1
|
Shatunova S, Aktar R, Peiris M, Lee JYP, Vetter I, Starobova H. The role of the gut microbiome in neuroinflammation and chemotherapy-induced peripheral neuropathy. Eur J Pharmacol 2024; 979:176818. [PMID: 39029779 DOI: 10.1016/j.ejphar.2024.176818] [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: 03/04/2024] [Revised: 06/05/2024] [Accepted: 07/17/2024] [Indexed: 07/21/2024]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is one of the most debilitating adverse effects caused by chemotherapy drugs such as paclitaxel, oxaliplatin and vincristine. It is untreatable and often leads to the discontinuation of cancer therapy and a decrease in the quality of life of cancer patients. It is well-established that neuroinflammation and the activation of immune and glial cells are among the major drivers of CIPN. However, these processes are still poorly understood, and while many chemotherapy drugs alone can drive the activation of these cells and consequent neuroinflammation, it remains elusive to what extent the gut microbiome influences these processes. In this review, we focus on the peripheral mechanisms driving CIPN, and we address the bidirectional pathways by which the gut microbiome communicates with the immune and nervous systems. Additionally, we critically evaluate literature addressing how chemotherapy-induced dysbiosis and the consequent imbalance in bacterial products may contribute to the activation of immune and glial cells, both of which drive neuroinflammation and possibly CIPN development, and how we could use this knowledge for the development of effective treatment strategies.
Collapse
Affiliation(s)
- Svetlana Shatunova
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Rubina Aktar
- Centre for Neuroscience, Surgery and Trauma, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Madusha Peiris
- Centre for Neuroscience, Surgery and Trauma, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Jia Yu Peppermint Lee
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia; The School of Pharmacy, The University of Queensland, Woollsiana, QLD, Australia
| | - Hana Starobova
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia.
| |
Collapse
|
2
|
Pan C, Xu Y, Jiang Z, Fan C, Chi Z, Zhang Y, Miao M, Ren Y, Wu Z, Xu L, Mei C, Chen Q, Xi Y, Chen X. Naringenin relieves paclitaxel-induced pain by suppressing calcitonin gene-related peptide signalling and enhances the anti-tumour action of paclitaxel. Br J Pharmacol 2024; 181:3136-3159. [PMID: 38715438 DOI: 10.1111/bph.16397] [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: 06/16/2023] [Revised: 02/01/2024] [Accepted: 03/21/2024] [Indexed: 08/03/2024] Open
Abstract
BACKGROUND AND PURPOSE Chemotherapy-induced peripheral neuropathy (CIPN) commonly causes neuropathic pain, but its pathogenesis remains unclear, and effective therapies are lacking. Naringenin, a natural dihydroflavonoid compound, has anti-inflammatory, anti-nociceptive and anti-tumour activities. However, the effects of naringenin on chemotherapy-induced pain and chemotherapy effectiveness remain unexplored. EXPERIMENTAL APPROACH Female and male mouse models of chemotherapy-induced pain were established using paclitaxel. Effects of naringenin were assessed on pain induced by paclitaxel or calcitonin gene-related peptide (CGRP) and on CGRP expression in dorsal root ganglia (DRG) and spinal cord tissue. Additionally, we examined peripheral macrophage infiltration, glial activation, c-fos expression, DRG neuron excitability, microglial M1/M2 polarization, and phosphorylation of spinal NF-κB. Furthermore, we investigated the synergic effect and related mechanisms of naringenin and paclitaxel on cell survival of cancer cells in vitro. KEY RESULTS Systemic administration of naringenin attenuated paclitaxel-induced pain in both sexes. Naringenin reduced paclitaxel-enhanced CGRP expression in DRGs and the spinal cord, and alleviated CGRP-induced pain in naïve mice of both sexes. Naringenin mitigated macrophage infiltration and reversed paclitaxel-elevated c-fos expression and DRG neuron excitability. Naringenin decreased spinal glial activation and NF-κB phosphorylation in both sexes but influenced microglial M1/M2 polarization only in females. Co-administration of naringenin with paclitaxel enhanced paclitaxel's anti-tumour effect, impeded by an apoptosis inhibitor. CONCLUSION AND IMPLICATIONS Naringenin's anti-nociceptive mechanism involves CGRP signalling and neuroimmunoregulation. Furthermore, naringenin facilitates paclitaxel's anti-tumour action, possibly involving apoptosis. This study demonstrates naringenin's potential as a supplementary treatment in cancer therapy by mitigating side effects and potentiating efficacy of chemotherapy.
Collapse
Affiliation(s)
- Chen Pan
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Yuhao Xu
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Zongsheng Jiang
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Chengjiang Fan
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Zhexi Chi
- Department of Anesthesiology, Ningbo No. 2 Hospital, Ningbo, China
| | - Yu Zhang
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Mengmeng Miao
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Yuxuan Ren
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Ziyi Wu
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Linbin Xu
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Changqing Mei
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Qingge Chen
- Department of Anesthesiology, Ningbo No. 2 Hospital, Ningbo, China
- Department of Anesthesiology, The People's Hospital of Bozhou, Bozhou, China
| | - Yang Xi
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Xiaowei Chen
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| |
Collapse
|
3
|
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
|
4
|
Saito K, Koizumi S. A promising drug for neuropathic pain: identification of vesicular nucleotide transporter as a novel target of eicosapentaenoic acid. Purinergic Signal 2023; 19:587-589. [PMID: 36627401 PMCID: PMC10754788 DOI: 10.1007/s11302-022-09918-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 12/26/2022] [Indexed: 01/12/2023] Open
Affiliation(s)
- Kozo Saito
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
- Yamanashi GLIA Center, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan.
