1
|
Losgott T, Schicker KW, Hilber K, Boehm S, Salzer I. Gaussian white noise stimulation as an alternative method to excite sensory neurons. Front Pharmacol 2025; 16:1561905. [PMID: 40331198 PMCID: PMC12053156 DOI: 10.3389/fphar.2025.1561905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
Introduction Peripheral nerve endings of dorsal root ganglion (DRG) neurons act as nociceptors and generate action potentials in response to noxious stimuli. Primary cultures of dissociated DRG have been used extensively to study changes neuronal excitability caused by either analgesics or pathological conditions, such as inflammation. The dissociation procedure can be viewed as a form of axotomy, and one might expect a resulting increase in excitability of the neurons during the subsequent culture period. However, changes in firing properties of DRG neurons over time in vitro have not been investigated systematically. Methods Thus, the current experiments compared action potential firing in dissociated DRG neurons after one to 7 days in culture and examined Gaussian white noise as novel stimulation paradigm. Primary cultures of DRG neurons were recorded in perforated patch current-clamp. Action potentials were evoked either by a sequence of five rectangular current pulses with increasing amplitudes or by Gaussian white noise of varying RMS amplitudes and frequencies. Results Conventional rectangular current injections triggered 19 ± 20 action potentials in cells when recorded within 24 h after dissociation. After 7 days in culture, DRG neurons fired 4.3 ± 0.7 action potentials in response to current pulses. Inflammatory mediators increased numbers of action potentials evoked by rectangular current pulses within 24 h after dissociation to 66 ± 54, but left those elicited after 7 days in vitro unaltered (4.3 ± 0.5). In the same set of neurons kept in culture for 7 days, Gaussian white noise stimuli triggered 1,540 ± 470 action potentials, and this number was increased to 2089 ± 685 by inflammatory mediators. The Kv7 channel activator retigabine and the paracetamol metabolite n-acetyl-p-benzoquinone imine (NAPQI) decreased numbers of action potentials triggered by Gaussian white noise, but failed to do so when rectangular current pulses were used as stimuli, both in neurons after 7 days in culture. Discussion These results demonstrate a decrease in the excitability of DRG neurons from day one to 7 after dissociation and reveal Gaussian white noise as reliable trigger of action potential firing in these neurons.
Collapse
Affiliation(s)
| | | | | | | | - Isabella Salzer
- Division of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
2
|
Guenther KG, Wirt JL, Oliva I, Saberi SA, Crystal JD, Hohmann AG. The cannabinoid CB 2 agonist LY2828360 suppresses neuropathic pain behavior and attenuates morphine tolerance and conditioned place preference in rats. Neuropharmacology 2025; 265:110257. [PMID: 39644993 PMCID: PMC11729772 DOI: 10.1016/j.neuropharm.2024.110257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/06/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Cannabinoid CB2 agonists show promise as analgesics because they lack unwanted side effects associated with direct activation of CB1 receptors. CB2 receptor activation suppresses pathological pain in animal models, but the types of pain that best respond to CB2 agonists are incompletely understood. This gap in knowledge may contribute to failures in clinical translation. We previously showed that the G protein-biased CB2 receptor agonist LY2828360 attenuated the maintenance of neuropathic pain behavior in mouse models of inflammatory and neuropathic pain. Whether this finding generalizes to neuropathic pain induced by traumatic nerve injury or occurs in multiple rodent species remains unknown. Here we show that LY2828360 (3 and 10 mg/kg i.p.), administered acutely, reversed paclitaxel-induced mechanical hypersensitivity in male rats. By contrast, LY2828360 (10 mg/kg i.p.), administered acutely, attenuated mechanical hypersensitivity in a spared nerve injury (SNI) rat model, whereas the low dose (3 mg/kg i.p.) was ineffective. In both models, efficacy of LY2828360 was sustained following 10 days of repeated dosing. LY2828360 (3 mg/kg i.p.) also prevented development of tolerance to the opioid analgesic morphine (6 mg/kg i.p.) in rats with SNI when co-administered. LY2828360 (3 mg/kg i.p.) did not produce preference or aversion in the conditioned place preference (CPP) test in rats when administered alone but blocked CPP to morphine (6 mg/kg i.p.). Lastly, LY2828360 (3 mg/kg i.p.) did not alter the acquisition of i.v. morphine self-administration under fixed ratio 1 (FR1) and 3 (FR3) or motivation to work for morphine under a progressive ratio (PR) schedule of reinforcement.
Collapse
Affiliation(s)
- Kelsey G Guenther
- Program in Neuroscience, Indiana University, Bloomington, IN, USA; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Jonah L Wirt
- Program in Neuroscience, Indiana University, Bloomington, IN, USA; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Idaira Oliva
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA; Gill Institute for Neuroscience, Indiana University, Bloomington, IN, USA
| | - Shahin A Saberi
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Jonathon D Crystal
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA; Gill Institute for Neuroscience, Indiana University, Bloomington, IN, USA
| | - Andrea G Hohmann
- Program in Neuroscience, Indiana University, Bloomington, IN, USA; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA; Gill Institute for Neuroscience, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
3
|
Balcazar-Ochoa LG, Ángeles-López GE, Chavarría A, Ramírez-Carreto RJ, González-Hernández A, Guzmán-Ruiz MA, Segovia-Mendoza M, Ochoa-Aguilar A, Ventura-Martínez R. Clavulanic acid prevents paclitaxel-induced neuropathic pain through a systemic and central anti-inflammatory effect in mice. Neurotherapeutics 2025; 22:e00522. [PMID: 39794241 PMCID: PMC12014412 DOI: 10.1016/j.neurot.2024.e00522] [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: 09/19/2024] [Revised: 12/13/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Paclitaxel (PCX) based treatments, commonly used to treat breast, ovarian and lung cancers, have the highest incidence of chemotherapy-induced neuropathic pain, affecting from 38 to 94 % of patients. Unfortunately, analgesic treatments are not always effective for PCX-induced neuropathic pain (PINP). This study aimed to evaluate the antinociceptive effect of clavulanic acid (CLAV), a clinically used β-lactam molecule, in both therapeutic and preventive contexts in mice with PINP. A single dose of CLAV administered after the onset of PINP significantly reduced mechanical hyperalgesia. Interestingly, preventive administration of CLAV prevented PINP development. The effect of preventive CLAV on PINP was associated with increased levels of IL-10 and IFN-β in serum, and decreased levels of IL-1β and TNF-α in both the serum and CNS. Immunostaining experiments revelated that CLAV increased the levels of glutamate transporter type 1 (GLT-1) and toll-like receptor type 4 (TLR4) in the spinal cord, while reducing levels of the astrocytic marker the glial fibrillary acidic protein (GFAP). Notably, co-incubation with CLAV and PCX in triple-negative breast cancer cells did not interfere with PCX-induced cytotoxic effects. Hence, these findings suggest that CLAV could be employed as a clinical treatment aimed at preventing PINP without compromission the cytotoxic efficacy of PCX.
Collapse
Affiliation(s)
- Luis Gerardo Balcazar-Ochoa
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico
| | | | - Anahí Chavarría
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico
| | - Ricardo Jair Ramírez-Carreto
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico
| | | | - Mara Alaide Guzmán-Ruiz
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico
| | - Mariana Segovia-Mendoza
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico
| | | | - Rosa Ventura-Martínez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico.
| |
Collapse
|
4
|
Gao P, Peng F, Liu J, Wu W, Zhao G, Liu C, Cao H, Li Y, Qiu F, Zhang W. Lidocaine Enhanced Antitumor Efficacy and Relieved Chemotherapy-Induced Hyperalgesia in Mice with Metastatic Gastric Cancer. Int J Mol Sci 2025; 26:828. [PMID: 39859541 PMCID: PMC11766172 DOI: 10.3390/ijms26020828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
With the widespread use of lidocaine for pain control in cancer therapy, its antitumor activity has attracted considerable attention in recent years. This paper provides a simple strategy of combining lidocaine with chemotherapy drugs for cancer therapy, aiming to relieve chemotherapy-induced pain and achieve stronger antitumor efficacy. However, there is still a lack of substantial pre-clinical evidence for the efficacy and related mechanisms of such combinations, obstructing their potential clinical application. In this study, we propose intraperitoneal chemotherapy (IPC) against gastric cancer (GC) as an ideal scenario to evaluate the efficacy of a lidocaine/paclitaxel combination. Firstly, we used human GC cells MKN-45-luc to investigate the antitumor activity and related mechanisms of the lidocaine/paclitaxel combination in vitro. Then, we used C57BL/6 mice with intraperitoneal drug suffusion to evaluate the efficacy of lidocaine to suppress paclitaxel-induced hyperalgesia and related mechanisms. Lastly, in BALB/c tumor-bearing nude mice we evaluated the synergistic antitumor activity and pain-relieving effect of the lidocaine/paclitaxel combination. Our results showed enhanced antitumor activity for the lidocaine/paclitaxel combination, which induced apoptosis, inhibited migration, and the invasion of GC cells in a synergistic manner. In animal models, the lidocaine/paclitaxel combination effectively inhibited growth and peritoneal metastasis of the tumor, resulting in prolonged survival time. Meanwhile, lidocaine showed considerable anti-inflammatory activity alongside its anesthetic effect, which, in combination, effectively relieved hyperalgesia induced by paclitaxel. These results suggested that intraperitoneal suffusion with lidocaine/paclitaxel could be a pain-free IPC formulation with enhanced antitumor activity, which could provide a promising treatment for GC with peritoneal metastasis.
Collapse
Affiliation(s)
- Peiwen Gao
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (P.G.); (J.L.); (W.W.)
| | - Fei Peng
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| | - Jing Liu
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (P.G.); (J.L.); (W.W.)
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| | - Weiwei Wu
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (P.G.); (J.L.); (W.W.)
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| | - Guoyan Zhao
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| | - Congyan Liu
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| | - Hangxue Cao
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| | - Yuncheng Li
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| | - Feng Qiu
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (P.G.); (J.L.); (W.W.)
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| | - Wensheng Zhang
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (P.G.); (J.L.); (W.W.)
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| |
Collapse
|
5
|
Doyle TM, Janes K, Xiao WH, Kolar GR, Luecke HF, Gratton MA, Tosh DK, Jacobson KA, Bennett GJ, Salvemini D. Mitochondrial A 3 Adenosine Receptor as a Mechanism for the Protective Effects of A 3AR Agonists on Chemotherapy-Induced Neuropathic Pain. J Neurosci 2025; 45:e1268242024. [PMID: 39653498 PMCID: PMC11735668 DOI: 10.1523/jneurosci.1268-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/11/2024] [Accepted: 10/29/2024] [Indexed: 01/18/2025] Open
Abstract
Alterations in mitochondrial function are the linchpin in numerous disease states including in the development of chemotherapy-induced neuropathic pain (CIPN), a major dose-limiting toxicity of widely used chemotherapeutic cytotoxins. In CIPN, mitochondrial dysfunction is characterized by deficits in mitochondrial bioenergetics (e.g., decreased ATP production) that are thought to drive the degeneration of the peripheral nerve sensory axon terminal sensory arbors in the skin (the intraepidermal nerve fibers; IENFs) and induce abnormal spontaneous discharge in peripheral nerve sensory axons. Preserving mitochondrial function is anticipated to prevent CIPN. We have now discovered that the G-protein-coupled receptor, A3 adenosine receptor subtype (A3AR), is expressed on the mitochondrial outer membrane. Ex vivo application of a highly selective A3AR agonist, MRS5980, to saphenous nerve microfilaments harvested from male oxaliplatin-treated rats reversed the loss in ATP production underscoring mitoprotective effects resulting from A3AR activation on mitochondria. Moreover, in vivo administration of A3AR agonists to rats during oxaliplatin treatment was associated with reduced IENF loss and a lower incidence of spontaneous discharge in peripheral afferent axons. These effects are accompanied by improved mitochondrial ATP production in primary afferent sensory axons and overall inhibition of the development of neuropathic pain. These data identify for the first time mitochondrial A3AR and indicate that activation of A3AR protects mitochondrial function in primary afferent sensory axons against chemotherapy-induced neurotoxicity. Repurposing A3AR agonists that are already in clinical trials as anticancer agents as adjunct to chemotherapeutics will address a major unmet medical need for which there are no FDA-approved drugs.
Collapse
Affiliation(s)
- Timothy M Doyle
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri 63104
- Institute for Translational Neuroscience, Saint Louis University, Saint Louis, Missouri 63104
| | - Kali Janes
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri 63104
| | - Wen Hua Xiao
- Department of Anesthesiology, University of California San Diego, San Diego, California 92103
| | - Grant R Kolar
- Institute for Translational Neuroscience, Saint Louis University, Saint Louis, Missouri 63104
- Department of Pathology, Saint Louis University School of Medicine, Saint Louis, Missouri 63104
| | - Hans F Luecke
- Chemical Proteomics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Michael Anne Gratton
- Center for Sensory Neuroscience, Boys Town National Research Hospital, Boys Town, Nebraska 68010
| | - Dilip K Tosh
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Gary J Bennett
- Department of Anesthesiology, University of California San Diego, San Diego, California 92103
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri 63104
- Institute for Translational Neuroscience, Saint Louis University, Saint Louis, Missouri 63104
| |
Collapse
|
6
|
Abd Elaleem WS, Ghaiad HR, Abd Elmawla MA, Shaheen AA. Attenuation of p38 MAPK/NF-κB/TRPV1/CGRP is involved in the antinociceptive effect of hesperidin methyl chalcone and taxifolin in paclitaxel-induced peripheral neuropathy. Biofactors 2025; 51:e2125. [PMID: 39353740 DOI: 10.1002/biof.2125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024]
Abstract
Paclitaxel (PTX)-induced peripheral neuropathy (PIPN) is a disabling side effect of PTX, which adversely affects the life quality of cancer patients. Flavonoids such as hesperidin methyl chalcone (HMC) and taxifolin (TAX) can alleviate neuropathic pain via their anti-inflammatory, antioxidant, neuroprotective, and antinociceptive properties. The current study aimed to assess the efficacy of HMC and TAX in preventing PIPN individually or in combination. Pretreatment with HMC and TAX mitigated PTX-induced mechanical allodynia and hyperalgesia, cold allodynia, and thermal hyperalgesia as well as restore the normal histological architecture. Remarkably, neuropathic pain was relieved by suppression of nerve growth factor (NGF), p38 mitogen-activated protein kinase (p38 MAPK), and transient receptor potential vanilloid type-1 (TRPV1), which ultimately lead to reduced calcitonin gene-related peptide (CGRP). Furthermore, both HMC or TAX enhanced nuclear factor erythroid 2-related factor 2 (Nrf2), leading to elevated glutathione (GSH) and total antioxidant capacity (TAC) along with lowered malondialdehyde (MDA), which in turn, downregulated nuclear factor kappa B P65 (NF-κB P65) and its phosphorylated form and eventually reduced tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1β) then lowered the apoptotic indices. Promisingly, the combination of both agents was superior to each drug alone through targeting more diverse signaling pathways and achieving synergistic and comprehensive therapeutic effects. In conclusion, pretreatment with HMC and TAX separately or in combination alleviated PIPN via modulating NGF/p38 MAPK/NF-κB P65/TRPV1/CGRP pathway.
