESP, São Carlos, Brazil.,Institute of Neuroscience and Behavior, Ribeirão Preto, Brazil
| | - Lígia Renata Rodrigues Tavares
- Department of Psychology, Federal University of São Carlos-UFSCar, São Carlos, Brazil.,Joint Graduate Program in Physiological Sciences, UFSCar/UNESP, São Carlos, Brazil
| | - Elke Mayumi Furuya-da-Cunha
- Department of Psychology, Federal University of São Carlos-UFSCar, São Carlos, Brazil.,Joint Graduate Program in Physiological Sciences, UFSCar/UNESP, São Carlos, Brazil
| | - Paulo Eduardo Carneiro de Oliveira
- Department of Psychology, Federal University of São Carlos-UFSCar, São Carlos, Brazil.,Joint Graduate Program in Physiological Sciences, UFSCar/UNESP, São Carlos, Brazil
| | - Lucas Canto-de-Souza
- Institute of Neuroscience and Behavior, Ribeirão Preto, Brazil.,Laboratory of Pharmacology, School of Pharmaceutical Sciences, Univ. Estadual Paulista - UNESP, Araraquara, Brazil
| | - Ricardo Luiz Nunes-de-Souza
- Joint Graduate Program in Physiological Sciences, UFSCar/UNESP, São Carlos, Brazil.,Institute of Neuroscience and Behavior, Ribeirão Preto, Brazil.,Laboratory of Pharmacology, School of Pharmaceutical Sciences, Univ. Estadual Paulista - UNESP, Araraquara, Brazil
| | - Azair Canto-de-Souza
- Department of Psychology, Federal University of São Carlos-UFSCar, São Carlos, Brazil.,Joint Graduate Program in Physiological Sciences, UFSCar/UNESP, São Carlos, Brazil.,Institute of Neuroscience and Behavior, Ribeirão Preto, Brazil.,Graduate Program in Psychology UFSCar, São Carlos, Brazil
| |
Collapse
|
32
|
Sierra S, Lippold KM, Stevens DL, Poklis JL, Dewey WL, González-Maeso J. Adjunctive effect of the serotonin 5-HT 2C receptor agonist lorcaserin on opioid-induced antinociception in mice. Neuropharmacology 2020; 167:107949. [PMID: 31987863 DOI: 10.1016/j.neuropharm.2020.107949] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/11/2019] [Accepted: 01/08/2020] [Indexed: 12/25/2022]
Abstract
Opioid-sparing adjuncts are treatments that aim to reduce the overall dose of opioids needed to achieve analgesia, hence decreasing the burden of side effects through alternative mechanisms of action. Lorcaserin is a serotonin 5-HT2C receptor (5-HT2CR) agonist that has recently been reported to reduce abuse-related effects of the opioid analgesic oxycodone. The goal of our studies was to evaluate the effects of adjunctive lorcaserin on opioid-induced analgesic-like behavior using the tail-flick reflex (TFR) test as a mouse model of acute thermal nociception. We show that whereas subcutaneous (s.c.) administration of lorcaserin alone was inactive on the TFR test, adjunctive lorcaserin (s.c.) significantly increased the potency of oxycodone as an antinociceptive drug. This effect was prevented by the 5-HT2CR antagonist SB242084. A similar lorcaserin (s.c.)-induced adjunctive phenotype was observed upon administration of the opioid analgesics morphine and fentanyl. Remarkably, we also show that, opposite to the effects observed via s.c. administration, intrathecal (i.t.) administration of lorcaserin alone induced antinociceptive TFR behavior, an effect that was not prevented by the opioid receptor antagonist naloxone. This route of administration (i.t.) also led to a significant augmentation of oxycodone-induced antinociception. Lorcaserin (s.c.) did not alter the brain or blood concentrations of oxycodone, which suggests that its adjunctive effects on opioid-induced antinociception do not depend upon changes in opioid metabolism. Together, these data indicate that lorcaserin-mediated activation of the 5-HT2CR may represent a new pharmacological approach to augment opioid-induced antinociception. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.