- Yamanashi GLIA Center, University of Yamanashi, Chuo, Yamanashi, Japan.
| |
Collapse
|
5
|
Sun C, Deng J, Ma Y, Meng F, Cui X, Li M, Li J, Li J, Yin P, Kong L, Zhang L, Tang P. The dual role of microglia in neuropathic pain after spinal cord injury: Detrimental and protective effects. Exp Neurol 2023; 370:114570. [PMID: 37852469 DOI: 10.1016/j.expneurol.2023.114570] [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: 07/04/2023] [Revised: 09/21/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023]
Abstract
Spinal cord injury (SCI) is a debilitating condition that is frequently accompanied by neuropathic pain, resulting in significant physical and psychological harm to a vast number of individuals globally. Despite the high prevalence of neuropathic pain following SCI, the precise underlying mechanism remains incompletely understood. Microglia are a type of innate immune cell that are present in the central nervous system (CNS). They have been observed to have a significant impact on neuropathic pain following SCI. This article presents a comprehensive overview of recent advances in understanding the role of microglia in the development of neuropathic pain following SCI. Specifically, the article delves into the detrimental and protective effects of microglia on neuropathic pain following SCI, as well as the mechanisms underlying their interconversion. Furthermore, the article provides a thorough overview of potential avenues for future research in this area.
Collapse
Affiliation(s)
- Chang Sun
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China; Department of Orthopedics, Air Force Medical Center, PLA, Beijing, China
| | - Junhao Deng
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China; School of Life Sciences, Tsinghua University, Beijing, China; State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instruments, Tsinghua University, Beijing, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Yifei Ma
- School of Medicine, Nankai University, Tianjin, China
| | - Fanqi Meng
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiang Cui
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Ming Li
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Jiantao Li
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Jia Li
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Pengbin Yin
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Lingjie Kong
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instruments, Tsinghua University, Beijing, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
| | - Licheng Zhang
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China.
| | - Peifu Tang
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China.
| |
Collapse
|
6
|
Wu S, Xiong T, Guo S, Zhu C, He J, Wang S. An up-to-date view of paclitaxel-induced peripheral neuropathy. J Cancer Res Ther 2023; 19:1501-1508. [PMID: 38156915 DOI: 10.4103/jcrt.jcrt_1982_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 07/12/2023] [Indexed: 01/03/2024]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN),referring to the damage to the peripheral nerves caused by exposure to a neurotoxic chemotherapeutic agent, is a common side effect amongst patients undergoing chemotherapy. Paclitaxel-induced peripheral neuropathy (PIPN) can lead to dose reduction or early cessation of chemotherapy, which is not conducive to patients'survival. Even after treatment is discontinued, PIPN symptoms carried a greater risk of worsening and plagued the patient's life, leading to long-term morbidity in survivors. Here, we summarize the research progress for clinical manifestations, risk factors, pathogenesis, prevention and treatment of PIPN, so as to embark on the path of preventing PIPN with prolongation of patient's life quality on a long-term basis.
Collapse
Affiliation(s)
- Shan Wu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Pharmacy, People's Hospital of Leshan, Shizhong, Leshan, China
| | - Tu Xiong
- Department of Radiology, People's Hospital of Leshan, Shizhong, Leshan, China
| | - Shenglan Guo
- Department of Pharmacy, Huadu District People's Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Cuiyi Zhu
- Department of Pharmacy, People's Hospital of Leshan, Shizhong, Leshan, China
| | - Jing He
- Department of Pharmacy, People's Hospital of Leshan, Shizhong, Leshan, China
| | - Shurong Wang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
7
|
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: 0] [Impact Index Per Article: 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
|
8
|
Mei C, Pan C, Xu L, Miao M, Lu Q, Yu Y, Lin P, Wu W, Ni F, Gao Y, Xu Y, Xu J, Chen X. Trimethoxyflavanone relieves Paclitaxel-induced neuropathic pain via inhibiting expression and activation of P2X7 and production of CGRP in mice. Neuropharmacology 2023; 236:109584. [PMID: 37225085 DOI: 10.1016/j.neuropharm.2023.109584] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 04/28/2023] [Accepted: 05/07/2023] [Indexed: 05/26/2023]
Abstract
Paclitaxel (PTX) is an anticancer drug used to treat solid tumors, but one of its common adverse effects is chemotherapy-induced peripheral neuropathy (CIPN). Currently, there is limited understanding of neuropathic pain associated with CIPN and effective treatment strategies are inadequate. Previous studies report the analgesic actions of Naringenin, a dihydroflavonoid compound, in pain. Here we observed that the anti-nociceptive action of a Naringenin derivative, Trimethoxyflavanone (Y3), was superior to Naringenin in PTX-induced pain (PIP). An intrathecal injection of Y3 (1 μg) reversed the mechanical and thermal thresholds of PIP and suppressed the PTX-induced hyper-excitability of dorsal root ganglion (DRG) neurons. PTX enhanced the expression of ionotropic purinergic receptor P2X7 (P2X7) in satellite glial cells (SGCs) and neurons in DRGs. The molecular docking simulation predicts possible interactions between Y3 and P2X7. Y3 reduced the PTX-enhanced P2X7 expression in DRGs. Electrophysiological recordings revealed that Y3 directly inhibited P2X7-mediated currents in DRG neurons of PTX-treated mice, suggesting that Y3 suppressed both expression and function of P2X7 in DRGs post-PTX administration. Y3 also reduced the production of calcitonin gene-related peptide (CGRP) in DRGs and at the spinal dorsal horn. Additionally, Y3 suppressed the PTX-enhanced infiltration of Iba1-positive macrophage-like cells in DRGs and overactivation of spinal astrocytes and microglia. Therefore, our results indicate that Y3 attenuates PIP via inhibiting P2X7 function, CGRP production, DRG neuron sensitization, and abnormal spinal glial activation. Our study implies that Y3 could be a promising drug candidate against CIPN-associated pain and neurotoxicity.