Collapse
Affiliation(s)
- Wafaa S Abd Elaleem
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Heba R Ghaiad
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mai A Abd Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Amira A Shaheen
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
7
|
Li Y, Uhelski ML, North RY, Farson LB, Bankston CB, Roland GH, Fan DH, Sheffield KN, Jia A, Orlando D, Heles M, Yaksh TL, Miller YI, Kosten TA, Dougherty PM. ApoA-I binding protein (AIBP) regulates transient receptor potential vanilloid 1 (TRPV1) activity in rat dorsal root ganglion neurons by selective disruption of toll-like receptor 4 (TLR4)-lipid rafts. Brain Behav Immun 2025; 123:644-655. [PMID: 39414176 DOI: 10.1016/j.bbi.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024] Open
Abstract
Toll-like receptor 4 (TLR4) and the transient receptor potential vanilloid subtype 1 (TRPV1) are both upregulated and play key roles in the induction and expression of paclitaxel-related chemotherapy-induced peripheral neuropathy (CIPN). Using Apolipoprotein A-I binding protein, non-specific cholesterol depletion, TLR4 mis-sense rats and a TLR4 inhibitor, we demonstrate that co-localization of TRPV1 with TLR4 to cholesterol-rich lipid membrane rafts in nociceptors is essential for its normal activation as well as for its exaggerated activation that underlies the development and expression of CIPN. The findings suggest that TLR4-lipid rafts may have an essential role in numerous neuroinflammatory and neuropathic pain conditions. This mechanism is also generalized to female rats for the first time.
Collapse
Affiliation(s)
- Yan Li
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, the United States of America
| | - Megan L Uhelski
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, the United States of America
| | - Robert Y North
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, the United States of America
| | - Luke B Farson
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | - Christopher B Bankston
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | - Gavin H Roland
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | - Dwight H Fan
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | | | - Amy Jia
- Northwestern University, Evanston, IL 60208, the United States of America
| | - Dana Orlando
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | - Mario Heles
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, the United States of America
| | - Tony L Yaksh
- The Department of Anesthesiology, the University of California San Diego, La Jolla, CA, 92093, the United States of America
| | - Yury I Miller
- Department of Medicine, the University of California San Diego, La Jolla, CA, 92093, the United States of America
| | - Therese A Kosten
- Department of Psychology, Health Building 1, 4349 Martin Luther King Blvd, Houston, TX 77204, the United States of America
| | - Patrick M Dougherty
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, the United States of America.
| |
Collapse
|
8
|
Zamani A, EmamiAref P, Kubíčková L, Hašanová K, Šandor O, Dubový P, Joukal M. Paclitaxel triggers molecular and cellular changes in the choroid plexus. FRONTIERS IN PAIN RESEARCH 2024; 5:1488369. [PMID: 39654799 PMCID: PMC11625821 DOI: 10.3389/fpain.2024.1488369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024] Open
Abstract
Paclitaxel is a widely used chemotherapeutic agent for treating various solid tumors. However, resulting neuropathic pain, often a lifelong side effect of paclitaxel, can limit dosing and compromise optimal treatment. The choroid plexus, located in the brain ventricles, spreads peripheral inflammatory reactions into the brain. Our study is the first to analyze the effects of paclitaxel on inflammatory alterations in the choroid plexus. We hypothesized that the choroid plexus could respond directly to paclitaxel and simultaneously be indirectly altered via circulating damage-associated molecular patterns (DAMPs) produced by paclitaxel application. Using immunohistochemical and Western blot analysis, we examined the levels of toll-like receptor 9 (TLR9) and formyl peptide receptor 2 (FPR2), along with the pro-inflammatory cytokines interleukin 6 (IL6) and tumor necrosis factor α (TNFα) in choroid plexus epithelial cells of male Wistar rats following paclitaxel treatment. Moreover, we utilized an in vitro model of choroid plexus epithelial cells, the Z310 cells, to investigate the changes in these cells in response to paclitaxel and DAMPs (CpG ODN). Our results demonstrate that paclitaxel increases TLR9 and FPR2 levels in the choroid plexus while inducing IL6 and TNFα upregulation in both acute and chronic manners. In vitro experiments further revealed that paclitaxel directly interacts with epithelial cells of the choroid plexus, leading to increased levels of TLR9, FPR2, IL6, and TNFα. Additionally, treatment of cells with CpG ODN, an agonist of TLR9, elicited upregulation of IL6 and TNFα. Our findings determined that paclitaxel influences the choroid plexus through both direct and indirect mechanisms, resulting in inflammatory profile alterations. Given the pivotal role of the choroid plexus in brain homeostasis, a compromised choroid plexus following chemotherapy may facilitate the spread of peripheral inflammation into the brain, consequently exacerbating the development of neuropathic pain.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marek Joukal
- Department of Anatomy, Alemeh Zamani Research Group, Faculty of Medicine, Masaryk University, Brno, Czechia
| |
Collapse
|
9
|
Xiao XY, Chen YM, Zhu J, Yin MY, Huang CN, Qin HM, Liu SX, Xiao Y, Fang HW, Zhuang T, Chen Y. The synergistic anti-nociceptive effects of nefopam and gabapentinoids in inflammatory, osteoarthritis, and neuropathic pain mouse models. Eur J Pharmacol 2024; 977:176738. [PMID: 38876275 DOI: 10.1016/j.ejphar.2024.176738] [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: 07/13/2023] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/16/2024]
Abstract
Pain is a common public health problem and remains as an unmet medical need. Currently available analgesics usually have limited efficacy or are accompanied by many adverse side effects. To achieve satisfactory pain relief by multimodal analgesia, new combinations of nefopam and gabapentinoids (pregabalin/gabapentin) were designed and assessed in inflammatory, osteoarthritis and neuropathic pain. Isobolographic analysis was performed to analyze the interactions between nefopam and gabapentinoids in carrageenan-induced inflammatory pain, mono-iodoacetate-induced osteoarthritis pain and paclitaxel-induced peripheral neuropathic pain in mice. The anti-inflammatory effect and motor performance of monotherapy or their combinations were evaluated in the carrageenan-induced inflammatory responses and rotarod test, respectively. Nefopam (1, 3, 5, 10, 30 mg/kg, p.o.), pregabalin (3, 6, 12, 24 mg/kg, p.o.) or gabapentin (25, 50, 75, 100 mg/kg, p.o.) dose-dependently reversed mechanical allodynia in three pain models. Isobolographic analysis indicated that the combinations of nefopam and gabapentinoids exerted synergistic anti-nociceptive effects in inflammatory, osteoarthritis, and neuropathic pain mouse models, as evidenced by the experimental ED50 (median effective dose) falling below the predicted additive line. Moreover, the combination of nefopam-pregabalin/gabapentin alleviated carrageenan-induced inflammation and edema, and also prevented gabapentinoids-related sedation or ataxia by lowering their effective doses. Collectively, the co-administration of nefopam and gabapentinoids showed synergistic analgesic effects and may result in improved therapeutic benefits for treating pain.
Collapse
Affiliation(s)
- Xin-Yi Xiao
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yan-Ming Chen
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jin Zhu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Ming-Yue Yin
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Chao-Nan Huang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Hui-Min Qin
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Shu-Xian Liu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yang Xiao
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Heng-Wei Fang
- School of Pharmacy, Henan University, Kaifeng, 475001, China
| | - Tao Zhuang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China.
| | - Yin Chen
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
10
|
Filipiuc LE, Creangă-Murariu I, Tamba BI, Ababei DC, Rusu RN, Stanciu GD, Ștefanescu R, Ciorpac M, Szilagyi A, Gogu R, Filipiuc SI, Tudorancea IM, Solcan C, Alexa-Stratulat T, Cumpăt MC, Cojocaru DC, Bild V. JWH-182: a safe and effective synthetic cannabinoid for chemotherapy-induced neuropathic pain in preclinical models. Sci Rep 2024; 14:16242. [PMID: 39004628 PMCID: PMC11247095 DOI: 10.1038/s41598-024-67154-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024] Open
Abstract
Chemotherapy-induced neuropathic pain (CINP), a condition with unmet treatment needs, affects over half of cancer patients treated with chemotherapeutics. Researchers have recently focused on the endocannabinoid system because of its critical role in regulating our bodies' most important functions, including pain. We used in vitro and in vivo methods to determine the toxicity profile of a synthetic cannabinoid, JWH-182, and whether it could be potentially effective for CINP alleviation. In vitro, we evaluated JWH-182 general toxicity, measuring fibroblast viability treated with various concentrations of compound, and its neuroprotection on dorsal root ganglion neurons treated with paclitaxel. In vivo, we performed an evaluation of acute and 28-day repeated dose toxicity in mice, with monitoring of health status and a complete histopathological examination. Finally, we evaluated the efficacy of JWH-182 on a CINP model in mice using specific pain assessment tests. JWH-182 has an acceptable toxicity profile, in both, in vitro and in vivo studies and it was able to significantly reduce pain perception in a CINP model in mice. However, the translation of these results to the clinic needs further investigation.
Collapse
Affiliation(s)
- Leontina-Elena Filipiuc
- Advanced Research and Development Center for Experimental Medicine (CEMEX), "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
- Department of Pharmacology, Clinical Pharmacology and Algesiology, "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
| | - Ioana Creangă-Murariu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
- Department of Pharmacology, Clinical Pharmacology and Algesiology, "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
| | - Bogdan-Ionel Tamba
- Advanced Research and Development Center for Experimental Medicine (CEMEX), "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania.
- Department of Pharmacology, Clinical Pharmacology and Algesiology, "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania.
| | - Daniela-Carmen Ababei
- Advanced Research and Development Center for Experimental Medicine (CEMEX), "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
- Pharmacodynamics and Clinical Pharmacy Department, "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
| | - Răzvan-Nicolae Rusu
- Pharmacodynamics and Clinical Pharmacy Department, "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
| | - Gabriela-Dumitrița Stanciu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
| | - Raluca Ștefanescu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
| | - Mitică Ciorpac
- Advanced Research and Development Center for Experimental Medicine (CEMEX), "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
| | - Andrei Szilagyi
- Advanced Research and Development Center for Experimental Medicine (CEMEX), "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
| | - Raluca Gogu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
| | - Silviu-Iulian Filipiuc
- Advanced Research and Development Center for Experimental Medicine (CEMEX), "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
| | - Ivona-Maria Tudorancea
- Advanced Research and Development Center for Experimental Medicine (CEMEX), "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
- Department of Pharmacology, Clinical Pharmacology and Algesiology, "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
| | - Carmen Solcan
- Faculty of Veterinary Medicine, "Ion Ionescu de La Brad" University of Life Sciences, 700490, Iasi, Romania
| | - Teodora Alexa-Stratulat
- Oncology Department, Regional Institute of Oncology, Iasi, Romania
- Department of Medical Oncology-Radiotherapy, "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
| | - Marinela-Carmen Cumpăt
- Department of Medical Specialties I and III, "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
- Clinical Rehabilitation Hospital, Cardiovascular and Respiratory Rehabilitation Clinic, Pantelimon Halipa Street No. 14, 700661, Iasi, Romania
| | - Doina-Clementina Cojocaru
- Department of Medical Specialties I and III, "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
- Clinical Rehabilitation Hospital, Cardiovascular and Respiratory Rehabilitation Clinic, Pantelimon Halipa Street No. 14, 700661, Iasi, Romania
| | - Veronica Bild
- Advanced Research and Development Center for Experimental Medicine (CEMEX), "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
- Pharmacodynamics and Clinical Pharmacy Department, "Grigore T. Popa" University of Medicine and Pharmacy, University Street No. 16, 700115, Iasi, Romania
- Center of Biomedical Research, Romanian Academy, Iasi Branch, Iasi, Romania
| |
Collapse
|
11
|
Li WW, Zhao Y, Liu HC, Liu J, Chan SO, Zhong YF, Zhang TY, Liu Y, Zhang W, Xia YQ, Chi XC, Xu J, Wang Y, Wang J. Roles of Thermosensitive Transient Receptor Channels TRPV1 and TRPM8 in Paclitaxel-Induced Peripheral Neuropathic Pain. Int J Mol Sci 2024; 25:5813. [PMID: 38892000 PMCID: PMC11171746 DOI: 10.3390/ijms25115813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Paclitaxel, a microtubule-stabilizing chemotherapy drug, can cause severe paclitaxel-induced peripheral neuropathic pain (PIPNP). The roles of transient receptor potential (TRP) ion channel vanilloid 1 (TRPV1, a nociceptor and heat sensor) and melastatin 8 (TRPM8, a cold sensor) in PIPNP remain controversial. In this study, Western blotting, immunofluorescence staining, and calcium imaging revealed that the expression and functional activity of TRPV1 were upregulated in rat dorsal root ganglion (DRG) neurons in PIPNP. Behavioral assessments using the von Frey and brush tests demonstrated that mechanical hyperalgesia in PIPNP was significantly inhibited by intraperitoneal or intrathecal administration of the TRPV1 antagonist capsazepine, indicating that TRPV1 played a key role in PIPNP. Conversely, the expression of TRPM8 protein decreased and its channel activity was reduced in DRG neurons. Furthermore, activation of TRPM8 via topical application of menthol or intrathecal injection of WS-12 attenuated the mechanical pain. Mechanistically, the TRPV1 activity triggered by capsaicin (a TRPV1 agonist) was reduced after menthol application in cultured DRG neurons, especially in the paclitaxel-treated group. These findings showed that upregulation of TRPV1 and inhibition of TRPM8 are involved in the generation of PIPNP, and they suggested that inhibition of TRPV1 function in DRG neurons via activation of TRPM8 might underlie the analgesic effects of menthol.
Collapse
Affiliation(s)
- Wen-Wen Li
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Yan Zhao
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Huai-Cun Liu
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Jiao Liu
- Center of Medical and Health Analysis, Peking University Health Science Center, Beijing 100191, China;
| | - Sun-On Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yi-Fei Zhong
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Tang-Yu Zhang
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Yu Liu
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Wei Zhang
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Yu-Qi Xia
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Xiao-Chun Chi
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Jian Xu
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Yun Wang
- Neuroscience Research Institute and Department of Neurobiology, Key Laboratory for Neuroscience of Ministry of Education and Neuroscience, Peking University Health Science Center, Beijing 100191, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Jun Wang
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| |
Collapse
|
12
|
Xu Y, Yao L, Guo Y, Shi C, Zhou J, Hua M. The Potential Antinociceptive Effect and Mechanism of Cannabis sativa L. Extract on Paclitaxel-Induced Neuropathic Pain in Rats Uncovered by Multi-Omics Analysis. Molecules 2024; 29:1958. [PMID: 38731449 PMCID: PMC11085863 DOI: 10.3390/molecules29091958] [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: 03/16/2024] [Revised: 04/12/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
Cannabis sativa L. (hemp) is a herbaceous plant rich in cannabinoids with a long history of use in pain treatment. The most well-characterized cannabinoids, cannabidiol (CBD) and Δ9-tetrahydrocannabinol (Δ9-THC), garnered much attention in chemotherapy-induced peripheral neuropathy (CIPN) treatment. However, few studies have investigated the biological benefits and mechanism of hemp extract on CIPN. In the present study, hemp extract (JG) rich in cannabinoids was extracted by supercritical fluid carbon dioxide extraction (SFCE). The antinociceptive efficacy was evaluated using a paclitaxel-induced peripheral neuropathy (PIPN) rat model based on behavioral tests. Further omics-based approaches were applied to explore the potential mechanisms. The results showed that JG decreased mechanical allodynia, thermal hyperalgesia, and inflammatory cytokines in PIPN rats significantly. Transcriptome analysis identified seven key genes significantly regulated by JG in PIPN model rats, mainly related to the neuroactive ligand-receptor interaction pathway, PPAR signaling pathway, and cAMP signaling pathway. In metabolomic analysis, a total of 39 significantly altered metabolites were identified, mainly correlated with pentose and glucuronate interconversions and the glycerophospholipid metabolism pathway. Gut microbiota analysis suggested that increased community Lachnoclostridium and Lachnospiraceae_UCG-006 in PIPN rats can be reversed significantly by JG. In conclusion, hemp extract exhibited antinociceptive effects on PIPN. The analgesic mechanism was probably related to the regulation of inflammation, neuroactive ligand-receptor interaction pathway, sphingolipid metabolism, etc. This study provides novel insights into the functional interactions of Cannabis sativa L. extract on PIPN.