Collapse
Affiliation(s)
- Salvador Sierra
- Department of Physiology & Biophysics Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Kumiko M Lippold
- Department of Pharmacology & Toxicology Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - David L Stevens
- Department of Pharmacology & Toxicology Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Justin L Poklis
- Department of Pharmacology & Toxicology Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - William L Dewey
- Department of Pharmacology & Toxicology Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Javier González-Maeso
- Department of Physiology & Biophysics Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA.
| |
Collapse
|
33
|
Valproate reverses stress-induced somatic hyperalgesia and visceral hypersensitivity by up-regulating spinal 5-HT 2C receptor expression in female rats. Neuropharmacology 2019; 165:107926. [PMID: 31883927 DOI: 10.1016/j.neuropharm.2019.107926] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 11/15/2019] [Accepted: 12/20/2019] [Indexed: 12/15/2022]
Abstract
Sodium valproate (VPA) has analgesic effects in clinical and experimental studies, but the mechanisms are still unclear. The present study examined the effects of VPA on stress-induced somatic hyperalgesia and visceral hypersensitivity and the role of 5-HT2C receptors in the spinal cord. Repeated 3 day forced swim (FS) significantly reduced the thermal withdrawal latency and mechanical withdrawal threshold, and increased the magnitude of the visceromotor response to colorectal distention compared to the baseline values in rats. The somatic hyperalgesia and visceral hypersensitivity were accompanied by significant down-regulation of 5-HT2C receptor expression in the L4-L5 and L6-S1 dorsal spinal cord. Intraperitoneal administration of VPA (300 mg/kg) before each FS and 1 day post FS prevented the development of somatic hyperalgesia and visceral hypersensitivity induced by FS stress, as well as down-regulation of 5-HT2C receptors in the spinal cord. The reversal of somatic hyperalgesia and visceral hypersensitivity by VPA in FS rats was blocked by intrathecal administration of the selective 5-HT2C receptor antagonist RS-102221 (30 μg/10 μL) 30 min after each VPA injection. The results suggest that VPA attenuates FS-induced somatic hyperalgesia and visceral hypersensitivity by restoring down-regulated function of 5-HT2C receptors in the spinal cord.
Collapse
|
34
|
Phaseolus vulgaris L. Extract: Alpha-Amylase Inhibition against Metabolic Syndrome in Mice. Nutrients 2019; 11:nu11081778. [PMID: 31374931 PMCID: PMC6723332 DOI: 10.3390/nu11081778] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 01/07/2023] Open
Abstract
To examine the effects of the alpha-amylase inhibitor isoform 1 called phaseolamin, a standardized extract from white kidney beans (Phaseolus vulgaris L.) was tested against the hallmarks of metabolic syndrome. The efficacy of a per os repeated treatment with P. vulgaris extract (500 mg/kg) was compared with metformin (100 mg/kg) and atorvastatin (10 mg/kg) in a model of metabolic syndrome evoked by prolonged high fat diet (HFD; week 1 to week 19) in C57BL/6 mice. Bean extract and compounds administration started after metabolic syndrome establishment (week 11). P. vulgaris extract reduced the body weight overtime, as well as effectively lowered glycaemia, triglycerides, and cholesterol. On week 19, bean extract normalized the HFD-evoked tolerance to glucose and insulin. According to the phytochemical characterization, it inhibited the alpha-amylase activity. Animals treated with the extract were rescued from motor impairments and nociceptive threshold alterations induced by HFD. Specific organs analysis revealed that P. vulgaris extract decreased hepatic steatosis and lipid peroxidation in liver. It protected the heart from HFD oxidative alterations increasing the expression of the detoxifying enzymes catalase and glutathione reductase, and normalizing NADH dehydrogenase level. The histological analysis of aorta showed a protection about the development of fatty streaks in the muscular layers. In conclusion, a prolonged treatment with the standardized extract of P. vulgaris significantly reduced several pathological features related to a metabolic syndrome-like condition; a multifactorial approach that candidates this vegetal product as a possible therapeutic option against metabolic syndrome.