Collapse
Affiliation(s)
- Changqing Mei
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Chen Pan
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Linbin Xu
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Mengmeng Miao
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Qichen Lu
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yang Yu
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Pengyu Lin
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Wenwei Wu
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Feng Ni
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, China; LeadArt Technologies Ltd., Ningbo, 315201, China
| | - Yinping Gao
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yuhao Xu
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Jia Xu
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Xiaowei Chen
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
9
|
Bagher AM, Binmahfouz LS, Shaik RA, Eid BG. Cannabinoid receptor 1 positive allosteric modulator (GAT229) attenuates cisplatin-induced neuropathic pain in mice. Saudi Pharm J 2023; 31:255-264. [PMID: 36942271 PMCID: PMC10023546 DOI: 10.1016/j.jsps.2022.12.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is one of chemotherapies' most often documented side effects. Patients with CIPN experience spontaneous burning, numbness, tingling, and neuropathic pain in their feet and hands. Currently, there is no effective pharmacological treatment to prevent or treat CIPN. Activating the cannabinoid receptor type 1 (CB1) by orthosteric agonists has shown promising results in alleviating the pain and neuroinflammation associated with CIPN. However, the use of CB1 orthosteric agonists is linked to undesirable side effects. Unlike the CB1 orthosteric agonists, CB1 positive allosteric modulators (PAMs) don't produce any psychoactive effects, tolerance, or dependence. Previous studies have shown that CB1 PAMs exhibit antinociceptive effects in inflammatory and neuropathic rodent models. This study aimed to investigate the potential benefits of the newly synthesized GAT229, a pure CB1 PAM, in alleviating neuropathic pain and slowing the progression of CIPN. GAT229 was evaluated in a cisplatin-induced (CIS) mouse model of peripheral neuropathic pain (3 mg/kg/d, 28 d, i.p.). GAT229 attenuated and slowed the progression of thermal hyperalgesia and mechanical allodynia induced by CIS, as evaluated by the hotplate test and von Frey filament test. GAT229 reduced the expression of proinflammatory cytokines in the dorsal root ganglia (DRG) neurons. Furthermore, GAT229 attenuated nerve injuries by normalizing the brain-derived neurotrophic factor and the nerve growth factor mRNA expression levels in the DRG neurons. The CB1 receptor antagonist/inverse agonist AM251 blocked GAT229-mediated beneficial effects. According to our data, we suggest that CB1 PAMs might be beneficial in alleviating neuropathic pain and slowing the progression of CIPN.
Collapse
Affiliation(s)
- Amina M. Bagher
- Corresponding author at: Department of Pharmacology and Toxicology, King Abdulaziz University, Jeddah, Saudi Arabia.
| | | | | | | |
Collapse
|
10
|
Microglial Cannabinoid CB 2 Receptors in Pain Modulation. Int J Mol Sci 2023; 24:ijms24032348. [PMID: 36768668 PMCID: PMC9917135 DOI: 10.3390/ijms24032348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023] Open
Abstract
Pain, especially chronic pain, can strongly affect patients' quality of life. Cannabinoids ponhave been reported to produce potent analgesic effects in different preclinical pain models, where they primarily function as agonists of Gi/o protein-coupled cannabinoid CB1 and CB2 receptors. The CB1 receptors are abundantly expressed in both the peripheral and central nervous systems. The central activation of CB1 receptors is strongly associated with psychotropic adverse effects, thus largely limiting its therapeutic potential. However, the CB2 receptors are promising targets for pain treatment without psychotropic adverse effects, as they are primarily expressed in immune cells. Additionally, as the resident immune cells in the central nervous system, microglia are increasingly recognized as critical players in chronic pain. Accumulating evidence has demonstrated that the expression of CB2 receptors is significantly increased in activated microglia in the spinal cord, which exerts protective consequences within the surrounding neural circuitry by regulating the activity and function of microglia. In this review, we focused on recent advances in understanding the role of microglial CB2 receptors in spinal nociceptive circuitry, highlighting the mechanism of CB2 receptors in modulating microglia function and its implications for CB2 receptor- selective agonist-mediated analgesia.
Collapse
|
11
|
Tay N, Laakso EL, Schweitzer D, Endersby R, Vetter I, Starobova H. Chemotherapy-induced peripheral neuropathy in children and adolescent cancer patients. Front Mol Biosci 2022; 9:1015746. [PMID: 36310587 PMCID: PMC9614173 DOI: 10.3389/fmolb.2022.1015746] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/20/2022] [Indexed: 11/22/2022] Open
Abstract
Brain cancer and leukemia are the most common cancers diagnosed in the pediatric population and are often treated with lifesaving chemotherapy. However, chemotherapy causes severe adverse effects and chemotherapy-induced peripheral neuropathy (CIPN) is a major dose-limiting and debilitating side effect. CIPN can greatly impair quality of life and increases morbidity of pediatric patients with cancer, with the accompanying symptoms frequently remaining underdiagnosed. Little is known about the incidence of CIPN, its impact on the pediatric population, and the underlying pathophysiological mechanisms, as most existing information stems from studies in animal models or adult cancer patients. Herein, we aim to provide an understanding of CIPN in the pediatric population and focus on the 6 main substance groups that frequently cause CIPN, namely the vinca alkaloids (vincristine), platinum-based antineoplastics (cisplatin, carboplatin and oxaliplatin), taxanes (paclitaxel and docetaxel), epothilones (ixabepilone), proteasome inhibitors (bortezomib) and immunomodulatory drugs (thalidomide). We discuss the clinical manifestations, assessments and diagnostic tools, as well as risk factors, pathophysiological processes and current pharmacological and non-pharmacological approaches for the prevention and treatment of CIPN.