Collapse
Affiliation(s)
| | | | | | | | | | - Moli Hua
- National Key Laboratory of Lead Druggability Research, Shanghai Institute of Pharmaceutical Industry Co., Ltd., China State Institute of Pharmaceutical Industry, Shanghai 201203, China; (Y.X.); (L.Y.); (Y.G.); (C.S.); (J.Z.)
| |
Collapse
|
13
|
Li H, Ward SJ. Paclitaxel-Associated Mechanical Sensitivity and Neuroinflammation Are Sex-, Time-, and Site-Specific and Prevented through Cannabigerol Administration in C57Bl/6 Mice. Int J Mol Sci 2024; 25:4277. [PMID: 38673862 PMCID: PMC11050247 DOI: 10.3390/ijms25084277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is one of the most prevalent and dose-limiting complications in chemotherapy patients. One identified mechanism underlying CIPN is neuroinflammation. Most of this research has been conducted in only male or female rodent models, making direct comparisons regarding the role of sex differences in the neuroimmune underpinnings of CIPN limited. Moreover, most measurements have focused on the dorsal root ganglia (DRG) and/or spinal cord, while relatively few studies have been aimed at characterizing neuroinflammation in the brain, for example the periaqueductal grey (PAG). The overall goals of the present study were to determine (1) paclitaxel-associated changes in markers of inflammation in the PAG and DRG in male and female C57Bl6 mice and (2) determine the effect of prophylactic administration of an anti-inflammatory cannabinoid, cannabigerol (CBG). In Experiment 1, male and female mice were treated with paclitaxel (8-32 mg/kg/injection, Days 1, 3, 5, and 7) and mechanical sensitivity was measured using Von Frey filaments on Day 7 (Cohort 1) and Day 14 (Cohort 2). Cohorts were euthanized on Day 8 or 15, respectively, and DRG and PAG were harvested for qPCR analysis of the gene expression of markers of pain and inflammation Aig1, Gfap, Ccl2, Cxcl9, Tlr4, Il6, and Calca. In Experiment 2, male and female mice were treated with vehicle or 10 mg/kg CBG i.p. 30 min prior to each paclitaxel injection. Mechanical sensitivity was measured on Day 14. Mice were euthanized on Day 15, and PAG were harvested for qPCR analysis of the gene expression of Aig1, Gfap, Ccl2, Cxcl9, Tlr4, Il6, and Calca. Paclitaxel produced a transient increase in potency to produce mechanical sensitivity in male versus female mice. Regarding neuroinflammation, more gene expression changes were apparent earlier in the DRG and at a later time point in the PAG. Also, more changes were observed in females in the PAG than males. Overall, sex differences were observed for most markers at both time points and regions. Importantly, in both the DRG and PAG, most increases in markers of neuroinflammation and pain occurred at paclitaxel doses higher than those associated with significant changes in the mechanical threshold. Two analytes that demonstrated the most compelling sexual dimorphism and that changed more in males were Cxcl9 and Ccl2, and Tlr4 in females. Lastly, prophylactic administration of CBG protected the male and female mice from increased mechanical sensitivity and female mice from neuroinflammation in the PAG. Future studies are warranted to explore how these sex differences may shed light on the mechanisms of CIPN and how non-psychoactive cannabinoids such as CBG may engage these targets to prevent or attenuate the effects of paclitaxel and other chemotherapeutic agents on the nervous system.
Collapse
Affiliation(s)
| | - Sara Jane Ward
- Center for Substance Abuse Research, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| |
Collapse
|
14
|
Chen X, Gan Y, Au NPB, Ma CHE. Current understanding of the molecular mechanisms of chemotherapy-induced peripheral neuropathy. Front Mol Neurosci 2024; 17:1345811. [PMID: 38660386 PMCID: PMC11039947 DOI: 10.3389/fnmol.2024.1345811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is the most common off-target adverse effects caused by various chemotherapeutic agents, such as cisplatin, oxaliplatin, paclitaxel, vincristine and bortezomib. CIPN is characterized by a substantial loss of primary afferent sensory axonal fibers leading to sensory disturbances in patients. An estimated of 19-85% of patients developed CIPN during the course of chemotherapy. The lack of preventive measures and limited treatment options often require a dose reduction or even early termination of life-saving chemotherapy, impacting treatment efficacy and patient survival. In this Review, we summarized the current understanding on the pathogenesis of CIPN. One prominent change induced by chemotherapeutic agents involves the disruption of neuronal cytoskeletal architecture and axonal transport dynamics largely influenced by the interference of microtubule stability in peripheral neurons. Due to an ineffective blood-nerve barrier in our peripheral nervous system, exposure to some chemotherapeutic agents causes mitochondrial swelling in peripheral nerves, which lead to the opening of mitochondrial permeability transition pore and cytochrome c release resulting in degeneration of primary afferent sensory fibers. The exacerbated nociceptive signaling and pain transmission in CIPN patients is often linked the increased neuronal excitability largely due to the elevated expression of various ion channels in the dorsal root ganglion neurons. Another important contributing factor of CIPN is the neuroinflammation caused by an increased infiltration of immune cells and production of inflammatory cytokines. In the central nervous system, chemotherapeutic agents also induce neuronal hyperexcitability in the spinal dorsal horn and anterior cingulate cortex leading to the development of central sensitization that causes CIPN. Emerging evidence suggests that the change in the composition and diversity of gut microbiota (dysbiosis) could have direct impact on the development and progression of CIPN. Collectively, all these aspects contribute to the pathogenesis of CIPN. Recent advances in RNA-sequencing offer solid platform for in silico drug screening which enable the identification of novel therapeutic agents or repurpose existing drugs to alleviate CIPN, holding immense promises for enhancing the quality of life for cancer patients who undergo chemotherapy and improve their overall treatment outcomes.
Collapse
Affiliation(s)
- Xinyu Chen
- Department of Neuroscience, Hong Kong Special Administrative Region (HKSAR), City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Yumeng Gan
- Department of Neuroscience, Hong Kong Special Administrative Region (HKSAR), City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Ngan Pan Bennett Au
- Department of Neuroscience, Hong Kong Special Administrative Region (HKSAR), City University of Hong Kong, Kowloon, Hong Kong SAR, China
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
- Institute of Life Sciences and Healthcare, University of Portsmouth, Portsmouth, United Kingdom
| | - Chi Him Eddie Ma
- Department of Neuroscience, Hong Kong Special Administrative Region (HKSAR), City University of Hong Kong, Kowloon, Hong Kong SAR, China
| |
Collapse
|
15
|
Lu Y, Liu S, Wang P, Guo X, Qin Z, Hou H, Tao T. A novel microglia-targeting strategy based on nanoparticle-mediated delivery of miR-26a-5p for long-lasting analgesia in chronic pain. J Nanobiotechnology 2024; 22:128. [PMID: 38519978 PMCID: PMC10960380 DOI: 10.1186/s12951-024-02420-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 03/18/2024] [Indexed: 03/25/2024] Open
Abstract
Accumulating evidence supports the notion that microglia play versatile roles in different chronic pain conditions. However, therapeutic strategies of chronic pain by targeting microglia remain largely overlooked. This study seeks to develop a miRNA-loaded nano-delivery system by targeting microglia, which could provide a decent and long-lasting analgesia for chronic pain. Surface aminated mesoporous silica nanoparticles were adopted to load miR-26a-5p, a potent analgesic miRNA, by electrostatic adsorption, which can avoid miR-26a-5p is rapidly released and degraded. Then, targeting peptide MG1 was modified on the surface of aminated mesoporous silica particles for microglia targeting. In peripheral nerve injury induced neuropathic pain model, a satisfactory anti-allodynia effect with about 6 weeks pain-relief duration were achieved through targeting microglia strategy, which decreased microglia activation and inflammation by Wnt5a, a non-canonical Wnt pathway. In inflammatory pain and chemotherapy induced peripheral neuropathic pain, microglia targeting strategy also exhibited more efficient analgesia and longer pain-relief duration than others. Overall, we developed a microglia-targeting nano-delivery system, which facilitates precisely miR-26a-5p delivery to enhance analgesic effect and duration for several chronic pain conditions.
Collapse
Affiliation(s)
- Yitian Lu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Shuai Liu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Peng Wang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xiangna Guo
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Zaisheng Qin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Honghao Hou
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Tao Tao
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
- Department of Anesthesiology, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China.
| |
Collapse
|
16
|
Whitaker EE, Mecum NE, Cott RC, Goode DJ. Expression of MHC II in DRG neurons attenuates paclitaxel-induced cold hypersensitivity in male and female mice. PLoS One 2024; 19:e0298396. [PMID: 38330029 PMCID: PMC10852343 DOI: 10.1371/journal.pone.0298396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
Chemotherapy is often a life-saving treatment, but the development of intractable pain caused by chemotherapy-induced peripheral neuropathy (CIPN) is a major dose-limiting toxicity that restricts cancer survival rates. Recent reports demonstrate that paclitaxel (PTX) robustly increases anti-inflammatory CD4+ T cells in the dorsal root ganglion (DRG), and that T cells and anti-inflammatory cytokines are protective against CIPN. However, the mechanism by which CD4+ T cells are activated, and the extent cytokines released by CD4+ T cells target DRG neurons are unknown. Here, we are the first to detect major histocompatibility complex II (MHCII) protein in mouse DRG neurons and to find CD4+ T cells breaching the satellite glial cell barrier to be in close proximity to neurons, together suggesting CD4+ T cell activation and targeted cytokine release. MHCII protein is primarily expressed in small nociceptive neurons in male and female mouse DRG but increased after PTX in small nociceptive neurons in only female DRG. Reducing one copy of MHCII in small nociceptive neurons decreased anti-inflammatory IL-10 and IL-4 producing CD4+ T cells in naïve male DRG and increased their hypersensitivity to cold. Administration of PTX to male and female mice that lacked one copy of MHCII in nociceptive neurons decreased anti-inflammatory CD4+ T cells in the DRG and increased the severity of PTX-induced cold hypersensitivity. Collectively, our results demonstrate expression of MHCII protein in mouse DRG neurons, which modulates cytokine producing CD4+ T cells in the DRG and attenuates cold hypersensitivity during homeostasis and after PTX treatment.
Collapse
Affiliation(s)
- Emily E. Whitaker
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, United States of America
| | - Neal E. Mecum
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, United States of America
| | - Riley C. Cott
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, United States of America
| | - Diana J. Goode
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, United States of America
| |
Collapse
|
17
|
Gadepalli A, Ummadisetty O, Akhilesh, Chouhan D, Yadav KE, Tiwari V. Peripheral mu-opioid receptor activation by dermorphin [D-Arg2, Lys4] (1-4) amide alleviates behavioral and neurobiological aberrations in rat model of chemotherapy-induced neuropathic pain. Neurotherapeutics 2024; 21:e00302. [PMID: 38241153 PMCID: PMC10903092 DOI: 10.1016/j.neurot.2023.10.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 01/21/2024] Open
Abstract
Paclitaxel, a frequently utilized chemotherapeutic agent, often gives rise to severe and distressing sensory neuropathy in patients undergoing chemotherapy. Unfortunately, current therapeutics for chemotherapy-induced neuropathic pain (CINP) demonstrate limited effectiveness and are burdened with the potential for central side effects such as sedation, respiratory depression, cognitive impairment, and addiction, posing substantial clinical challenges. In light of these limitations, present study is designed to investigate the therapeutic potential of Dermorphin [D-Arg2, Lys4] (1-4) amide (DALDA), a preferential peripherally acting mu-opioid receptor agonist, in rat model of CINP. The primary objective was to assess the analgesic properties of DALDA and elucidate the underlying mechanisms governing its therapeutic activity. Our findings revealed that DALDA treatment significantly ameliorated paclitaxel-induced evoked and spontaneous ongoing pain in rats without causing drug addiction and other central side effects. Molecular analyses further unveiled that paclitaxel administration resulted in increased expression of TRP channels, NR2B, voltage-gated sodium channels (VGSCs) and neuroinflammatory markers in both the dorsal root ganglion (DRG) and the spinal cord (L4-L5 region) of rats. DALDA treatment significantly downregulated ion channels (TRPs, VGSCs) and NR2B expressions, concomitant with the inhibition of microglial activation, resulting in the suppression of oxido-nitrosative stress and neuroinflammatory cascade. Findings from the current study suggests that peripheral mu-opioid receptors may offer a potential target for the treatment of patients suffering from CINP, offering new avenues for improved pain relief while minimizing central side effects.
Collapse
Affiliation(s)
- Anagha Gadepalli
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| | - Obulapathi Ummadisetty
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| | - Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| | - Deepak Chouhan
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| | - Krushna Eknath Yadav
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
18
|
Akhilesh, Uniyal A, Mehta A, Tiwari V. Combination chemotherapy in rodents: a model for chemotherapy-induced neuropathic pain and pharmacological screening. Metab Brain Dis 2024; 39:43-65. [PMID: 37991674 DOI: 10.1007/s11011-023-01315-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/09/2023] [Indexed: 11/23/2023]
Abstract
Chemotherapy-induced neuropathic pain (CINP) remains a therapeutic challenge, with no US-FDA approved drugs or effective treatments available. Despite significant progress in unravelling the pathophysiology of CINP, the clinical translation of this knowledge into tangible outcome remains elusive. Here, we employed behavioural and pharmacological approaches to establish and validate a novel combination-based chemotherapeutic model of peripheral neuropathy. Male Sprague Dawley rats were subjected to chemotherapy administration followed by assessment of pain behaviour at different time-points post-chemotherapy. Paclitaxel-treated animals displayed an enhanced thermal and mechanical hypersensitivity from day four onwards which continued till day thirty-five post last paclitaxel injection. Notably, rats subjected to combination chemotherapy, displayed prolonged hypersensitivity that emerged on day four and persisted until day fifty-six. RT-PCR analysis revealed significant upregulation in DRG and spinal mRNA expressions of TRP channels (TRPA1, TRPV1, & TRPM8), pro-inflammatory cytokines (TNF-α & IL-1β) and neuropeptides, Substance P and CGRP in both the pain models. Interestingly, the combination chemotherapy model demonstrated a significant increase in DRG and spinal NR2B expressions compared to rats solely treated with paclitaxel. Pharmacological investigations revealed that gabapentin treatment substantially mitigates pain hypersensitivity in both the combined chemotherapy and paclitaxel-administered groups, with the simultaneous reversal of cellular and molecular changes observed in the lumbar DRG and spinal cord of rats. The findings from this study suggests that combination chemotherapy model exhibits heightened and prolonged hypersensitivity in comparison to the conventional paclitaxel-induced neuropathic pain model. This model not only recapitulates clinical biomarkers of neuropathy but also presents a potential alternative platform for screening analgesic drugs targeted at CINP.