Collapse
|
35
|
Baptista-de-Souza D, Pelarin V, Canto-de-Souza L, Nunes-de-Souza RL, Canto-de-Souza A. Interplay between 5-HT 2C and 5-HT 1A receptors in the dorsal periaqueductal gray in the modulation of fear-induced antinociception in mice. Neuropharmacology 2018; 140:100-106. [PMID: 30056125 DOI: 10.1016/j.neuropharm.2018.07.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/05/2018] [Accepted: 07/23/2018] [Indexed: 11/27/2022]
Abstract
The confinement of rodents to the open arm of the elevated-plus maze provokes antinociception (OAA). As a type of defensive reaction, the OAA has been investigated through systemic and intramesencephalic (e.g., dorsal portion of the periaqueductal gray - dPAG) injections of anxiolytic-like drugs [e.g., serotonergic (5-HT) receptor agonists or antagonists]. Here we investigated the effects of (i) intra-dPAG injections of a 5HT2C receptor agonist (MK-212; 0.21 or 0.63 nmol) and antagonist (SB 242084; 0.01, 0.1 or 1.0 nmol); (ii) combined injections of SB 242084 and MK-212 into the dPAG; (iii) combined injections of SB 242084 with 8-OHDPAT (10 nmol) into the dPAG on the OAA in male Swiss mice. Nociception was assessed with the writhing test induced by acetic acid injection. Results showed that (i) intra-dPAG injection of MK-212 (0.63 nmol) increased the OAA; (ii) intra-dPAG SB 242084 (1.0 nmol) prevented the OAA; (iii) SB 242084 (0.1 nmol, a dose devoid of intrinsic effect on nociception) blocked the OAA enhancement provoked by MK-212 and enabled 8-OH-DPAT to prevent the OAA. These results suggest that OAA is mediated by 5-HT2C receptors within the dPAG. Intra-dPAG SB242084 administration provoked similar results on the effects produced by MK-212 and 8-OH-DPAT on OAA. In addition, the dPAG 5-HT1A and 5-HT2C receptors interact each other in the modulation of OAA.
Collapse
Affiliation(s)
- Daniela Baptista-de-Souza
- Dept. Psychology, Federal University of São Carlos-UFSCar, São Carlos, SP, 13565-905, Brazil; Joint Graduate Program in Physiological Sciences UFSCar/UNESP, São Carlos, SP, 13565-905, Brazil; Institute of Neuroscience and Behavior, Av. Do Café, 2.450, 14050-220, Ribeirão Preto, SP, Brazil
| | - Vinícius Pelarin
- Dept. Psychology, Federal University of São Carlos-UFSCar, São Carlos, SP, 13565-905, Brazil; Joint Graduate Program in Physiological Sciences UFSCar/UNESP, São Carlos, SP, 13565-905, Brazil
| | - Lucas Canto-de-Souza
- Lab. Pharmacology, School of Pharmaceutical Sciences, Univ. Estadual Paulista - UNESP, Araraquara, SP, 14800-903, Brazil; Institute of Neuroscience and Behavior, Av. Do Café, 2.450, 14050-220, Ribeirão Preto, SP, Brazil
| | - Ricardo Luiz Nunes-de-Souza
- Lab. Pharmacology, School of Pharmaceutical Sciences, Univ. Estadual Paulista - UNESP, Araraquara, SP, 14800-903, Brazil; Joint Graduate Program in Physiological Sciences UFSCar/UNESP, São Carlos, SP, 13565-905, Brazil; Institute of Neuroscience and Behavior, Av. Do Café, 2.450, 14050-220, Ribeirão Preto, SP, Brazil
| | - Azair Canto-de-Souza
- Dept. Psychology, Federal University of São Carlos-UFSCar, São Carlos, SP, 13565-905, Brazil; Joint Graduate Program in Physiological Sciences UFSCar/UNESP, São Carlos, SP, 13565-905, Brazil; Graduate Program in Psychology UFSCar, Rod. Washington Luís, Km 235, São Carlos, SP, 13565-905, Brazil; Institute of Neuroscience and Behavior, Av. Do Café, 2.450, 14050-220, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
36
|
Tavares LRR, Baptista-de-Souza D, Canto-de-Souza A. Activation of 5-HT2C (but not 5-HT1A) receptors in the amygdala enhances fear-induced antinociception: Blockade with local 5-HT2C antagonist or systemic fluoxetine. Neuropharmacology 2018; 135:376-385. [DOI: 10.1016/j.neuropharm.2018.03.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 03/05/2018] [Accepted: 03/06/2018] [Indexed: 12/28/2022]
|
37
|
Mascarenhas DC, Gomes KS, Sorregotti T, Nunes-de-Souza RL. Blockade of Cannabinoid CB1 Receptors in the Dorsal Periaqueductal Gray Unmasks the Antinociceptive Effect of Local Injections of Anandamide in Mice. Front Pharmacol 2017; 8:695. [PMID: 29046638 PMCID: PMC5632997 DOI: 10.3389/fphar.2017.00695] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 09/19/2017] [Indexed: 12/12/2022] Open