Collapse
Affiliation(s)
- Nicolette Tay
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - E-Liisa Laakso
- Mater Research Institute-The University of Queensland, South Brisbane, QLD, Australia
| | - Daniel Schweitzer
- Mater Research Institute-The University of Queensland, South Brisbane, QLD, Australia
| | - Raelene Endersby
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
- The School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Hana Starobova
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
- *Correspondence: Hana Starobova,
| |
Collapse
|
12
|
Samandar F, Tehranizadeh ZA, Saberi MR, Chamani J. CB1 as a novel target for Ginkgo biloba's terpene trilactone for controlling chemotherapy-induced peripheral neuropathy (CIPN). J Mol Model 2022; 28:283. [PMID: 36044079 DOI: 10.1007/s00894-022-05284-8] [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: 01/28/2022] [Accepted: 08/19/2022] [Indexed: 11/24/2022]
Abstract
The application of antineoplastic chemotherapeutic agents causes a common side effect known as chemotherapy-induced peripheral neuropathy (CIPN) that leads to reducing the quality of patient's life. This research involves the performance of molecular docking and molecular dynamic (MD) simulation studies to explore the impact of terpenoids of Ginkgo biloba on the targets (CB-1, TLR4, FAAH-1, COX-1, COX-2) that can significantly affect the controlling of CIPN's symptoms. According to the in-vitro and in-vivo investigations, terpenoids, particularly ginkgolides B, A, and bilobalide, can cause significant effects on neuropathic pain. The molecular docking results disclosed the tendency of our ligands to interact with mainly CB1 and FAAH-1, as well as partly with TLR4, throughout their interactions with targets. Terpene trilactone can exhibit a lower rate of binding energy than CB1's inhibitor (7dy), while being precisely located in the CB1's active site and capable of inducing stable interactions by forming hydrogen bonds. The analyses of MD simulation proved that ginkgolide B was a more suitable activator and inhibitor for CB1 and TLR4, respectively, when compared to bilobalide and ginkgolide A. Moreover, bilobalide is capable of inhibiting FAAH-1 more effectively than the two other ligands. According to the analyses of ADME, every three ligands followed the Lipinski's rule of five. Considering these facts, the exertion of three ligands is recommended for their anti-inflammatory, neuroprotective, and anti-nociception influences caused by primarily activating CB1 and inhibiting FAAH-1 and TLR4; in this regard, these compounds can stand as potential candidates for the control and treatment of CIPN's symptoms.
Collapse
Affiliation(s)
- Farzaneh Samandar
- Department of Biology, Faculty of Sciences, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Zeinab Amiri Tehranizadeh
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Reza Saberi
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Bioinformatics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jamshidkhan Chamani
- Department of Biology, Faculty of Sciences, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| |
Collapse
|
13
|
Practical Considerations for the Use of Cannabis in Cancer Pain Management—What a Medical Oncologist Should Know. J Clin Med 2022; 11:jcm11175036. [PMID: 36078963 PMCID: PMC9457511 DOI: 10.3390/jcm11175036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 12/29/2022] Open
Abstract
Pain is a highly debilitating emotional and sensory experience that significantly affects quality of life (QoL). Numerous chronic conditions, including cancer, are associated with chronic pain. In the setting of malignancy, pain can be a consequence of the tumor itself or of life-saving interventions, including surgery, chemotherapy, and radiotherapy. Despite significant pharmacological advances and awareness campaigns, pain remains undertreated in one-third of patients. To date, opioids have been the mainstay of cancer pain management. The problematic side effects and unsatisfactory pain relief of opioids have revived patients’ and physicians’ interest in finding new solutions, including cannabis and cannabinoids. The medical use of cannabis has been prohibited for decades, and it remains in Schedule 1 of the Misuse of Drugs Regulations. Currently, the legal context for its usage has become more permissive. Various preclinical and observational studies have aimed to prove that cannabinoids could be effective in cancer pain management. However, their clinical utility must be further supported by high-quality clinical trials.