Collapse
Affiliation(s)
- Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, 221005, India
| | - Ankit Uniyal
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, 221005, India
| | - Anuj Mehta
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, 221005, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, 221005, India.
| |
Collapse
|
19
|
Bacalhau C, Costa-Pereira JT, Tavares I. Preclinical research in paclitaxel-induced neuropathic pain: a systematic review. Front Vet Sci 2023; 10:1264668. [PMID: 38188718 PMCID: PMC10766764 DOI: 10.3389/fvets.2023.1264668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/21/2023] [Indexed: 01/09/2024] Open
Abstract
Introduction Chemotherapy-induced peripheral neuropathy (CIPN) is a common consequence of cancer treatment and pain is a frequent complaint of the patients. Paclitaxel, a cytostatic drug, generates a well-described peripheral nerve injury and neuroinflammation, which may be experimentally mimicked in animal models. We conducted a systematic review analyzing the experimental design, reporting and mechanisms underlying paclitaxel-induced neuropathy in the included studies to establish the perspectives of translation of the current literature in models of CIPN. Methods We elected studies published in Pubmed and Scopus between 1 January 2018 and 3 December 2022. Results According to a defined mesh of keywords searched, and after applying exclusion and inclusion criteria, 70 original studies were included and analyzed in detail. Most studies used male Sprague-Dawley rats to induce paclitaxel-induced neuropathy, used low doses of paclitaxel, and the analyzed studies mainly focused at 14-28 days of CIPN. Mechanical nociceptive tests were preferred in the behavioral evaluation. The mechanisms under study were mainly neuroinflammation of peripheral nerves. The overall methodological quality was considered moderate, and the risk of bias was unclear. Discussion Despite the ample preclinical research in paclitaxel-induced neuropathy, this systematic review alerts to some flaws in the experimental design along with limitations in reporting, e.g., lack of representation of both sexes in experimental work and the lack of reporting of the ARRIVE guidelines. This may limit the reproducibility of preclinical studies in CIPN. In addition, the clinical features of CIPN should be considered when designing animal experiments, such as sex and age of the CIPN patients. In this way the experimental studies aiming to establish the mechanisms of CIPN may allow the development of new drugs to treat CIPN and translation in the research of CIPN could be improved.
Collapse
Affiliation(s)
- Carolina Bacalhau
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - José Tiago Costa-Pereira
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Porto, Portugal
- I3S-Institute of Investigation and Innovation in Health, University of Porto, Porto, Portugal
- Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal
| | - Isaura Tavares
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Porto, Portugal
- I3S-Institute of Investigation and Innovation in Health, University of Porto, Porto, Portugal
| |
Collapse
|
20
|
Xu J, Huang P, Bie B, Dai Y, Ben-Salem S, Borjini N, Zhang L, Chen J, Olman M, Cheng J, Lin F. Complement Receptor C3aR1 Contributes to Paclitaxel-Induced Peripheral Neuropathic Pain in Mice and Rats. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1736-1746. [PMID: 37861348 PMCID: PMC10841827 DOI: 10.4049/jimmunol.2300252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 09/22/2023] [Indexed: 10/21/2023]
Abstract
Cancer chemotherapy-induced neuropathic pain is a devastating pain syndrome without effective therapies. We previously reported that rats deficient in complement C3, the central component of complement activation cascade, showed a reduced degree of paclitaxel-induced mechanical allodynia (PIMA), suggesting that complement is integrally involved in the pathogenesis of this model. However, the underlying mechanism was unclear. Complement activation leads to the production of C3a, which mediates inflammation through its receptor C3aR1. In this article, we report that the administration of paclitaxel induced a significantly higher expression level of C3aR1 on dorsal root ganglion (DRG) macrophages and expansion of these macrophages in DRGs in wild-type (WT) compared with in C3aR1 knockout (KO) mice. We also found that paclitaxel induced less severe PIMA, along with a reduced DRG expression of transient receptor potential channels of the vanilloid subtype 4 (TRPV4), an essential mediator for PIMA, in C3aR1 KO than in WT mice. Treating WT mice or rats with a C3aR1 antagonist markedly attenuated PIMA in association with downregulated DRG TRPV4 expression, reduced DRG macrophages expansion, suppressed DRG neuron hyperexcitability, and alleviated peripheral intraepidermal nerve fiber loss. Administration of C3aR1 antagonist to TRPV4 KO mice further protected them from PIMA. These results suggest that complement regulates PIMA development through C3aR1 to upregulate TRPV4 on DRG neurons and promote DRG macrophage expansion. Targeting C3aR1 could be a novel therapeutic approach to alleviate this debilitating pain syndrome.
Collapse
Affiliation(s)
- Jijun Xu
- Department of Pain Management, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Ping Huang
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Bihua Bie
- Department of Pain Management, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Yang Dai
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Salma Ben-Salem
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Nozha Borjini
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Lingjun Zhang
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jin Chen
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Mitchell Olman
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jianguo Cheng
- Department of Pain Management, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
- Department of Neurosciences, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Feng Lin
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
21
|
Gadepalli A, Ummadisetty O, Akhilesh, Chouhan D, Anmol, Tiwari V. Loperamide, a peripheral Mu-Opioid receptor agonist, attenuates chemotherapy-induced neuropathic pain in rats. Int Immunopharmacol 2023; 124:110944. [PMID: 37801968 DOI: 10.1016/j.intimp.2023.110944] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/28/2023] [Accepted: 09/12/2023] [Indexed: 10/08/2023]
Abstract
Opioids are employed in the management of chemotherapy-induced neuropathic pain (CINP) when other pain management approaches have failed and proven ineffective. However, their use in CINP is generally considered as a second-line or adjunctive therapy owing to their central side effects and development of tolerance with their long-term usage. Targeting peripheral sites may offer several advantages over the conventional CNS-based approaches as peripheral targets modulate pain signals at their source, thereby relieving pain with higher specificity, efficacy and minimizing adverse effects associated with off-site CNS actions. Therefore, present study was designed with an aim to investigate the effect of loperamide, a peripherally acting mu-opioid receptor agonist, on paclitaxel-induced neuropathic pain in rats and elucidate its underlying mechanism. Loperamide treatment significantly attenuated mechanical, and cold hypersensitivity and produced significant place preference behaviour in neuropathic rats indicating its potential to treat both evoked and spontaneous pain. More importantly, loperamide treatment in naïve rats did not produce place preference to drug-paired chamber pointing towards its non-addictive analgesic potential. Further, molecular investigations revealed increased expression of ion channels such as TRPA1, TRPM8; voltage-gated sodium channels (VGSCs) and neuroinflammatory markers in the dorsal root ganglion (DRG) and lumbar (L4-L5) spinal cord of neuropathic rats, which was significantly downregulated upon loperamide treatment. These findings collectively suggest that activation of peripheral mu-opioid receptors contributes to the amelioration of both evoked and spontaneous pain in neuropathic rats by downregulating TRP channels and VGSCs along with suppression of oxido-nitrosative stress and neuroinflammatory cascade.
Collapse
Affiliation(s)
- Anagha Gadepalli
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| | - Obulapathi Ummadisetty
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| | - Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| | - Deepak Chouhan
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| | - Anmol
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
22
|
Cristiano C, Giorgio C, Cocchiaro P, Boccella S, Cesta MC, Castelli V, Liguori FM, Cuozzo MR, Brandolini L, Russo R, Allegretti M. Inhibition of C5aR1 as a promising approach to treat taxane-induced neuropathy. Cytokine 2023; 171:156370. [PMID: 37722320 DOI: 10.1016/j.cyto.2023.156370] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/19/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a common side effect of several antitumor agents resulting in progressive and often irreversible damage of peripheral nerves. In addition to their known anticancer effects, taxanes, including paclitaxel, can also induce peripheral neuropathy by activating microglia and astrocytes, which release pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin 1-beta (IL-1β), and chemokine (C-C motif) ligand 2 (CCL-2). All these events contribute to the maintenance of neuropathic or inflammatory response. Complement component 5a (C5a)/C5a receptor 1 (C5aR1) signaling was very recently shown to play a crucial role in paclitaxel-induced peripheral neuropathy. Our recent findings highlighted that taxanes have the previously unreported property of binding and activating C5aR1, and that C5aR1 inhibition by DF3966A is effective in preventing paclitaxel-induced peripheral neuropathy (PIPN) in animal models. Here, we investigated if C5aR1 inhibition maintains efficacy in reducing PIPN in a therapeutic setting. Furthermore, we characterized the role of C5aR1 activation by paclitaxel and the CIPN-associated activation of nod-like receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasome. Our results clearly show that administration of the C5aR1 inhibitor strongly reduced cold and mechanical allodynia in mice when given both during the onset of PIPN and when neuropathy is well established. C5aR1 activation by paclitaxel was found to be a key event in the induction of inflammatory factors in spinal cord, such as TNF-α, ionized calcium-binding adapter molecule 1 (Iba-1), and glial fibrillary acidic protein (GFAP). In addition, C5aR1 inhibition significantly mitigated paclitaxel-induced inflammation and inflammasome activation by reducing IL-1β and NLRP3 expression at both sciatic and dorsal root ganglia level, confirming the involvement of inflammasome in PIPN. Moreover, paclitaxel-induced upregulation of C5aR1 was significantly reduced by DF3966A treatment in central nervous system. Lastly, the antinociceptive effect of C5aR1 inhibition was confirmed in an in vitro model of sensory neurons in which we focused on receptor channels usually activated upon neuropathy. In conclusion, C5aR1 inhibition is proposed as a therapeutic option with the potential to exert long-term protective effect on PIPN-associated neuropathic pain and inflammation.
Collapse
Affiliation(s)
- C Cristiano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - C Giorgio
- Dompé farmaceutici SpA, Via De Amicis, 80131 Naples, Italy
| | - P Cocchiaro
- Dompé farmaceutici SpA, Via De Amicis, 80131 Naples, Italy
| | - S Boccella
- Dompé farmaceutici SpA, Via De Amicis, 80131 Naples, Italy
| | - M C Cesta
- Dompé farmaceutici SpA, Via Campo di Pile, 67100 L'Aquila, Italy
| | - V Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - F M Liguori
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - M R Cuozzo
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - L Brandolini
- Dompé farmaceutici SpA, Via Campo di Pile, 67100 L'Aquila, Italy
| | - R Russo
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - M Allegretti
- Dompé farmaceutici SpA, Via Campo di Pile, 67100 L'Aquila, Italy.
| |
Collapse
|
23
|
Yang X, Huang X, Lu W, Yan F, Ye Y, Wang L, Tang X, Zeng W, Huang J, Xie J. Transcriptome Profiling of miRNA-mRNA Interactions and Associated Mechanisms in Chemotherapy-Induced Neuropathic Pain. Mol Neurobiol 2023; 60:5672-5690. [PMID: 37332017 DOI: 10.1007/s12035-023-03398-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/23/2023] [Indexed: 06/20/2023]
Abstract
Chemotherapy-induced neuropathic pain (CINP) is a dose-limiting adverse event affecting 40% of chemotherapy patients. MiRNA-mRNA interaction plays an important role in various processes. However, detailed profiling of miRNA-mRNA interactions in CINP remains unclear. Here, a rat-based CINP model was established using paclitaxel, followed by nociceptive behavioral tests related to mechanical allodynia, thermal hyperalgesia, and cold allodynia. The landscape of miRNA-mRNA interaction in the spinal dorsal horn was investigated through mRNA transcriptomics and small RNA sequencing. Under CINP condition, 86 differentially expressed mRNAs and 56 miRNAs were identified. Gene Set Enrichment Analysis (GSEA), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses indicated the activity of Odorant binding, postsynaptic specialization and synaptic density, extracellular matrix, mitochondrial matrix, retrograde endocannabinoid signaling, and GTPase activity. Protein-protein interaction (PPI), networks of circRNA-miRNA-mRNA, lncRNA-miRNA-mRNA, and TF-genes were demonstrated. We next explored the immune infiltration microenvironment and found a higher infiltration abundance of Th17 and a lower abundance of MDSC in CINP. RT-qPCR and dual-luciferase assays were used to verify the sequencing results, and single-cell analysis based on the SekSeeq database was conducted. Combined with bioinformatics analyses and experimental validations, Mpz, a protein-coding gene specifically expressed in Schwann cells, was found critical in maintaining CINP under miRNA regulation. Therefore, these data highlight the expression patterns of miRNA-mRNA, and the underlying mechanism in the spinal dorsal horn under CINP condition, and Mpz may serve as a promising therapeutic target for patients with CINP.
Collapse
Affiliation(s)
- Xiaohua Yang
- State Key Laboratory of Oncology in Southern China, Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Xiqiang Huang
- Department of Anesthesiology, Zhongshan People's Hospital, Zhongshan, 528400, Guangdong, China
| | - Weicheng Lu
- State Key Laboratory of Oncology in Southern China, Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Fang Yan
- State Key Laboratory of Oncology in Southern China, Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Yaqi Ye
- State Key Laboratory of Oncology in Southern China, Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Linjie Wang
- Department of Human Anatomy and Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510060, Guangdong, China
| | - Xiaole Tang
- State Key Laboratory of Oncology in Southern China, Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Weian Zeng
- State Key Laboratory of Oncology in Southern China, Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Jingxiu Huang
- State Key Laboratory of Oncology in Southern China, Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China.
| | - Jingdun Xie
- State Key Laboratory of Oncology in Southern China, Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China.
| |
Collapse
|
24
|
Whitaker EE, Mecum NE, Cott RC, Goode DJ. Novel expression of major histocompatibility complex II in dorsal root ganglion neurons attenuates paclitaxel-induced cold hypersensitivity in male and female mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.31.535136. [PMID: 37066176 PMCID: PMC10103942 DOI: 10.1101/2023.03.31.535136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Chemotherapy is often a life-saving treatment, but the development of intractable pain caused by chemotherapy-induced peripheral neuropathy (CIPN) is a major dose-limiting toxicity that restricts survival rates. Recent reports demonstrate that paclitaxel (PTX) robustly increases anti-inflammatory CD4+ T cells in the dorsal root ganglion (DRG), and that T cells and anti-inflammatory cytokines are protective against CIPN. However, the mechanism by which CD4+ T cells are activated, and the extent cytokines released by CD4+ T cells target DRG neurons are unknown. Here, we found novel expression of functional major histocompatibility complex II (MHCII) protein in DRG neurons, and CD4+ T cells in close proximity to DRG neurons, together suggesting CD4+ T cell activation and targeted cytokine release. MHCII protein is primarily expressed in small nociceptive neurons in male mouse DRG regardless of PTX, while MHCII is induced in small nociceptive neurons in female DRG after PTX. Accordingly, reducing MHCII in small nociceptive neurons increased hypersensitivity to cold only in naive male mice, but increased severity of PTX-induced cold hypersensitivity in both sexes. Collectively, our results demonstrate expression of MHCII on DRG neurons and a functional role during homeostasis and inflammation.