Abstract
Divergent results in pain management account for the growing number of studies aiming at elucidating the pharmacology of the endocannabinoid/endovanilloid anandamide (AEA) within several pain-related brain structures. For instance, the stimulation of both Transient Receptor Potential Vanilloid type 1 (TRPV1) and Cannabinoid type 1 (CB1) receptors led to paradoxical effects on nociception. Here, we attempted to propose a clear and reproducible methodology to achieve the antinociceptive effect of exogenous AEA within the dorsal periaqueductal gray (dPAG) of mice exposed to the tail-flick test. Accordingly, male Swiss mice received intra-dPAG injection of AEA (CB1/TRPV1 agonist), capsaicin (TRPV1 agonist), WIN (CB1 agonist), AM251 (CB1 antagonist), and 6-iodonordihydrocapsaicin (6-IODO) (TRPV1 selective antagonist) and their nociceptive response was assessed with the tail-flick test. In order to assess AEA effects on nociception specifically at vanilloid or cannabinoid (CB) substrates into the dPAG, mice underwent an intrinsically inactive dose of AM251 or 6-IODO followed by local AEA injections and were subjected to the same test. While intra-dPAG AEA did not change acute pain, local injections of capsaicin or WIN induced a marked TRPV1- and CB1-dependent antinociceptive effect, respectively. Regarding the role of AEA specifically at CB/vanilloid substrates, while the blockade of TRPV1 did not change the lack of effects of intra-dPAG AEA on nociception, local pre-treatment of AM251, a CB1 antagonist, led to a clear AEA-induced antinociception. It seems that the exogenous AEA-induced antinociception is unmasked when it selectively binds to vanilloid substrates, which might be useful to address acute pain in basic and perhaps clinical trials.
Collapse
Affiliation(s)
- Diego C Mascarenhas
- Joint Graduate Program in Physiological Sciences, Federal University of São Carlos and São Paulo State University, São Carlos, Brazil.,Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil
| | - Karina S Gomes
- Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil
| | - Tatiani Sorregotti
- Joint Graduate Program in Physiological Sciences, Federal University of São Carlos and São Paulo State University, São Carlos, Brazil.,Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil
| | - Ricardo L Nunes-de-Souza
- Joint Graduate Program in Physiological Sciences, Federal University of São Carlos and São Paulo State University, São Carlos, Brazil.,Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil
| |
Collapse
|
38
|
Chenaf C, Chapuy E, Libert F, Marchand F, Courteix C, Bertrand M, Gabriel C, Mocaër E, Eschalier A, Authier N. Agomelatine: a new opportunity to reduce neuropathic pain-preclinical evidence. Pain 2017; 158:149-160. [PMID: 27984527 DOI: 10.1097/j.pain.0000000000000738] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Antidepressants are first-line treatments of neuropathic pain but not all these drugs are really effective. Agomelatine is an antidepressant with a novel mode of action, acting as an MT1/MT2 melatonergic receptor agonist and a 5-HT2C receptor antagonist that involves indirect norepinephrine release. Melatonin, serotonin, and norepinephrine have been involved in the pathophysiology of neuropathic pain. Yet, no study has been conducted to determine agomelatine effects on neuropathic pain in animal models. Using 3 rat models of neuropathic pain of toxic (oxaliplatin/OXA), metabolic (streptozocin/STZ), and traumatic (sciatic nerve ligation/CCI [chronic constriction nerve injury]) etiologies, we investigated the antihypersensitivity effect of acute and repeated agomelatine administration. We then determined the influence of melatonergic, 5-HT2C, α-2 and β-1/2 adrenergic receptor antagonists in the antihypersensitivity effect of agomelatine. The effect of the combination of agomelatine + gabapentin was evaluated using an isobolographic approach. In STZ and CCI models, single doses of agomelatine significantly and dose dependently reduced mechanical hypersensitivity. After daily administrations for 2 weeks, this effect