Collapse
|
14
|
Formulated Curcumin Prevents Paclitaxel-Induced Peripheral Neuropathy through Reduction in Neuroinflammation by Modulation of α7 Nicotinic Acetylcholine Receptors. Pharmaceutics 2022; 14:pharmaceutics14061296. [PMID: 35745868 PMCID: PMC9227889 DOI: 10.3390/pharmaceutics14061296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 01/04/2023] Open
Abstract
Paclitaxel is widely used in the treatment of various types of solid malignancies. Paclitaxel-induced peripheral neuropathy (PIPN) is often characterized by burning pain, cold, and mechanical allodynia in patients. Currently, specific pharmacological treatments against PIPN are lacking. Curcumin, a polyphenol of Curcuma longa, shows antioxidant, anti-inflammatory, and neuroprotective effects and has recently shown efficacy in the mitigation of various peripheral neuropathies. Here, we tested, for the first time, the therapeutic effect of 1.5% dietary curcumin and Meriva (a lecithin formulation of curcumin) in preventing the development of PIPN in C57BL/6J mice. Curcumin or Meriva treatment was initiated one week before injection of paclitaxel and continued throughout the study (21 days). Mechanical and cold sensitivity as well as locomotion/motivation were tested by the von Frey, acetone, and wheel-running tests, respectively. Additionally, sensory-nerve-action-potential (SNAP) amplitude by caudal-nerve electrical stimulation, electronic microscopy of the sciatic nerve, and inflammatory-protein quantification in DRG and the spinal cord were measured. Interestingly, a higher concentration of curcumin was observed in the spinal cord with the Meriva diet than the curcumin diet. Our results showed that paclitaxel-induced mechanical hypersensitivity was partially prevented by the curcumin diet but completely prevented by Meriva. Both the urcumin diet and the Meriva diet completely prevented cold hypersensitivity, the reduction in SNAP amplitude and reduced mitochondrial pathology in sciatic nerves observed in paclitaxel-treated mice. Paclitaxel-induced inflammation in the spinal cord was also prevented by the Meriva diet. In addition, an increase in α7 nAChRs mRNA, known for its anti-inflammatory effects, was also observed in the spinal cord with the Meriva diet in paclitaxel-treated mice. The use of the α7 nAChR antagonist and α7 nAChR KO mice showed, for the first time in vivo, that the anti-inflammatory effects of curcumin in peripheral neuropathy were mediated by these receptors. The results presented in this study represent an important advance in the understanding of the mechanism of action of curcumin in vivo. Taken together, our results show the therapeutic potential of curcumin in preventing the development of PIPN and further confirms the role of α7 nAChRs in the anti-inflammatory effects of curcumin.
Collapse
|
15
|
García MM, Molina-Álvarez M, Rodríguez-Rivera C, Paniagua N, Quesada E, Uranga JA, Rodríguez-Franco MI, Pascual D, Goicoechea C. Antinociceptive and modulatory effect of pathoplastic changes in spinal glia of a TLR4/CD14 blocking molecule in two models of pain in rat. Biomed Pharmacother 2022; 150:112986. [PMID: 35462333 DOI: 10.1016/j.biopha.2022.112986] [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: 02/08/2022] [Revised: 04/02/2022] [Accepted: 04/14/2022] [Indexed: 11/02/2022] Open
Abstract
The role of spinal glia in the development and maintenance of chronic pain has become over the last years a subject of increasing interest. In this regard, toll-like receptor 4 (TLR4) signaling has been proposed as a major trigger mechanism. Hence, in this study we explored the implications of TLR4 inhibition in the periphery and primarily in the CNS, focusing on the impact this inhibition renders in pain development and glia activation in the dorsal horn in two models of pain. Making use of a synthetic cluster of differentiation 14 (CD14)/TLR4 antagonist, the effect of TLR4 blockade on tactile allodynia and heat hyperalgesia was evaluated in osteoarthritic and postoperative rat models. An in vitro parallel artificial membrane permeation assay was performed to determine the proneness of the drug to permeate the blood-brain barrier prior to systemic and central administration. Findings suggest a dominant role of peripheral TLR4 in the model of incisional pain, whilst both peripheral and central TLR4 seem to be responsible for osteoarthritic pain. That is, central and peripheral TLR4 may be differently involved in the etiopathology of diverse types of pain what potentially seems a promising approach in the management of pain.
Collapse
Affiliation(s)
- Miguel M García
- Area of Pharmacology and Nutrition and Bromatology, Department of Basic Health Sciences, School of Health Sciences, Universidad Rey Juan Carlos, High Performance Experimental Pharmacology Research Group, Universidad Rey Juan Carlos (PHARMAKOM), Madrid, Spain; Unidad Asociada I+D+i Instituto de Química Médica (IQM-CSIC)-URJC, Madrid, Spain
| | - Miguel Molina-Álvarez
- Area of Pharmacology and Nutrition and Bromatology, Department of Basic Health Sciences, School of Health Sciences, Universidad Rey Juan Carlos, High Performance Experimental Pharmacology Research Group, Universidad Rey Juan Carlos (PHARMAKOM), Madrid, Spain; Unidad Asociada I+D+i Instituto de Química Médica (IQM-CSIC)-URJC, Madrid, Spain
| | - Carmen Rodríguez-Rivera
- Area of Pharmacology and Nutrition and Bromatology, Department of Basic Health Sciences, School of Health Sciences, Universidad Rey Juan Carlos, High Performance Experimental Pharmacology Research Group, Universidad Rey Juan Carlos (PHARMAKOM), Madrid, Spain; Unidad Asociada I+D+i Instituto de Química Médica (IQM-CSIC)-URJC, Madrid, Spain
| | - Nancy Paniagua
- Area of Pharmacology and Nutrition and Bromatology, Department of Basic Health Sciences, School of Health Sciences, Universidad Rey Juan Carlos, High Performance Experimental Pharmacology Research Group, Universidad Rey Juan Carlos (PHARMAKOM), Madrid, Spain; Unidad Asociada I+D+i Instituto de Química Médica (IQM-CSIC)-URJC, Madrid, Spain
| | - Ernesto Quesada
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain; Unidad Asociada I+D+i Instituto de Química Médica (IQM-CSIC)-URJC, Madrid, Spain
| | - José Antonio Uranga
- Area of Histology, Department of Basic Health Sciences, School of Health Sciences, Universidad Rey Juan Carlos, High Performance Research Group in Physiopathology and Pharmacology of the Digestive System, Universidad Rey Juan Carlos (NEUGUT), Madrid, Spain
| | | | - David Pascual
- Area of Pharmacology and Nutrition and Bromatology, Department of Basic Health Sciences, School of Health Sciences, Universidad Rey Juan Carlos, High Performance Experimental Pharmacology Research Group, Universidad Rey Juan Carlos (PHARMAKOM), Madrid, Spain; Unidad Asociada I+D+i Instituto de Química Médica (IQM-CSIC)-URJC, Madrid, Spain.