Collapse
|
25
|
Cao F, Wang X, Ye Q, Yan F, Lu W, Xie J, Bi B, Wang X. Identifying circRNA-miRNA-mRNA Regulatory Networks in Chemotherapy-Induced Peripheral Neuropathy. Curr Issues Mol Biol 2023; 45:6804-6822. [PMID: 37623249 PMCID: PMC10453290 DOI: 10.3390/cimb45080430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/13/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a frequent and severe side effect of first-line chemotherapeutic agents. The association between circular RNAs (circRNAs) and CIPN remains unclear. In this study, CIPN models were constructed with Taxol, while 134 differentially expressed circRNAs, 353 differentially expressed long non-coding RNAs, and 86 differentially expressed messenger RNAs (mRNAs) were identified utilizing RNA sequencing. CircRNA-targeted microRNAs (miRNAs) were predicted using miRanda, and miRNA-targeted mRNAs were predicted using TargetScan and miRDB. The intersection of sequencing and mRNA prediction results was selected to establish the circRNA-miRNA-mRNA networks, which include 15 circRNAs, 18 miRNAs, and 11 mRNAs. Functional enrichment pathway analyses and immune infiltration analyses revealed that differentially expressed mRNAs were enriched in the immune system, especially in T cells, monocytes, and macrophages. Cdh1, Satb2, Fas, P2ry2, and Zfhx2 were further identified as hub genes and validated by RT-qPCR, correlating with macrophages, plasmacytoid dendritic cells, and central memory CD4 T cells in CIPN. Additionally, we predicted the associated diseases, 36 potential transcription factors (TFs), and 30 putative drugs for hub genes using the DisGeNET, TRRUST, and DGIdb databases, respectively. Our results indicated the crucial role of circRNAs, and the immune microenvironment played in CIPN, providing novel insights for further research.
Collapse
Affiliation(s)
- Fei Cao
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (F.C.); (X.W.); (Q.Y.); (F.Y.); (W.L.); (J.X.)
| | - Xintong Wang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (F.C.); (X.W.); (Q.Y.); (F.Y.); (W.L.); (J.X.)
| | - Qingqing Ye
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (F.C.); (X.W.); (Q.Y.); (F.Y.); (W.L.); (J.X.)
| | - Fang Yan
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (F.C.); (X.W.); (Q.Y.); (F.Y.); (W.L.); (J.X.)
| | - Weicheng Lu
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (F.C.); (X.W.); (Q.Y.); (F.Y.); (W.L.); (J.X.)
| | - Jingdun Xie
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (F.C.); (X.W.); (Q.Y.); (F.Y.); (W.L.); (J.X.)
| | - Bingtian Bi
- Department of Clinical Trial Center, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xudong Wang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (F.C.); (X.W.); (Q.Y.); (F.Y.); (W.L.); (J.X.)
| |
Collapse
|
26
|
Kim N, Chung G, Son SR, Park JH, Lee YH, Park KT, Cho IH, Jang DS, Kim SK. Magnolin Inhibits Paclitaxel-Induced Cold Allodynia and ERK1/2 Activation in Mice. PLANTS (BASEL, SWITZERLAND) 2023; 12:2283. [PMID: 37375908 DOI: 10.3390/plants12122283] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a common side effect of anti-cancer drugs. The main symptoms often include sensory disturbances and neuropathic pain, and currently there is no effective treatment for this condition. This study aimed to investigate the suppressive effects of magnolin, an extracellular signal-regulated kinase (ERK) inhibitor substance derived from a 95% EtOH extract of the seeds of Magnolia denudata, on the symptoms of CIPN. A taxol-based anti-cancer drug paclitaxel (PTX) was repeatedly injected (2 mg/kg/day, total 8 mg/kg) into mice to induce CIPN. A neuropathic pain symptom was assessed using a cold allodynia test that scores behaviors of licking and shaking paw after plantar administration of acetone drop. Magnolin was administered intraperitoneally (0.1, 1, or 10 mg/kg) and behavioral changes to acetone drop were measured. The effect of magnolin administration on ERK expression in the dorsal root ganglion (DRG) was investigated using western blot analysis. The results showed that the repeated injections of PTX induced cold allodynia in mice. Magnolin administration exerted an analgesic effect on the PTX-induced cold allodynia and inhibited the ERK phosphorylation in the DRG. These results suggest that magnolin could be developed as an alternative treatment to suppress paclitaxel-induced neuropathic pain symptoms.
Collapse
Affiliation(s)
- Nari Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Geehoon Chung
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - So-Ri Son
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jae Hyun Park
- Department of East-West Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Young Hyun Lee
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Keon-Tae Park
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ik-Hyun Cho
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Dae Sik Jang
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sun Kwang Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of East-West Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
27
|
Huang Y, Chen H, Jin D, Chen SR, Pan HL. NMDA Receptors at Primary Afferent-Excitatory Neuron Synapses Differentially Sustain Chemotherapy- and Nerve Trauma-Induced Chronic Pain. J Neurosci 2023; 43:3933-3948. [PMID: 37185237 PMCID: PMC10217996 DOI: 10.1523/jneurosci.0183-23.2023] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/30/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
The spinal dorsal horn contains vesicular glutamate transporter-2 (VGluT2)-expressing excitatory neurons and vesicular GABA transporter (VGAT)-expressing inhibitory neurons, which normally have different roles in nociceptive transmission. Spinal glutamate NMDAR hyperactivity is a crucial mechanism of chronic neuropathic pain. However, it is unclear how NMDARs regulate primary afferent input to spinal excitatory and inhibitory neurons in neuropathic pain. Also, the functional significance of presynaptic NMDARs in neuropathic pain has not been defined explicitly. Here we showed that paclitaxel treatment or spared nerve injury (SNI) similarly increased the NMDAR-mediated mEPSC frequency and dorsal root-evoked EPSCs in VGluT2 dorsal horn neurons in male and female mice. By contrast, neither paclitaxel nor SNI had any effect on mEPSCs or evoked EPSCs in VGAT neurons. In mice with conditional Grin1 (gene encoding GluN1) KO in primary sensory neurons (Grin1-cKO), paclitaxel treatment failed to induce pain hypersensitivity. Unexpectedly, SNI still caused long-lasting pain hypersensitivity in Grin1-cKO mice. SNI increased the amplitude of puff NMDA currents in VGluT2 neurons and caused similar depolarizing shifts in GABA reversal potentials in WT and Grin1-cKO mice. Concordantly, spinal Grin1 knockdown diminished SNI-induced pain hypersensitivity. Thus, presynaptic NMDARs preferentially amplify primary afferent input to spinal excitatory neurons in neuropathic pain. Although presynaptic NMDARs are required for chemotherapy-induced pain hypersensitivity, postsynaptic NMDARs in spinal excitatory neurons play a dominant role in traumatic nerve injury-induced chronic pain. Our findings reveal the divergent synaptic connectivity and functional significance of spinal presynaptic and postsynaptic NMDARs in regulating cell type-specific nociceptive input in neuropathic pain with different etiologies.SIGNIFICANCE STATEMENT Spinal excitatory neurons relay input from nociceptors, whereas inhibitory neurons repress spinal nociceptive transmission. Chronic nerve pain is associated with aberrant NMDAR activity in the spinal dorsal horn. This study demonstrates, for the first time, that chemotherapy and traumatic nerve injury preferentially enhance the NMDAR activity at primary afferent-excitatory neuron synapses but have no effect on primary afferent input to spinal inhibitory neurons. NMDARs in primary sensory neurons are essential for chemotherapy-induced chronic pain, whereas nerve trauma causes pain hypersensitivity predominantly via postsynaptic NMDARs in spinal excitatory neurons. Thus, presynaptic and postsynaptic NMDARs at primary afferent-excitatory neuron synapses are differentially engaged in chemotherapy- and nerve injury-induced chronic pain and could be targeted respectively for treating these painful conditions.
Collapse
Affiliation(s)
- Yuying Huang
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Hong Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Daozhong Jin
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Shao-Rui Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Hui-Lin Pan
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| |
Collapse
|
28
|
Lobos N, Lux S, Zepeda RJ, Pelissier T, Marcos JL, Bustos-Quevedo G, Hernández A, Constandil L. Rosuvastatin Synergistically Enhances the Antinociceptive Efficacy of Duloxetine in Paclitaxel-Induced Neuropathic Pain in Mice. Int J Mol Sci 2023; 24:ijms24098359. [PMID: 37176065 PMCID: PMC10179025 DOI: 10.3390/ijms24098359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Paclitaxel, a widely used cancer chemotherapeutic agent, has high incidence of neurotoxicity associated with the production of neuropathic pain, for which only duloxetine has shown significant but moderate analgesic effect. Since statins, classically used to reduce hypercholesterolemia, have shown antinociceptive effect in preclinical studies on neuropathic pain, we studied whether the antinociceptive efficacy of duloxetine could be synergistically potentiated by rosuvastatin in a model of paclitaxel-induced neuropathy in mice. The astrocytic and microglial responses in the spinal cord of paclitaxel-treated mice were also assessed by measuring GFAP and CD11b proteins, respectively. Paclitaxel treatment did not impair motor coordination and balance in rotarod testing. Rosuvastatin, duloxetine, and the rosuvastatin/duloxetine combination (combined at equieffective doses) dose-dependently decreased mechanical allodynia (ED30, von Frey testing) and thermal hyperalgesia (ED50, hot plate testing) in paclitaxel-treated mice. Isobolographic analysis showed a superadditive interaction for rosuvastatin and duloxetine, as both the ED30 and ED50 for the rosuvastatin/duloxetine combination contained only a quarter of each drug compared to the individual drugs. The rosuvastatin/duloxetine combination reversed paclitaxel-induced GFAP overexpression, indicating that such effects might depend in part on astrocyte inactivation. Results suggest that statins could be useful in synergistically enhancing the efficacy of duloxetine in some chemotherapy-induced neuropathic conditions.
Collapse
Affiliation(s)
- Nicolás Lobos
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago 9170022, Chile
| | - Sebastián Lux
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago 9170022, Chile
- Critical Care Unit, Barros Luco Trudeau Hospital, Santiago 8900085, Chile
| | - Ramiro Javier Zepeda
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Teresa Pelissier
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago 9170022, Chile
| | - José Luis Marcos
- Escuela de Ciencias Agrícolas y Veterinarias, Universidad Viña del Mar, Viña del Mar 2572007, Chile
| | - Gonzalo Bustos-Quevedo
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago 9170022, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Santiago 9170124, Chile
| | - Alejandro Hernández
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago 9170022, Chile
| | - Luis Constandil
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago 9170022, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Santiago 9170124, Chile
| |
Collapse
|
29
|
Shinouchi R, Sasaki A, Takaki T, Tsuji M, Kiuchi Y, Nobe K. The effect of hand therapy on alleviating chemotherapy-induced peripheral neuropathy in a model mouse. Neurosci Lett 2023; 800:137138. [PMID: 36813075 DOI: 10.1016/j.neulet.2023.137138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/04/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023]
Abstract
The use of neurotoxic chemotherapeutic agents induces numbness in the limbs through chemotherapy-induced peripheral neuropathy (CIPN). Recently, we found that hand therapy involving finger massage improved mild to moderate numbness in CIPN patients. In this study, we behaviorally, physiologically, pathologically, and histologically investigated the mechanisms underlying hand therapy-induced numbness improvement in a CIPN model mouse. Hand therapy was performed for 21 days after the disease induction. Its effects were evaluated using mechanical and thermal thresholds and blood flow in the bilateral hind paw. Moreover, 14 days after the hand therapy was administered, we assessed the blood flow and conduction velocity in the sciatic nerve, the level of serum galectin-3, and the histological myelin and epidermis-related changes in the hindfoot tissue. Hand therapy significantly improved allodynia, hyperalgesia, blood flow, conduction velocity, serum galectin-3, and epidermal thickness in the CIPN model mouse. Furthermore, we observed the images of repairs of the myelin degeneration. Thus, we found that hand therapy could improve numbness in the CIPN model mouse and that it could help to repair peripheral nerves by promoting blood circulation in the limbs.
Collapse
Affiliation(s)
- Ryosuke Shinouchi
- Division of Pharmacology, Department of Pharmacology, Toxicology and Therapeutics, School of Pharmacy, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan.
| | - Akiko Sasaki
- Department of Pharmacology, School of Medicine, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Takashi Takaki
- Section of Electron Microscopy, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Mayumi Tsuji
- Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Yuji Kiuchi
- Department of Pharmacology, School of Medicine, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Koji Nobe
- Division of Pharmacology, Department of Pharmacology, Toxicology and Therapeutics, School of Pharmacy, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| |
Collapse
|
30
|
Haroun R, Wood JN, Sikandar S. Mechanisms of cancer pain. FRONTIERS IN PAIN RESEARCH 2023; 3:1030899. [PMID: 36688083 PMCID: PMC9845956 DOI: 10.3389/fpain.2022.1030899] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/14/2022] [Indexed: 01/05/2023] Open
Abstract
Personalised and targeted interventions have revolutionised cancer treatment and dramatically improved survival rates in recent decades. Nonetheless, effective pain management remains a problem for patients diagnosed with cancer, who continue to suffer from the painful side effects of cancer itself, as well as treatments for the disease. This problem of cancer pain will continue to grow with an ageing population and the rapid advent of more effective therapeutics to treat the disease. Current pain management guidelines from the World Health Organisation are generalised for different pain severities, but fail to address the heterogeneity of mechanisms in patients with varying cancer types, stages of disease and treatment plans. Pain is the most common complaint leading to emergency unit visits by patients with cancer and over one-third of patients that have been diagnosed with cancer will experience under-treated pain. This review summarises preclinical models of cancer pain states, with a particular focus on cancer-induced bone pain and chemotherapy-associated pain. We provide an overview of how preclinical models can recapitulate aspects of pain and sensory dysfunction that is observed in patients with persistent cancer-induced bone pain or neuropathic pain following chemotherapy. Peripheral and central nervous system mechanisms of cancer pain are discussed, along with key cellular and molecular mediators that have been highlighted in animal models of cancer pain. These include interactions between neuronal cells, cancer cells and non-neuronal cells in the tumour microenvironment. Therapeutic targets beyond opioid-based management are reviewed for the treatment of cancer pain.