was confirmed in the CCI model and agomelatine also displayed a marked antihypersensitivity effect in the OXA model. The antihypersensitivity effect of agomelatine involved melatonergic, 5-HT2C, and α-2 adrenergic receptors but not beta adrenoceptors. The isobolographic analysis demonstrated that the combination of agomelatine + gabapentin had additive effects. Agomelatine exerts a clear-cut antihypersensitivity effect in 3 different neuropathic pain models. Its effect is mediated by melatonergic and 5-HT2C receptors and, although agomelatine has no affinity, also by α-2 adrenergic receptors. Finally, agomelatine combined with gabapentin produces an additive antihypersensitivity effect.
Collapse
Affiliation(s)
- Chouki Chenaf
- Université Clermont Auvergne, Université d'Auvergne, INSERM UMR 1107 Neuro-Dol Equipe Pharmacologie Fondamentale et Clinique de la Douleur, CHU Clermont-Ferrand Service de Pharmacologie Médicale, Institut Analgesia, Faculté de Médecine, Clermont-Ferrand, France
| | - Eric Chapuy
- Université Clermont Auvergne, Université d'Auvergne, INSERM UMR 1107 Neuro-Dol Equipe Pharmacologie Fondamentale et Clinique de la Douleur, Institut Analgesia, Faculté de Médecine, Clermont-Ferrand, France
| | - Frédéric Libert
- Université Clermont Auvergne, Université d'Auvergne, INSERM UMR 1107 Neuro-Dol Equipe Pharmacologie Fondamentale et Clinique de la Douleur, CHU Clermont-Ferrand Service de Pharmacologie Médicale, Institut Analgesia, Faculté de Médecine, Clermont-Ferrand, France
| | - Fabien Marchand
- Université Clermont Auvergne, Université d'Auvergne, INSERM UMR 1107 Neuro-Dol Equipe Pharmacologie Fondamentale et Clinique de la Douleur, Institut Analgesia, Faculté de Médecine, Clermont-Ferrand, France
| | - Christine Courteix
- Université Clermont Auvergne, Université d'Auvergne, INSERM UMR 1107 Neuro-Dol Equipe Pharmacologie Fondamentale et Clinique de la Douleur, Institut Analgesia, Faculté de Médecine, Clermont-Ferrand, France
| | - Marianne Bertrand
- Neuropsychiatry Division, Institut de Recherches Internationales Servier, Suresnes, France
| | - Cecilia Gabriel
- Neuropsychiatry Division, Institut de Recherches Internationales Servier, Suresnes, France
| | - Elisabeth Mocaër
- Neuropsychiatry Division, Institut de Recherches Internationales Servier, Suresnes, France
| | - Alain Eschalier
- Université Clermont Auvergne, Université d'Auvergne, INSERM UMR 1107 Neuro-Dol Equipe Pharmacologie Fondamentale et Clinique de la Douleur, CHU Clermont-Ferrand Service de Pharmacologie Médicale, Institut Analgesia, Faculté de Médecine, Clermont-Ferrand, France
| | - Nicolas Authier
- Université Clermont Auvergne, Université d'Auvergne, INSERM UMR 1107 Neuro-Dol Equipe Pharmacologie Fondamentale et Clinique de la Douleur, CHU Clermont-Ferrand Service de Pharmacologie Médicale, Institut Analgesia, Faculté de Médecine, Clermont-Ferrand, France
| |
Collapse
|
39
|
Voulalas PJ, Ji Y, Jiang L, Asgar J, Ro JY, Masri R. Loss of dopamine D1 receptors and diminished D1/5 receptor-mediated ERK phosphorylation in the periaqueductal gray after spinal cord lesion. Neuroscience 2016; 343:94-105. [PMID: 27932310 DOI: 10.1016/j.neuroscience.2016.11.040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 11/18/2016] [Accepted: 11/25/2016] [Indexed: 12/20/2022]
Abstract
Neuropathic pain resulting from spinal cord injury is often accompanied by maladaptive plasticity of the central nervous system, including the opioid receptor-rich periaqueductal gray (PAG). Evidence suggests that sensory signaling via the PAG is robustly modulated by dopamine D1- and D2-like receptors, but the effect of damage to the spinal cord on D1 and D2 receptor protein expression and function in the PAG has not been examined. Here we show that 21days after a T10 or C6 spinothalamic tract lesion, both mice and rats display a remarkable decline in the expression of D1 receptors in the PAG, revealed by western blot analysis. These changes were associated with a significant reduction in hindpaw withdrawal thresholds in lesioned animals compared to sham-operated controls. We investigated the consequences of diminished D1 receptor levels by quantifying