| | - Carlos Goicoechea
- Area of Pharmacology and Nutrition and Bromatology, Department of Basic Health Sciences, School of Health Sciences, Universidad Rey Juan Carlos, High Performance Experimental Pharmacology Research Group, Universidad Rey Juan Carlos (PHARMAKOM), Madrid, Spain; Unidad Asociada I+D+i Instituto de Química Médica (IQM-CSIC)-URJC, Madrid, Spain
| |
Collapse
|
16
|
Bagues A, López-Tofiño Y, Llorente-Berzal Á, Abalo R. Cannabinoid drugs against chemotherapy-induced adverse effects: focus on nausea/vomiting, peripheral neuropathy and chemofog in animal models. Behav Pharmacol 2022; 33:105-129. [PMID: 35045012 DOI: 10.1097/fbp.0000000000000667] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Although new drugs are being developed for cancer treatment, classical chemotherapeutic agents are still front-line therapies, despite their frequent association with severe side effects that can hamper their use. Cannabinoids may prevent or palliate some of these side effects. The aim of the present study is to review the basic research which has been conducted evaluating the effects of cannabinoid drugs in the treatment of three important side effects induced by classical chemotherapeutic agents: nausea and vomiting, neuropathic pain and cognitive impairment. Several published studies have demonstrated that cannabinoids are useful in preventing and reducing the nausea, vomits and neuropathy induced by different chemotherapy regimens, though other side effects can occur, such as a reduction of gastrointestinal motility, along with psychotropic effects when using centrally-acting cannabinoids. Thus, peripherally-acting cannabinoids and new pharmacological options are being investigated, such as allosteric or biased agonists. Additionally, due to the increase in the survival of cancer patients, there are emerging data that demonstrate an important cognitive deterioration due to chemotherapy, and because the cannabinoid drugs have a neuroprotective effect, they could be useful in preventing chemotherapy-induced cognitive impairment (as demonstrated through studies in other neurological disorders), but this has not yet been tested. Thus, although cannabinoids seem a promising therapeutic approach in the treatment of different side effects induced by chemotherapeutic agents, future research will be necessary to find pharmacological options with a safer profile. Moreover, a new line of research awaits to be opened to elucidate their possible usefulness in preventing cognitive impairment.
Collapse
Affiliation(s)
- Ana Bagues
- Área de Farmacología y Nutrición, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón
- High Performance Research Group in Experimental Pharmacology (PHARMAKOM-URJC)
- Unidad Asociada I+D+i del Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Yolanda López-Tofiño
- Área de Farmacología y Nutrición, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System NeuGut-URJC
| | - Álvaro Llorente-Berzal
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland
- Centre for Pain Research and Galway Neuroscience Centre, NCBES, National University of Ireland, Galway, Ireland
| | - Raquel Abalo
- Área de Farmacología y Nutrición, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón
- Unidad Asociada I+D+i del Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System NeuGut-URJC
- Grupo de Trabajo de Ciencias Básicas en Dolor y Analgesia de la Sociedad Española del Dolor, Madrid, Spain
| |
Collapse
|
17
|
van den Hoogen NJ, Harding EK, Davidson CED, Trang T. Cannabinoids in Chronic Pain: Therapeutic Potential Through Microglia Modulation. Front Neural Circuits 2022; 15:816747. [PMID: 35069129 PMCID: PMC8777271 DOI: 10.3389/fncir.2021.816747] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/17/2021] [Indexed: 12/16/2022] Open
Abstract
Chronic pain is a complex sensory, cognitive, and emotional experience that imposes a great personal, psychological, and socioeconomic burden on patients. An estimated 1.5 billion people worldwide are afflicted with chronic pain, which is often difficult to treat and may be resistant to the potent pain-relieving effects of opioid analgesics. Attention has therefore focused on advancing new pain therapies directed at the cannabinoid system because of its key role in pain modulation. Endocannabinoids and exogenous cannabinoids exert their actions primarily through Gi/o-protein coupled cannabinoid CB1 and CB2 receptors expressed throughout the nervous system. CB1 receptors are found at key nodes along the pain pathway and their activity gates both the sensory and affective components of pain. CB2 receptors are typically expressed at low levels on microglia, astrocytes, and peripheral immune cells. In chronic pain states, there is a marked increase in CB2 expression which modulates the activity of these central and peripheral immune cells with important consequences for the surrounding pain circuitry. Growing evidence indicate that interventions targeting CB1 or CB2 receptors improve pain outcomes in a variety of preclinical pain models. In this mini-review, we will highlight recent advances in understanding how cannabinoids modulate microglia function and its implications for cannabinoid-mediated analgesia, focusing on microglia-neuron interactions within the spinal nociceptive circuitry.