Collapse
Affiliation(s)
- Rayan Haroun
- Division of Medicine, Wolfson Institute of Biomedical Research, University College London, London, United Kingdom
| | - John N Wood
- Division of Medicine, Wolfson Institute of Biomedical Research, University College London, London, United Kingdom
| | - Shafaq Sikandar
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
31
|
Sharma KK, Fatima N, Ali Z, Moshin M, Chandra P, Verma A, Goshain O, Kumar G. Neuropathy, its Profile and Experimental Nerve Injury Neuropathic Pain Models: A Review. Curr Pharm Des 2023; 29:3343-3356. [PMID: 38058089 DOI: 10.2174/0113816128274200231128065425] [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: 08/11/2023] [Revised: 10/17/2023] [Accepted: 11/06/2023] [Indexed: 12/08/2023]
Abstract
Neuropathy is a terrible disorder that has a wide range of etiologies. Drug-induced neuropathy, which happens whenever a chemical agent damages the peripheral nerve system, has been linked here to the iatrogenic creation of some drugs. It is potentially permanent and causes sensory impairments and paresthesia that typically affects the hands, feet, and stockings; motor participation is uncommon. It might appear suddenly or over time, and the long-term outlook varies. The wide range of chronic pain conditions experienced by people has been one of the main obstacles to developing new, more effective medications for the treatment of neuropathic pain. Animal models can be used to examine various neuropathic pain etiologies and symptoms. Several models investigate the peripheral processes of neuropathic pain, whereas some even investigate the central mechanisms, such as drug induce models like vincristine, cisplatin, bortezomib, or thalidomide, etc., and surgical models like sciatic nerve chronic constriction injury (CCI), sciatic nerve ligation through spinal nerve ligation (SNL), sciatic nerve damage caused by a laser, SNI (spared nerve injury), etc. The more popular animal models relying on peripheral nerve ligatures are explained. In contrast to chronic sciatic nerve contraction, which results in behavioral symptoms of less reliable stressful neuropathies, (SNI) spared nerve injury generates behavioral irregularities that are more feasible over a longer period. This review summarizes the latest methods models as well as clinical ideas concerning this mechanism. Every strongest current information on neuropathy is discussed, along with several popular laboratory models for causing neuropathy.
Collapse
Affiliation(s)
- Krishana Kumar Sharma
- Department of Pharmacology, Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, Uttar Pradesh 244001, India
| | - Nishat Fatima
- Department of Pharmacology, Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, Uttar Pradesh 244001, India
| | - Zeeshan Ali
- Department of Pharmacology, Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, Uttar Pradesh 244001, India
| | - Mohd Moshin
- Department of Pharmacology, Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, Uttar Pradesh 244001, India
| | - Phool Chandra
- Department of Pharmacology, Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, Uttar Pradesh 244001, India
| | - Anurag Verma
- Department of Pharmacology, Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, Uttar Pradesh 244001, India
| | - Omprakash Goshain
- Department of Pharmacology, Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, Uttar Pradesh 244001, India
| | - Gajendra Kumar
- Department of Chemistry, Constituent Government College (M.J.P.R.U.), Hasanpur, Uttar Pradesh 244241, India
| |
Collapse
|
32
|
Padín JF, Maroto M, Entrena JM, Egea J, Montell E, Vergés J, López MG, Cobos EJ, García AG. Small Synthetic Hyaluronan Disaccharide BIS014 Mitigates Neuropathic Pain in Mice. THE JOURNAL OF PAIN 2023; 24:68-83. [PMID: 36087908 DOI: 10.1016/j.jpain.2022.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/18/2022] [Accepted: 07/31/2022] [Indexed: 02/08/2023]
Abstract
Neuropathic pain (NP) is a challenging condition to treat, as the need for new drugs to treat NP is an unmet goal. We investigated the analgesic potential of a new sulfated disaccharide compound, named BIS014. Oral administration (p.o.) of this compound induced ameliorative effects in formalin-induced nociception and capsaicin-induced secondary mechanical hypersensitivity in mice, but also after partial sciatic nerve transection (spared nerve injury), chemotherapy (paclitaxel)-induced NP, and diabetic neuropathy induced by streptozotocin. Importantly, BIS014, at doses active on neuropathic hypersensitivity (60 mg/kg/p.o.), did not alter exploratory activity or motor coordination (in the rotarod test), unlike a standard dose of gabapentin (40 mg/kg/p.o.) which although inducing antiallodynic effects on the NP models, it also markedly decreased exploration and motor coordination. In docking and molecular dynamic simulation studies, BIS014 interacted with TRPV1, a receptor involved in pain transmission where it behaved as a partial agonist. Additionally, similar to capsaicin, BIS014 increased cytosolic Ca2+ concentration ([Ca2+]c) in neuroblastoma cells expressing TRPV1 receptors; these elevations were blocked by ruthenium red. BIS014 did not block capsaicin-elicited [Ca2+]c transients, but inhibited the increase in the firing rate of action potentials in bradykinin-sensitized dorsal root ganglion neurons stimulated with capsaicin. Perspective: We report that the oral administration of a new sulfated disaccharide compound, named BIS014, decreases neuropathic pain from diverse etiology in mice. Unlike the comparator gabapentin, BIS014 does not induce sedation. Thus, BIS014 has the potential to become a new efficacious non-sedative oral medication for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Juan-Fernando Padín
- Instituto-Fundación Teófilo Hernando, C/ Faraday 7, Parque Científico del Campus de Cantoblanco, Universidad Autónoma de Madrid, Madrid, Spain; Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Avda. Arzobispo Morcillo 4, Madrid, Spain; Departamento de Ciencias Médicas (Farmacología), Facultad de Medicina, Universidad de Castilla-La Mancha, Ciudad Real, Spain.
| | - Marcos Maroto
- Instituto-Fundación Teófilo Hernando, C/ Faraday 7, Parque Científico del Campus de Cantoblanco, Universidad Autónoma de Madrid, Madrid, Spain.
| | - José Manuel Entrena
- Unidad de Análisis de Comportamiento Animal, Centro de Instrumentación Científica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Armilla, Granada, Spain.
| | - Javier Egea
- Instituto de Investigación Sanitaria del Hospital Universitario La Princesa (IIS La Princesa), C/Diego de León 62 (1ª planta), Madrid, Spain.
| | - Eulàlia Montell
- Pre-Clinical R&D Department, Bioibérica, S.A., Barcelona, Spain.
| | - Josep Vergés
- Pre-Clinical R&D Department, Bioibérica, S.A., Barcelona, Spain.
| | - Manuela G López
- Instituto-Fundación Teófilo Hernando, C/ Faraday 7, Parque Científico del Campus de Cantoblanco, Universidad Autónoma de Madrid, Madrid, Spain; Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Avda. Arzobispo Morcillo 4, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Universitario La Princesa (IIS La Princesa), C/Diego de León 62 (1ª planta), Madrid, Spain.
| | - Enrique J Cobos
- Departamento de Farmacología e Instituto de Neurociencias, Facultad de Medicina, Universidad de Granada e Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain.
| | - Antonio G García
- Instituto-Fundación Teófilo Hernando, C/ Faraday 7, Parque Científico del Campus de Cantoblanco, Universidad Autónoma de Madrid, Madrid, Spain; Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Avda. Arzobispo Morcillo 4, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Universitario La Princesa (IIS La Princesa), C/Diego de León 62 (1ª planta), Madrid, Spain.
| |
Collapse
|
33
|
Costa-Pereira JT, Oliveira R, Guadilla I, Guillén MJ, Tavares I, López-Larrubia P. Neuroimaging uncovers neuronal and metabolic changes in pain modulatory brain areas in a rat model of chemotherapy-induced neuropathy - MEMRI and ex vivo spectroscopy studies. Brain Res Bull 2023; 192:12-20. [PMID: 36328144 DOI: 10.1016/j.brainresbull.2022.10.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/20/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
Chemotherapy-induced neuropathy (CIN) is one of the most common complications of cancer treatment with sensory dysfunctions which frequently include pain. The mechanisms underlying pain during CIN are starting to be uncovered. Neuroimaging allows the identification of brain circuitry involved in pain processing and modulation and has recently been used to unravel the disruptions of that circuitry by neuropathic pain. The present study evaluates the effects of paclitaxel, a cytostatic drug frequently used in cancer treatment, at the neuronal function in the anterior cingulate cortex (ACC), hypothalamus and periaqueductal gray (PAG) using manganese-enhanced magnetic resonance imaging (MEMRI). We also studied the metabolic profile at the prefrontal cortex (PFC) and hypothalamus using ex vivo spectroscopy. Wistar male rats were intraperitoneal injected with paclitaxel or vehicle solution (DMSO). The evaluation of mechanical sensitivity using von Frey test at baseline (BL), 21 (T21), 28 (T28), 49 (T49) and 56 days (T56) after CIN induction showed that paclitaxel-injected rats presented mechanical hypersensitivity from T21 until T56 after CIN induction. The evaluation of the locomotor activity and exploratory behaviors using open-field test at T28 and T56 after the first injection of paclitaxel revealed that paclitaxel-injected rats walked higher distance with higher velocity at late point of CIN accompanied with a sustained exhibition of anxiety-like behaviors. Imaging studies performed using MEMRI at T28 and T56 showed that paclitaxel treatment increased the neuronal activation in the hypothalamus and PAG at T56 in comparison with the control group. The analysis of data from ex vivo spectroscopy demonstrated that at T28 paclitaxel-injected rats presented an increase of N-acetyl aspartate (NAA) levels in the PFC and an increase of NAA and decrease of lactate (Lac) concentration in the hypothalamus compared to the control group. Furthermore, at T56 the paclitaxel-injected rats presented lower NAA and higher taurine (Tau) levels in the PFC. Together, MEMRI and metabolomic data indicate that CIN is associated with neuroplastic changes in brain areas involved in pain modulation and suggests that other events involving glial cells may be happening.
Collapse
Affiliation(s)
- José Tiago Costa-Pereira
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; IBMC-Institute of Molecular and Cell Biology, University of Porto, Portugal; I3S, Institute of Investigation and Innovation in Health, University of Porto, Portugal; Faculty of Nutrition and Food Sciences, University of Porto, Portugal
| | - Rita Oliveira
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; IBMC-Institute of Molecular and Cell Biology, University of Porto, Portugal; I3S, Institute of Investigation and Innovation in Health, University of Porto, Portugal
| | - Irene Guadilla
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Maria Jose Guillén
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Isaura Tavares
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; IBMC-Institute of Molecular and Cell Biology, University of Porto, Portugal; I3S, Institute of Investigation and Innovation in Health, University of Porto, Portugal
| | - Pilar López-Larrubia
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain.
| |
Collapse
|
34
|
Liu XG. Normalization of Neuroinflammation: A New Strategy for Treatment of Persistent Pain and Memory/Emotional Deficits in Chronic Pain. J Inflamm Res 2022; 15:5201-5233. [PMID: 36110505 PMCID: PMC9469940 DOI: 10.2147/jir.s379093] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 08/18/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic pain, which affects around 1/3 of the world population and is often comorbid with memory deficit and mood depression, is a leading source of suffering and disability. Studies in past decades have shown that hyperexcitability of primary sensory neurons resulting from abnormal expression of ion channels and central sensitization mediated pathological synaptic plasticity, such as long-term potentiation in spinal dorsal horn, underlie the persistent pain. The memory/emotional deficits are associated with impaired synaptic connectivity in hippocampus. Dysregulation of numerous endogenous proteins including receptors and intracellular signaling molecules is involved in the pathological processes. However, increasing knowledge contributes little to clinical treatment. Emerging evidence has demonstrated that the neuroinflammation, characterized by overproduction of pro-inflammatory cytokines and glial activation, is reliably detected in humans and animals with chronic pain, and is sufficient to induce persistent pain and memory/emotional deficits. The abnormal expression of ion channels and pathological synaptic plasticity in spinal dorsal horn and in hippocampus are resulting from neuroinflammation. The neuroinflammation is initiated and maintained by the interactions of circulating monocytes, glial cells and neurons. Obviously, unlike infectious diseases and cancer, which are caused by pathogens or malignant cells, chronic pain is resulting from alterations of cells and molecules which have numerous physiological functions. Therefore, normalization (counterbalance) but not simple inhibition of the neuroinflammation is the right strategy for treating neuronal disorders. Currently, no such agent is available in clinic. While experimental studies have demonstrated that intracellular Mg2+ deficiency is a common feature of chronic pain in animal models and supplement Mg2+ are capable of normalizing the neuroinflammation, activation of upregulated proteins that promote recovery, such as translocator protein (18k Da) or liver X receptors, has a similar effect. In this article, relevant experimental and clinical evidence is reviewed and discussed.
Collapse
Affiliation(s)
- Xian-Guo Liu
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
35
|
Singh J, Thapliyal S, Kumar A, Paul P, Kumar N, Bisht M, Naithani M, Rao S, Handu SS. Dimethyl Fumarate Ameliorates Paclitaxel-Induced Neuropathic Pain in Rats. Cureus 2022; 14:e28818. [PMID: 36225395 PMCID: PMC9536397 DOI: 10.7759/cureus.28818] [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] [Accepted: 09/06/2022] [Indexed: 11/30/2022] Open
Abstract
Background Paclitaxel (PTX)-induced peripheral neuropathy (PIPN) is nonresponsive to the currently available analgesics. Previous studies have shown the role of oxidative stress and central sensitization in the development of peripheral neuropathy. Dimethyl fumarate (DMF) acts as a nuclear factor erythroid-2-related factor 2 (Nrf2) activator with neuroprotective benefits and is approved for use in multiple sclerosis. Materials and methods In the current research, we evaluated the efficacy of DMF on paclitaxel-induced peripheral neuropathy in rats. Every alternate day for one week, paclitaxel 2 mg/kg dose was injected to establish a rat model of PIPN. Animals were treated with 25 mg/kg and 50 mg/kg of DMF. All the animals were assessed for thermal hyperalgesia, cold allodynia, and mechanical allodynia once a week. The gene expression of Nrf2 and the levels of pro-inflammatory mediators (interleukin (IL)-6, tumor necrosis factor-alpha (TNF-α), and IL-1β) were quantified in the sciatic nerves of these rats. The levels of p38 mitogen-activated protein kinase (MAPK) and brain-derived neurotrophic factor (BDNF) were quantified in the dorsal horn of the spinal cord. Results DMF significantly attenuated paclitaxel-induced thermal hyperalgesia and cold/mechanical allodynia. A significant decrease in the levels of pro-inflammatory cytokines with the levels of p38 MAPK and BDNF was observed in the DMF-treated animals. DMF treatment significantly upregulated the gene expression of Nrf2 in the sciatic nerve. Conclusion These findings suggest that DMF prevented the development of PIPN in rats through the activation of Nrf2 and the inhibition of p38 MAPK and BDNF.
Collapse
|
36
|
Kulkarni NP, Vaidya B, Narula AS, Sharma SS. Caffeic Acid Phenethyl Ester (CAPE) Attenuates Paclitaxel-induced Peripheral Neuropathy: A Mechanistic Study. Curr Neurovasc Res 2022; 19:293-302. [PMID: 36043777 DOI: 10.2174/1567202619666220829104851] [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: 06/12/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Chemotherapy-induced peripheral neuropathy is a debilitating pain syndrome produced as a side effect of antineoplastic drugs like paclitaxel. Despite efforts, the currently available therapeutics suffer from serious drawbacks like unwanted side effects and poor efficacy and provide only symptomatic relief. Hence, there is a need to find new therapeutic alternatives for the treatment of chemotherapy-induced peripheral neuropathy. OBJECTIVE The objective of this study was to explore the protective potential of caffeic acid phenethyl ester in paclitaxel-induced neuropathic pain. METHODS We examined the effects of caffeic acid phenethyl ester by administering paclitaxel (2 mg/kg, intraperitoneal) to female Sprague Dawley rats on four alternate days to induce neuropathic pain, followed by the administration of caffeic acid phenethyl ester (10 and 30 mg/kg, intraperitoneally). RESULTS Rats that were administered paclitaxel showed a substantially diminished pain threshold and nerve functions after 28 days. A significantly increased protein expression of Wnt signalling protein (β-catenin), inflammatory marker (matrix metalloproteinase 2) and a decrease in endogenous antioxidant (nuclear factor erythroid 2-related factor 2) levels were found in paclitaxel administered rats in comparison to the naïve control group. Caffeic acid phenethyl ester (10 and 30 mg/kg, intraperitoneal) showed improvements in behavioural and nerve function parameters along with reduced expression of β-catenin, matrix metalloproteinase 2 and an increase in nuclear factor erythroid 2- related factor 2 protein expression. CONCLUSION The present study suggests that caffeic acid phenethyl ester attenuates chemotherapyinduced peripheral neuropathy via inhibition of β-catenin and matrix metalloproteinase 2 and increases nuclear factor erythroid 2-related factor 2 activation.