D1-like receptor-mediated phosphorylation of ERK1,2 and CREB, events that have been observed in numerous brain structures. In naïve animals, western blot analysis revealed that ERK1,2, but not CREB phosphorylation was significantly increased in the PAG by the D1-like agonist SKF 81297. Using immunohistochemistry, we found that SKF 81297 increased ERK1,2 phosphorylation in the PAG of sham animals. However, in lesioned animals, basal pERK1,2 levels were elevated and did not significantly increase after exposure to SKF 81297. Our findings provide support for the hypothesis that molecular adaptations resulting in a decrease in D1 receptor expression and signaling in the PAG are a consequence of SCL.
Collapse
Affiliation(s)
- Pamela J Voulalas
- University of Maryland School of Dentistry, Department of Endodontics, Periodontics & Prosthodontics, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Yadong Ji
- University of Maryland School of Dentistry, Department of Endodontics, Periodontics & Prosthodontics, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Li Jiang
- University of Maryland School of Medicine, Department of Diagnostic Radiology, Baltimore, MD 21201, USA
| | - Jamila Asgar
- University of Maryland School of Dentistry, Department of Neural and Pain Sciences, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Jin Y Ro
- University of Maryland School of Dentistry, Department of Neural and Pain Sciences, Baltimore, MD 21201, USA; Kyung Hee University, School of Dentistry, Department of Oral Medicine, Seoul, Republic of Korea
| | - Radi Masri
- University of Maryland School of Dentistry, Department of Endodontics, Periodontics & Prosthodontics, 650 W. Baltimore Street, Baltimore, MD 21201, USA; University of Maryland School of Medicine, Department of Anatomy and Neurobiology, 650 W. Baltimore Street, Baltimore, MD 21201, USA.
| |
Collapse
|
40
|
Kremer M, Salvat E, Muller A, Yalcin I, Barrot M. Antidepressants and gabapentinoids in neuropathic pain: Mechanistic insights. Neuroscience 2016; 338:183-206. [PMID: 27401055 DOI: 10.1016/j.neuroscience.2016.06.057] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/21/2016] [Accepted: 06/30/2016] [Indexed: 01/20/2023]
Abstract
Neuropathic pain arises as a consequence of a lesion or disease affecting the somatosensory system. It is generally chronic and challenging to treat. The recommended pharmacotherapy for neuropathic pain includes the use of some antidepressants, such as tricyclic antidepressants (TCAs) (amitriptyline…) or serotonin and noradrenaline re-uptake inhibitors (duloxetine…), and/or anticonvulsants such as the gabapentinoids gabapentin or pregabalin. Antidepressant drugs are not acute analgesics but require a chronic treatment to relieve neuropathic pain, which suggests the recruitment of secondary downstream mechanisms as well as long-term molecular and neuronal plasticity. Noradrenaline is a major actor for the action of antidepressant drugs in a neuropathic pain context. Mechanistic hypotheses have implied the recruitment of noradrenergic descending pathways as well as the peripheral recruitment of noradrenaline from sympathetic fibers sprouting into dorsal root ganglia; and importance of both α2 and β2 adrenoceptors have been reported. These monoamine re-uptake inhibitors may also indirectly act as anti-proinflammatory cytokine drugs; and their therapeutic action requires the opioid system, particularly the mu (MOP) and/or delta (DOP) opioid receptors. Gabapentinoids, which target the voltage-dependent calcium channels α2δ-1 subunit, inhibit calcium currents, thus decreasing the excitatory transmitter release and spinal sensitization. Gabapentinoids also activate the descending noradrenergic pain inhibitory system coupled to spinal α2 adrenoceptors. Gabapentinoid treatment may also indirectly impact on neuroimmune actors, like proinflammatory cytokines. These drugs are effective against neuropathic pain both with acute administration at high dose and with repeated administration. This review focuses on mechanistic knowledge concerning chronic antidepressant treatment and gabapentinoid treatment in a neuropathic pain context.