Collapse
Affiliation(s)
- Nynke J. van den Hoogen
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Erika K. Harding
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Chloé E. D. Davidson
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Tuan Trang
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- *Correspondence: Tuan Trang
| |
Collapse
|
18
|
Semis HS, Kandemir FM, Kaynar O, Dogan T, Arikan SM. The protective effects of hesperidin against paclitaxel-induced peripheral neuropathy in rats. Life Sci 2021; 287:120104. [PMID: 34743946 DOI: 10.1016/j.lfs.2021.120104] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/21/2021] [Accepted: 10/24/2021] [Indexed: 01/15/2023]
Abstract
Paclitaxel (PTX), which is widely used in the treatment of solid tumors, leads to dose limitation because it causes peripheral neuropathy. This study was conducted to evaluate the potential effects of hesperidin (HES), which has various biological and pharmacological properties, against PTX-induced sciatic nerve damage. For this purpose, Sprague Dawley rats were given PTX 2 mg/kg/b.w for 5 days, then 100 or 200 mg/kg/b.w HES for 10 days, and behavioral tests were conducted at the end of the experiment. The data obtained show that PTX-induced MDA, NF-κB, IL-1β, TNF-α, COX-2, nNOS, JAK2, STAT3, and GFAP levels decreased with HES administration. Moreover, it was observed that SOD, CAT, and GPx activities inhibited by PTX increased with HES administration. It was determined that PTX caused apoptosis in the sciatic nerve by increasing Caspase-3 and Bax levels and suppressing Bcl-2 levels. HES, on the other hand, showed an anti-apoptotic effect, increasing Bcl-2 levels and decreasing Caspase-3 and Bax levels. Also, it was observed that PTX could cause endoplasmic reticulum stress (ERS) by increasing PERK, IRE1, ATF-6, GRP78 and CHOP mRNA transcript levels, while HES could alleviate ERS by suppressing them. The results indicate that neuropathic pain associated with PTX-induced peripheral neuropathy can be alleviated by HES administration and that it is a promising compound for cancer patients. In addition, it is thought that the results of the present study contain information that will shed light for researchers regarding further studies to be conducted with HES.
Collapse
Affiliation(s)
- Halil Sezgin Semis
- Department of Orthopedics and Traumatology, Private Buhara Hospital, Erzurum, Turkey
| | - Fatih Mehmet Kandemir
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| | - Ozgur Kaynar
- Department of Biochemistry, Faculty of Veterinary Medicine, Kastamonu University, Kastamonu, Turkey
| | - Tuba Dogan
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Sefik Murat Arikan
- Department of Orthopedics and Traumatology, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
19
|
Chen N, Ge MM, Li DY, Wang XM, Liu DQ, Ye DW, Tian YK, Zhou YQ, Chen JP. β2-adrenoreceptor agonist ameliorates mechanical allodynia in paclitaxel-induced neuropathic pain via induction of mitochondrial biogenesis. Biomed Pharmacother 2021; 144:112331. [PMID: 34673421 DOI: 10.1016/j.biopha.2021.112331] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/07/2021] [Accepted: 10/10/2021] [Indexed: 12/30/2022] Open
Abstract
Chemotherapy-induced neuropathic pain is a debilitating and common side effect of cancer treatment and so far no effective drug is available for treatment of the serious side effect. Previous studies have demonstrated β2-adrenoreceptor (ADRB2) agonists can attenuate neuropathic pain. However, the role of ADRB2 in paclitaxel -induced neuropathic pain (PINP) remains unclear. In this study, we investigated the effect of formoterol, a long-acting ADRB2 agonist, and related mechanisms in PINP. A rat model of PINP was established by intraperitoneal injection of paclitaxel (2 mg/kg) every other day with a final cumulative dose of 8 mg/kg. Hind paw withdrawal thresholds (PWTs) in response to von Frey filament stimuli were used to evaluate mechanical allodynia. Western blot was used to examine the expression of ADRB2, peroxisome proliferator-activated receptor coactivator-1α (PGC-1α), nuclear respiratory factors 1 (NRF1) and mitochondrial transcription factor A (TFAM) and the immunofluorescence was to detect the cellular localization of ADRB2 and PGC-1α in the spinal cord. Moreover, we measured mitochondrial DNA (mtDNA) copy number by qPCR. In our study, formoterol attenuated established PINP and delayed the onset of PINP. Formoterol restored ADRB2 expression as well as mtDNA copy number and PGC-1α, NRF1, and TFAM protein expression, which are major genes involved in mitochondrial biogenesis, in the spinal cord of PINP rats. Moreover, we found the analgesic effect of formoterol against PINP was partially abolished by PGC-1α inhibitor SR-18292. Collectively, these results demonstrated the activation of ADRB2 with formoterol ameliorates PINP at least partially through induction of mitochondrial biogenesis.
Collapse
MESH Headings
- Adrenergic beta-2 Receptor Agonists/pharmacology
- Analgesics/pharmacology
- Animals
- DNA, Mitochondrial/genetics
- DNA, Mitochondrial/metabolism
- Disease Models, Animal
- Formoterol Fumarate/pharmacology
- Male
- Mitochondria/drug effects
- Mitochondria/genetics
- Mitochondria/metabolism
- Neuralgia/drug therapy
- Neuralgia/genetics
- Neuralgia/metabolism
- Neuralgia/physiopathology
- Organelle Biogenesis
- Paclitaxel
- Pain Threshold/drug effects
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
- Rats, Sprague-Dawley
- Receptors, Adrenergic, beta-2/drug effects
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Spinal Cord/drug effects
- Spinal Cord/metabolism
- Spinal Cord/physiopathology
- Rats
Collapse
Affiliation(s)
- Nan Chen
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Meng-Meng Ge
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dan-Yang Li
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Mei Wang
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dai-Qiang Liu
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Da-Wei Ye
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu-Ke Tian
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ya-Qun Zhou
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Jian-Ping Chen
- Department of Pain Management, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
| |
Collapse
|
20
|
Campos RMP, Aguiar AFL, Paes-Colli Y, Trindade PMP, Ferreira BK, de Melo Reis RA, Sampaio LS. Cannabinoid Therapeutics in Chronic Neuropathic Pain: From Animal Research to Human Treatment. Front Physiol 2021; 12:785176. [PMID: 34916962 PMCID: PMC8669747 DOI: 10.3389/fphys.2021.785176] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/10/2021] [Indexed: 12/25/2022] Open
Abstract
Despite the importance of pain as a warning physiological system, chronic neuropathic pain is frequently caused by damage in the nervous system, followed by persistence over a long period, even in the absence of dangerous stimuli or after healing of injuries. Chronic neuropathic pain affects hundreds of millions of adults worldwide, creating a direct impact on quality of life. This pathology has been extensively characterized concerning its cellular and molecular mechanisms, and the endocannabinoid system (eCS) is widely recognized as pivotal in the development of chronic neuropathic pain. Scientific evidence has supported that phyto-, synthetic and endocannabinoids are efficient for pain management, while strong data arise from the therapeutic use of Cannabis-derived products. The use of medicinal Cannabis products is directed toward not only relieving symptoms of chronic pain, but also improving several aspects of patients’ welfare. Here, we review the involvement of eCS, along with other cellular and molecular elements, in chronic neuropathic pain pathology and how this system can be targeted for pain management.