Collapse
Affiliation(s)
- Namrata Pramod Kulkarni
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Punjab 160062, India
| | - Bhupesh Vaidya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Punjab 160062, India
| | - Acharan S Narula
- Narula Research Llc, 107 Boulder Bluff, Chapel Hill, North Carolina, NC 27516, USA
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Punjab 160062, India
| |
Collapse
|
37
|
7-Chloro-4-(Phenylselanyl) Quinoline Is a Novel Multitarget Therapy to Combat Peripheral Neuropathy and Comorbidities Induced by Paclitaxel in Mice. Mol Neurobiol 2022; 59:6567-6589. [DOI: 10.1007/s12035-022-02991-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/07/2022] [Indexed: 10/15/2022]
|
38
|
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
|
39
|
Nasser AH, Gendy AM, El-Yamany MF, El-Tanbouly DM. Upregulation of neuronal progranulin mediates the antinociceptive effect of trimetazidine in paclitaxel-induced peripheral neuropathy: Role of ERK1/2 signaling. Toxicol Appl Pharmacol 2022; 448:116096. [PMID: 35662665 DOI: 10.1016/j.taap.2022.116096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/23/2022] [Accepted: 05/28/2022] [Indexed: 10/18/2022]
Abstract
Neuronal progranulin (PGRN) overexpression is an endogenous adaptive pain defense following nerve injury. It allows the survival of injured neurons to block enhanced nociceptive responses. Trimetazidine (TMZ) is widely used by cardiac patients as an anti-anginal drug, reflecting its anti-ischemic property. TMZ promotes axonal regeneration of sciatic nerves after crush injury. This study explored the interplay between PGRN and extracellular signal-regulated kinases (ERK1/2) to address mechanisms underlying neuropathic pain alleviation following paclitaxel (PTX) administration. Rats were given four injections of PTX (2 mg/kg, i.p.) every other day. Two days after the last dose, rats received TMZ (25 mg/kg) with or without the ERK inhibitor, PD98059, daily for 21 days. TMZ preserved the integrity of myelinated nerve fibers, as evidenced by an obvious reduction in axonal damage biomarkers. Accordingly, it alleviated PTX-evoked thermal, cold, and mechanical hyperalgesia/allodynia. TMZ also promoted ERK1/2 phosphorylation with a profound upsurge in PGRN content. These effects were associated with a substantial increase in Notch1 receptor gene expression and a prominent anti-inflammatory effect with a marked increase in mRNA expression of secretory leukocyte protease inhibitor. Further, TMZ decreased oxidative stress and caspase-3 activity in the sciatic nerve. Conversely, co-administration of PD98059 completely abolished these beneficial effects. Thus, the robust antinociceptive effect of TMZ is largely attributed to upregulating PGRN and Notch1 receptors via ERK1/2 activation.
Collapse
Affiliation(s)
- Asmaa H Nasser
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, Giza, Egypt
| | - Abdallah M Gendy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, Giza, Egypt
| | - Mohammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Dalia M El-Tanbouly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
40
|
Brandolini L, d'Angelo M, Novelli R, Castelli V, Giorgio C, Sirico A, Cocchiaro P, D'Egidio F, Benedetti E, Cristiano C, Bugatti A, Ruocco A, Amendola PG, Talarico C, Manelfi C, Iaconis D, Beccari A, Quadros AU, Cunha TM, Caruso A, Russo R, Cimini A, Aramini A, Allegretti M. Paclitaxel binds and activates C5aR1: A new potential therapeutic target for the prevention of chemotherapy-induced peripheral neuropathy and hypersensitivity reactions. Cell Death Dis 2022; 13:500. [PMID: 35614037 PMCID: PMC9130998 DOI: 10.1038/s41419-022-04964-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 05/10/2022] [Accepted: 05/17/2022] [Indexed: 12/14/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) and hypersensitivity reactions (HSRs) are among the most frequent and impairing side effects of the antineoplastic agent paclitaxel. Here, we demonstrated that paclitaxel can bind and activate complement component 5a receptor 1 (C5aR1) and that this binding is crucial in the etiology of paclitaxel-induced CIPN and anaphylaxis. Starting from our previous data demonstrating the role of interleukin (IL)-8 in paclitaxel-induced neuronal toxicity, we searched for proteins that activate IL-8 expression and, by using the Exscalate platform for molecular docking simulations, we predicted the high affinity of C5aR1 with paclitaxel. By in vitro studies, we confirmed the specific and competitive nature of the C5aR1-paclitaxel binding and found that it triggers intracellularly the NFkB/P38 pathway and c-Fos. In F11 neuronal cells and rat dorsal root ganglia, C5aR1 inhibition protected from paclitaxel-induced neuropathological effects, while in paclitaxel-treated mice, the absence (knock-out mice) or the inhibition of C5aR1 significantly ameliorated CIPN symptoms-in terms of cold and mechanical allodynia-and reduced the chronic pathological state in the paw. Finally, we found that C5aR1 inhibition can counteract paclitaxel-induced anaphylactic cytokine release in macrophages in vitro, as well as the onset of HSRs in mice. Altogether these data identified C5aR1 as a key mediator and a new potential pharmacological target for the prevention and treatment of CIPN and HSRs induced by paclitaxel.
Collapse
Affiliation(s)
- Laura Brandolini
- Dompé Farmaceutici SpA, Via Campo di Pile, 67100, L'Aquila, Italy
| | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Rubina Novelli
- Dompé Farmaceutici SpA, Via S. Lucia, 20122, Milan, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Cristina Giorgio
- Dompé Farmaceutici SpA, Via Tommaso De Amicis, 80131, Naples, Italy
| | - Anna Sirico
- Dompé Farmaceutici SpA, Via Tommaso De Amicis, 80131, Naples, Italy
| | | | - Francesco D'Egidio
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Claudia Cristiano
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Antonella Bugatti
- Department of Molecular and Traslational Medicine, University of Brescia Medical School, 25123, Brescia, Italy
| | - Anna Ruocco
- Dompé Farmaceutici SpA, Via Tommaso De Amicis, 80131, Naples, Italy
| | | | - Carmine Talarico
- Dompé Farmaceutici SpA, Via Tommaso De Amicis, 80131, Naples, Italy
| | - Candida Manelfi
- Dompé Farmaceutici SpA, Via Tommaso De Amicis, 80131, Naples, Italy
| | - Daniela Iaconis
- Dompé Farmaceutici SpA, Via Tommaso De Amicis, 80131, Naples, Italy
| | - Andrea Beccari
- Dompé Farmaceutici SpA, Via Tommaso De Amicis, 80131, Naples, Italy
| | - Andreza U Quadros
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Thiago M Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Arnaldo Caruso
- Department of Molecular and Traslational Medicine, University of Brescia Medical School, 25123, Brescia, Italy
| | - Roberto Russo
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA, 19122, USA
| | - Andrea Aramini
- Dompé Farmaceutici SpA, Via Campo di Pile, 67100, L'Aquila, Italy
| | | |
Collapse
|
41
|
Systemic, Intrathecal, and Intracerebroventricular Antihyperalgesic Effects of the Calcium Channel Blocker CTK 01512–2 Toxin in Persistent Pain Models. Mol Neurobiol 2022; 59:4436-4452. [DOI: 10.1007/s12035-022-02864-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 04/28/2022] [Indexed: 11/25/2022]
|
42
|
Park SE, Neupane C, Noh C, Sharma R, Shin HJ, Pham TL, Lee GS, Park KD, Lee CJ, Kang DW, Lee SY, Kim HW, Park JB. Antiallodynic effects of KDS2010, a novel MAO-B inhibitor, via ROS-GABA inhibitory transmission in a paclitaxel-induced tactile hypersensitivity model. Mol Brain 2022; 15:41. [PMID: 35526002 PMCID: PMC9078011 DOI: 10.1186/s13041-022-00924-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/18/2022] [Indexed: 11/10/2022] Open
Abstract
Monoamine oxidase (MAO) inhibitors have been investigated for the treatment of neuropathic pain. Here, we assessed the antiallodynic effects of a novel MAO-B inhibitor, KDS2010, on paclitaxel (PTX)-induced mechanical hypersensitivity. Oral administration of KDS2010 effectively relieved PTX-induced mechanical hypersensitivity in a dose-dependent manner. KDS2010 (25 mg/Kg) significantly prevented and suppressed PTX-induced pain responses with minimal effects on the body weight, motor activity, and working memory. KDS2010 significantly reduced reactive astrocytosis and reactive oxygen species (ROS) level in the L4–L6 spinal cord of PTX-treated mice. Furthermore, KDS2010 reversed the attenuation of GABAergic spontaneous inhibitory postsynaptic current (sIPSC) frequency in spinal dorsal horn neurons, although it failed to restore the reduced tonic GABAA inhibition nor the increased GABA transporter 1 (GAT1) expression in PTX-treated mice. In addition, bath application of a reactive oxygen species (ROS) scavenger (PBN) restored the sIPSC frequency in PTX-treated mice but not in control and PTX + KDS2010-treated mice. These results indicated that the antiallodynic effect of KDS2010 is not due to a MAO-B-dependent GABA production. Finally, PBN alone also exerted a similar analgesic effect as KDS2010, but a co-treatment of PBN with KDS2010 showed no additive effect, suggesting that inhibition of MAO-B-dependent ROS production is responsible for the analgesic effect by KDS2010 on PTX-induced allodynia. Overall, KDS2010 attenuated PTX-induced pain behaviors by restoring the altered ROS level and GABAergic inhibitory signaling in the spinal cord, suggesting that KDS2010 is a promising therapeutic strategy for chemotherapy-induced peripheral neuropathy.
Collapse
Affiliation(s)
- Su Eun Park
- Department of Medical Sciences, Graduate School, Chungnam National University, Daejeon, 35015, Korea.,Department of Physiology, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, 35015, Korea
| | - Chiranjivi Neupane
- Department of Medical Sciences, Graduate School, Chungnam National University, Daejeon, 35015, Korea.,Department of Physiology, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, 35015, Korea.,Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Korea
| | - Chan Noh
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, 35015, South Korea
| | - Ramesh Sharma
- Department of Medical Sciences, Graduate School, Chungnam National University, Daejeon, 35015, Korea.,Department of Physiology, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, 35015, Korea.,Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Korea
| | - Hyun Jin Shin
- Department of Medical Sciences, Graduate School, Chungnam National University, Daejeon, 35015, Korea.,Department of Physiology, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, 35015, Korea
| | - Thuy Linh Pham
- Department of Medical Sciences, Graduate School, Chungnam National University, Daejeon, 35015, Korea.,Department of Physiology, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, 35015, Korea
| | - Gyu-Seung Lee
- Department of Medical Sciences, Graduate School, Chungnam National University, Daejeon, 35015, Korea.,Dong-Gu Health Promotion Center 301-01, 30 Bogeunso Avenue, Samseung-Dong, Dong-gu, Daejeon, South Korea
| | - Ki Duk Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Korea
| | - Dong-Wook Kang
- Department of Medical Sciences, Graduate School, Chungnam National University, Daejeon, 35015, Korea.,Department of Physiology, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, 35015, Korea
| | - So Yeong Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Korea
| | - Hyun-Woo Kim
- Department of Medical Sciences, Graduate School, Chungnam National University, Daejeon, 35015, Korea.,Department of Physiology, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, 35015, Korea
| | - Jin Bong Park
- Department of Medical Sciences, Graduate School, Chungnam National University, Daejeon, 35015, Korea. .,Department of Physiology, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, 35015, Korea. .,Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
43
|
Amaya C, Smith ER, Xu XX. Low Intensity Ultrasound as an Antidote to Taxane/Paclitaxel-induced Cytotoxicity. J Cancer 2022; 13:2362-2373. [PMID: 35517405 PMCID: PMC9066212 DOI: 10.7150/jca.71263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/04/2022] [Indexed: 11/25/2022] Open
Abstract
The taxane family of compounds, including Taxol/paclitaxel and Taxotere/docetaxel, are surprisingly successful drugs used in combination or alone for the treatment of most major solid tumors, especially metastatic cancer. The drugs are commonly used in regimen with other agents (often platinum drugs) as frontline treatment, or used as a single agent in a dose dense regimen for recurrent cancer. The major side effects of taxanes are peripheral neuropathy, alopecia, and neutropenia, which are grave burden for patients and limit the full potential of the taxane drugs. Especially in the current treatment protocol for peripheral neuropathy, taxane dosage is reduced once the symptoms present, resulting in the loss of full or optimal cancer killing activity. Substantial efforts have been made to address the problem of cytotoxic side effects of taxanes, though strategies remain very limited. Following administration of the taxane compound by infusion, taxane binds to cellular microtubules and is sequestered within the cells for several days. Taxane stabilizes and interferes with microtubule function, leading to ultimate death of cancer cells, but also damages hair follicles, peripheral neurons, and hemopoietic stem cells. Currently, cryo-treatment is practiced to limit exposure and side effects of the drug during infusion, though the effectiveness is uncertain or limited. A recent laboratory finding may provide a new strategy to counter taxane cytotoxicity, that a brief exposure to low density ultrasound waves was sufficient to eliminate paclitaxel cytotoxicity cells in culture by transiently breaking microtubule filaments, which were then relocated to lysosomes for disposal. Thus, ultrasonic force to break rigid microtubules is an effective solution to counter taxane cytotoxicity. The discovery and concept of low intensity ultrasound as an antidote may have the potential to provide a practical strategy to counter paclitaxel-induced peripheral neuropathy and alopecia that resulted from chemotherapy. Taxanes are a class of important drugs used in chemotherapy to treat several major cancers. This article reviews a new laboratory discovery that ultrasound can be used as an antidote for the peripheral cytotoxicity of taxane drugs and discusses the potential development and application of low intensity ultrasound to prevent side effects in chemotherapeutic treatment of cancer patients.