Collapse
Affiliation(s)
- Mélanie Kremer
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France; Université de Strasbourg, Strasbourg, France
| | - Eric Salvat
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France; Centre d'Etude et de Traitement de la Douleur, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - André Muller
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France; Centre d'Etude et de Traitement de la Douleur, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Ipek Yalcin
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Michel Barrot
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France.
| |
Collapse
|
41
|
Furuya-da-Cunha EM, Souza RRD, Canto-de-Souza A. Rat exposure in mice with neuropathic pain induces fear and antinociception that is not reversed by 5-HT2C receptor activation in the dorsal periaqueductal gray. Behav Brain Res 2016; 307:250-7. [PMID: 27059332 DOI: 10.1016/j.bbr.2016.04.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 01/14/2023]
Abstract
Previous studies have demonstrated that serotonin 5-HT2C receptors in the dorsal periaqueductal gray (dPAG) mediate both anxiety and antinociception in mice submitted to the elevated plus maze. The present study examined the effects of intra-dPAG infusion of the serotonin 5-HT2C receptor agonist (MK-212) in the defensive reactions and antinociception in mice with neurophatic pain confronted by a predator. Neuropathic pain was induced by chronic constriction injury (CCI) of the sciatic nerve, and predator confrontation was performed using the rat exposure test (RET). Our results demonstrated that both sham-operated and CCI mice exhibited intense defensive reactions when confronted by rats. However, rat-exposed CCI mice showed reduced pain reactivity in comparison to CCI mice exposed to a toy rat. Intra-dPAG infusion of MK-212 prior to predator exposure did not significantly alter defensive or antinociceptive responses. To our knowledge, our results represent the first evidence of RET-induced antinociception in mice. Moreover, the results of the present study suggest that 5-HT2C receptor activation in the dPAG is not critically involved in the control of predator-evoked fearful or antinociceptive responses.
Collapse
Affiliation(s)
- Elke Mayumi Furuya-da-Cunha
- Psychobiology Group/Department of Psychology/CECH-UFSCar, São Carlos, SP, 13565-905, Brazil; Joint Graduate Program in Physiological Sciences UFSCar/UNESP. Rod. Washington Luís, Km 235, São Carlos, SP, 13565-905, Brazil
| | - Rimenez Rodrigues de Souza
- Psychobiology Group/Department of Psychology/CECH-UFSCar, São Carlos, SP, 13565-905, Brazil; Graduate Program in Psychology UFSCar. Rod. Washington Luís, Km 235, São Carlos, SP, 13565-905, Brazil
| | - Azair Canto-de-Souza
- Psychobiology Group/Department of Psychology/CECH-UFSCar, São Carlos, SP, 13565-905, Brazil; Joint Graduate Program in Physiological Sciences UFSCar/UNESP. Rod. Washington Luís, Km 235, São Carlos, SP, 13565-905, Brazil; Graduate Program in Psychology UFSCar. Rod. Washington Luís, Km 235, São Carlos, SP, 13565-905, Brazil; Neuroscience and Behavioral Institute, Av. do Café, 2.450, 14050-220 Ribeirão Preto, SP, Brazil.
| |
Collapse
|