Collapse
Affiliation(s)
- Celina Amaya
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Elizabeth R Smith
- Department of Obstetrics, Gynecology and Reproductive Science, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Xiang-Xi Xu
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136.,Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, 1120 NW 14th Street, Miami, FL, USA
| |
Collapse
|
44
|
Bouchenaki H, Bernard A, Bessaguet F, Frachet S, Richard L, Sturtz F, Magy L, Bourthoumieu S, Demiot C, Danigo A. Neuroprotective Effect of Ramipril Is Mediated by AT2 in a Mouse MODEL of Paclitaxel-Induced Peripheral Neuropathy. Pharmaceutics 2022; 14:pharmaceutics14040848. [PMID: 35456682 PMCID: PMC9030366 DOI: 10.3390/pharmaceutics14040848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/06/2022] [Accepted: 04/11/2022] [Indexed: 02/06/2023] Open
Abstract
Paclitaxel (PTX)-induced peripheral neuropathy (PIPN) induces numerous symptoms affecting patient quality of life, leading to decreased doses or even to cessation of anticancer therapy. Previous studies have reported that a widely used drug, ramipril, improves neuroprotection in several rodent models of peripheral neuropathy. The protective role of the angiotensin II type 2 receptor (AT2) in the central and peripheral nervous systems is well-established. Here, we evaluate the effects of ramipril in the prevention of PIPN and the involvement of AT2 in this effect. Paclitaxel was administered in wild type or AT2-deficient mice on alternate days for 8 days, at a cumulative dose of 8 mg/kg (2 mg/kg per injection). Ramipril, PD123319 (an AT2 antagonist), or a combination of both were administered one day before PTX administration, and daily for the next twenty days. PTX-administered mice developed mechanical allodynia and showed a loss of sensory nerve fibers. Ramipril prevented the functional and morphological alterations in PTX mice. The preventive effect of ramipril against tactile allodynia was completely absent in AT2-deficient mice and was counteracted by PD123319 administration in wild type mice. Our work highlights the potential of ramipril as a novel preventive treatment for PIPN, and points to the involvement of AT2 in the neuroprotective role of ramipril in PIPN.
Collapse
Affiliation(s)
- Hichem Bouchenaki
- UR 20218-NeurIT, Faculties of Medicine and Pharmacy, University of Limoges, 87025 Limoges, France; (H.B.); (A.B.); (S.F.); (L.R.); (F.S.); (L.M.); (S.B.); (A.D.)
| | - Amandine Bernard
- UR 20218-NeurIT, Faculties of Medicine and Pharmacy, University of Limoges, 87025 Limoges, France; (H.B.); (A.B.); (S.F.); (L.R.); (F.S.); (L.M.); (S.B.); (A.D.)
| | - Flavien Bessaguet
- INSERM 1083 CNRS UMR 6015 Mitovasc Laboratory, CarMe Team, University of Angers, 49045 Angers, France;
| | - Simon Frachet
- UR 20218-NeurIT, Faculties of Medicine and Pharmacy, University of Limoges, 87025 Limoges, France; (H.B.); (A.B.); (S.F.); (L.R.); (F.S.); (L.M.); (S.B.); (A.D.)
- Department of Neurology, Reference Center for Rare Peripheral Neuropathies, University Hospital of Limoges, 87000 Limoges, France
| | - Laurence Richard
- UR 20218-NeurIT, Faculties of Medicine and Pharmacy, University of Limoges, 87025 Limoges, France; (H.B.); (A.B.); (S.F.); (L.R.); (F.S.); (L.M.); (S.B.); (A.D.)
- Department of Neurology, Reference Center for Rare Peripheral Neuropathies, University Hospital of Limoges, 87000 Limoges, France
| | - Franck Sturtz
- UR 20218-NeurIT, Faculties of Medicine and Pharmacy, University of Limoges, 87025 Limoges, France; (H.B.); (A.B.); (S.F.); (L.R.); (F.S.); (L.M.); (S.B.); (A.D.)
- Department of Biochemistry and Molecular Genetics, University Hospital of Limoges, 87000 Limoges, France
| | - Laurent Magy
- UR 20218-NeurIT, Faculties of Medicine and Pharmacy, University of Limoges, 87025 Limoges, France; (H.B.); (A.B.); (S.F.); (L.R.); (F.S.); (L.M.); (S.B.); (A.D.)
- Department of Neurology, Reference Center for Rare Peripheral Neuropathies, University Hospital of Limoges, 87000 Limoges, France
| | - Sylvie Bourthoumieu
- UR 20218-NeurIT, Faculties of Medicine and Pharmacy, University of Limoges, 87025 Limoges, France; (H.B.); (A.B.); (S.F.); (L.R.); (F.S.); (L.M.); (S.B.); (A.D.)
- Department of Cytogenetic, Medical Genetic and Reproduction Biology, University Hospital of Limoges, 87000 Limoges, France
| | - Claire Demiot
- UR 20218-NeurIT, Faculties of Medicine and Pharmacy, University of Limoges, 87025 Limoges, France; (H.B.); (A.B.); (S.F.); (L.R.); (F.S.); (L.M.); (S.B.); (A.D.)
- Correspondence: ; Tel.: +33-5554-35915
| | - Aurore Danigo
- UR 20218-NeurIT, Faculties of Medicine and Pharmacy, University of Limoges, 87025 Limoges, France; (H.B.); (A.B.); (S.F.); (L.R.); (F.S.); (L.M.); (S.B.); (A.D.)
| |
Collapse
|
45
|
Kim Y, Jung YH, Park SB, Kim H, Kwon JY, Kim HK, Lee HJ, Jeon S, Kim E. TMI-1, TNF-α-Converting Enzyme Inhibitor, Protects Against Paclitaxel-Induced Neurotoxicity in the DRG Neuronal Cells In Vitro. Front Pharmacol 2022; 13:842779. [PMID: 35250589 PMCID: PMC8889072 DOI: 10.3389/fphar.2022.842779] [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: 12/24/2021] [Accepted: 01/28/2022] [Indexed: 12/19/2022] Open
Abstract
Background: Chemotherapy-induced peripheral neuropathy (CIPN) negatively impacts cancer survivors' quality of life and is challenging to treat with existing drugs for neuropathic pain. TNF-α is known to potentiate TRPV1 activity, which contributes to CIPN. Here, we assessed the role of TMI-1, a TNF-α-converting enzyme inhibitor, in paclitaxel (PAC)-induced neurotoxicity in dorsal root ganglion (DRG) cells. Materials and Methods: Immortalized DRG neuronal 50B11 cells were cultured and treated with PAC or PAC with TMI-1 following neuronal differentiation. Cell viability, analysis of neurite growth, immunofluorescence, calcium flow cytometry, western blotting, quantitative RT-PCR, and cytokine quantitation by ELISA were performed to determine the role of TMI-1 in neurotoxicity in neuronal cells. Results: PAC administration decreased the length of neurites and upregulated the expression of TRPV1 in 50B11 cells. TMI-1 administration showed a protective effect by suppressing inflammatory signaling, and secretion of TNF-α. Conclusion: TMI-1 partially protects against paclitaxel-induced neurotoxicity by reversing the upregulation of TRPV1 and decreasing levels of inflammatory cytokines, including TNF-α, IL-1β, and IL-6 in neuronal cells.
Collapse
Affiliation(s)
- Yesul Kim
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Busan, South Korea
| | - Young-Hoon Jung
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Busan, South Korea
| | - Seung-Bin Park
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, South Korea
| | - Heekee Kim
- Department of Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Jae-young Kwon
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Busan, South Korea
| | - Hae-kyu Kim
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Busan, South Korea
| | - Hyeon-Jeong Lee
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Busan, South Korea
| | - Soeun Jeon
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Busan, South Korea
| | - Eunsoo Kim
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Busan, South Korea
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, South Korea
| |
Collapse
|
46
|
Silva NR, Gomes FIF, Lopes AHP, Cortez IL, Dos Santos JC, Silva CEA, Mechoulam R, Gomes FV, Cunha TM, Guimarães FS. The Cannabidiol Analog PECS-101 Prevents Chemotherapy-Induced Neuropathic Pain via PPARγ Receptors. Neurotherapeutics 2022; 19:434-449. [PMID: 34904193 PMCID: PMC9130439 DOI: 10.1007/s13311-021-01164-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2021] [Indexed: 01/03/2023] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is the main dose-limiting adverse effect of chemotherapy drugs such as paclitaxel (PTX). PTX causes marked molecular and cellular damage, mainly in the peripheral nervous system, including sensory neurons in the dorsal root ganglia (DRG). Several studies have shown the therapeutic potential of cannabinoids, including cannabidiol (CBD), the major non-psychotomimetic compound found in the Cannabis plant, to treat peripheral neuropathies. Here, we investigated the efficacy of PECS-101 (former HUF-101), a CBD fluorinated analog, on PTX-induced neuropathic pain in mice. PECS-101, administered after the end of treatment with PTX, did not reverse mechanical allodynia. However, PECS-101 (1 mg/kg) administered along with PTX treatment caused a long-lasting relief of the mechanical and cold allodynia. These effects were blocked by a PPARγ, but not CB1 and CB2 receptor antagonists. Notably, the effects of PECS-101 on the relief of PTX-induced mechanical and cold allodynia were not found in macrophage-specific PPARγ-deficient mice. PECS-101 also decreased PTX-induced increase in Tnf, Il6, and Aif1 (Iba-1) gene expression in the DRGs and the loss of intra-epidermal nerve fibers. PECS-101 did not alter motor coordination, produce tolerance, or show abuse potential. In addition, PECS-101 did not interfere with the chemotherapeutic effects of PTX. Thus, PECS-101, a new fluorinated CBD analog, could represent a novel therapeutic alternative to prevent mechanical and cold allodynia induced by PTX potentially through the activation of PPARγ in macrophages.
Collapse
Affiliation(s)
- Nicole Rodrigues Silva
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| | | | | | - Isadora Lopes Cortez
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | - Conceição Elidianne Aníbal Silva
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Raphael Mechoulam
- Department of Medicinal Chemistry and Natural Products, Hebrew University Medical Faculty, Jerusalem, Israel
| | - Felipe Villela Gomes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| | - Francisco Silveira Guimarães
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
- National Institute of Science and Translational Medicine, Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| |
Collapse
|
47
|
Methods and protocols for chemotherapy-induced peripheral neuropathy (CIPN) mouse models using paclitaxel. Methods Cell Biol 2022; 168:277-298. [DOI: 10.1016/bs.mcb.2021.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
48
|
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
|
49
|
The Protective Effect of Bergamot Polyphenolic Fraction (BPF) on Chemotherapy-Induced Neuropathic Pain. Pharmaceuticals (Basel) 2021; 14:ph14100975. [PMID: 34681199 PMCID: PMC8540578 DOI: 10.3390/ph14100975] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/13/2021] [Accepted: 09/23/2021] [Indexed: 12/14/2022] Open
Abstract
Paclitaxel is a chemotherapeutic drug used for cancer treatment. Chemotherapy-induced peripheral neuropathy (CIPN) is a common major dose-limiting side effect of many chemotherapeutic agents, including paclitaxel. CIPN is accompanied by mechanical and thermal hypersensitivity that resolves within weeks, months, or years after drug termination. To date, there is no available preventive strategy or effective treatment for CIPN due to the fact that its etiology has not been fully explained. It is clear that free radicals are implicated in many neurodegenerative diseases and recent studies have shown the important role of oxidative stress in development of CIPN. Here, we observed how, in rats, the administration of a natural antioxidant such as the bergamot polyphenolic extract (BPF), can play a crucial role in reducing CIPN. Paclitaxel administration induced mechanical allodynia and thermal hyperalgesia, which began to manifest on day seven, and reached its lowest levels on day fifteen. Paclitaxel-induced neuropathic pain was associated with nitration of proteins in the spinal cord including MnSOD, glutamine synthetase, and glutamate transporter GLT-1. This study showed that the use of BPF, probably by inhibiting the nitration of crucial proteins involved in oxidative stress, improved paclitaxel-induced pain behaviors relieving mechanical allodynia, thermal hyperalgesia, thus preventing the development of chemotherapy-induced neuropathic pain.
Collapse
|
50
|
Fotio Y, Sasso O, Ciccocioppo R, Piomelli D. Antinociceptive Profile of ARN19702, (2-Ethylsulfonylphenyl)-[(2S)-4-(6-fluoro-1,3-benzothiazol-2-yl)-2-methylpiperazin-1-yl]methanone, a Novel Orally Active N-Acylethanolamine Acid Amidase Inhibitor, in Animal Models. J Pharmacol Exp Ther 2021; 378:70-76. [PMID: 33986036 DOI: 10.1124/jpet.121.000674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 05/11/2021] [Indexed: 12/31/2022] Open
Abstract
N-Acylethanolamine acid amidase (NAAA) is an N-terminal cysteine hydrolase that stops the physiologic actions of palmitoylethanolamide, an endogenous lipid messenger that activates the transcription factor, peroxisome proliferator-activated receptor-α We have previously reported that the compound ARN19702 [(2-ethylsulfonylphenyl)-[(2S)-4-(6-fluoro-1,3-benzothiazol-2-yl)-2-methylpiperazin-1-yl]methanone] is an orally active, reversible NAAA inhibitor (IC50 on human NAAA = 230 nM) that produces remarkable protective effects against multiple sclerosis in mice. In the present study, we assessed the profile of ARN19702 in mouse and rat models of acute and neuropathic pain. Oral administration in male mice attenuated in a dose-dependent manner the spontaneous nocifensive response elicited by intraplantar formalin injection and the hypersensitivity caused by intraplantar carrageenan injection, paw incision, or sciatic nerve ligation. In male rats, ARN19702 reduced nociception associated with paclitaxel-induced neuropathy without development of subacute antinociceptive tolerance. Finally, ARN19702 (30 mg/kg, oral) did not produce place preference or alter exploratory motor behavior in male mice. The findings support the conclusion that NAAA is a suitable molecular target for the discovery of efficacious analgesic drugs devoid of rewarding potential. SIGNIFICANCE STATEMENT: This study evaluated the pharmacological profile of the orally bioavailable N-acylethanolamine acid amidase (NAAA) inhibitor (2-ethylsulfonylphenyl)-[(2S)-4-(6-fluoro-1,3-benzothiazol-2-yl)-2-methylpiperazin-1-yl]methanone (ARN19702) in mouse and rat models of neurogenic and inflammatory pain. The compound's potential rewarding and sedative effects were also examined. It is concluded that ARN19702 exhibits a broad analgesic profile that can be generalized across rodent species. The findings point to NAAA as a control node in the processing of neuropathic and inflammatory pain and to ARN19702 as a lead to uncover novel pain therapeutics devoid of addictive potential .
Collapse
Affiliation(s)
- Yannick Fotio
- Departments of Anatomy and Neurobiology (Y.F., D.P.), Biological Chemistry (D.P.), and Pharmaceutical Sciences (D.P.), University of California, Irvine, California; Drug Discovery and Development, Istituto Italiano di Technologia, Genova, Italy (O.S.); and School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy (R.C.)
| | - Oscar Sasso
- Departments of Anatomy and Neurobiology (Y.F., D.P.), Biological Chemistry (D.P.), and Pharmaceutical Sciences (D.P.), University of California, Irvine, California; Drug Discovery and Development, Istituto Italiano di Technologia, Genova, Italy (O.S.); and School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy (R.C.)
| | - Roberto Ciccocioppo
- Departments of Anatomy and Neurobiology (Y.F., D.P.), Biological Chemistry (D.P.), and Pharmaceutical Sciences (D.P.), University of California, Irvine, California; Drug Discovery and Development, Istituto Italiano di Technologia, Genova, Italy (O.S.); and School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy (R.C.)
| | - Daniele Piomelli
- Departments of Anatomy and Neurobiology (Y.F., D.P.), Biological Chemistry (D.P.), and Pharmaceutical Sciences (D.P.), University of California, Irvine, California; Drug Discovery and Development, Istituto Italiano di Technologia, Genova, Italy (O.S.); and School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy (R.C.)
| |
Collapse
|