Collapse
|
21
|
Vermeer CJC, Hiensch AE, Cleenewerk L, May AM, Eijkelkamp N. Neuro-immune interactions in paclitaxel-induced peripheral neuropathy. Acta Oncol 2021; 60:1369-1382. [PMID: 34313190 DOI: 10.1080/0284186x.2021.1954241] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Paclitaxel is a taxane-based chemotherapeutic agent used as a treatment in breast cancer. There is no effective prevention or treatment strategy for the most common side effect of peripheral neuropathy. In this manuscript, we reviewed the molecular mechanisms that contribute to paclitaxel-induced peripheral neuropathy (PIPN) with an emphasis on immune-related processes. METHODS A systematic search of the literature was conducted in PubMed, EMBASE and Cochrane Library. The SYRCLE's risk of bias tool was used to assess internal validity. RESULTS 156 studies conducted with rodent models were included. The risk of bias was high due to unclear methodology. Paclitaxel induces changes in myelinated axons, mitochondrial dysfunction, and mechanical hypersensitivity by affecting ion channels expression and function and facilitating spinal transmission. Paclitaxel-induced inflammatory responses are important contributors to PIPN. CONCLUSION Immune-related processes are an important mechanism contributing to PIPN. Studies in humans that validate these mechanistic data are highly needed to facilitate the development of therapeutic strategies.
Collapse
Affiliation(s)
- Cornelia J. C. Vermeer
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Anouk E. Hiensch
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Laurence Cleenewerk
- Center of Translational Immunology (CTI), University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Anne M. May
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Niels Eijkelkamp
- Center of Translational Immunology (CTI), University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
22
|
Pathomechanisms of Paclitaxel-Induced Peripheral Neuropathy. TOXICS 2021; 9:toxics9100229. [PMID: 34678925 PMCID: PMC8540213 DOI: 10.3390/toxics9100229] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/09/2021] [Accepted: 09/16/2021] [Indexed: 12/18/2022]
Abstract
Peripheral neuropathy is one of the most common side effects of chemotherapy, affecting up to 60% of all cancer patients receiving chemotherapy. Moreover, paclitaxel induces neuropathy in up to 97% of all gynecological and urological cancer patients. In cancer cells, paclitaxel induces cell death via microtubule stabilization interrupting cell mitosis. However, paclitaxel also affects cells of the central and peripheral nervous system. The main symptoms are pain and numbness in hands and feet due to paclitaxel accumulation in the dorsal root ganglia. This review describes in detail the pathomechanisms of paclitaxel in the peripheral nervous system. Symptoms occur due to a length-dependent axonal sensory neuropathy, where axons are symmetrically damaged and die back. Due to microtubule stabilization, axonal transport is disrupted, leading to ATP undersupply and oxidative stress. Moreover, mitochondria morphology is altered during paclitaxel treatment. A key player in pain sensation and axonal damage is the paclitaxel-induced inflammation in the spinal cord as well as the dorsal root ganglia. An increased expression of chemokines and cytokines such as IL-1β, IL-8, and TNF-α, but also CXCR4, RAGE, CXCL1, CXCL12, CX3CL1, and C3 promote glial activation and accumulation, and pain sensation. These findings are further elucidated in this review.
Collapse
|
23
|
Hore ZL, Villa-Hernandez S, Denk F. Probing the peripheral immune response in mouse models of oxaliplatin-induced peripheral neuropathy highlights their limited translatability. Wellcome Open Res 2021; 6:68. [PMID: 34250264 PMCID: PMC8243229 DOI: 10.12688/wellcomeopenres.16635.2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Chemotherapy-induced peripheral neuropathy (CIPN) is a disabling side effect of various chemotherapeutic agents, including oxaliplatin. It is highly prevalent amongst cancer patients, causing sensory abnormalities and pain. Unfortunately, as the underlying mechanisms remain poorly understood, effective therapeutics are lacking. Neuro-immune interactions have been highlighted as potential contributors to the development and maintenance of CIPN, however, whether this is the case in oxaliplatin-induced peripheral neuropathy (OIPN) is yet to be fully established. Methods: In this study we used flow cytometry to examine the peripheral immune response of male C57BL/6 mice following both single and repeated oxaliplatin administration. In animals exposed to repeated dosing, we also undertook mechanical and thermal behavioural assays to investigate how oxaliplatin alters phenotype, and conducted RT-qPCR experiments on bone marrow derived macrophages in order to further inspect the effects of oxaliplatin on immune cells. Results: In contrast to other reports, we failed to observe substantial changes in overall leukocyte, lymphocyte or myeloid cell numbers in dorsal root ganglia, sciatic nerves or inguinal lymph nodes. We did however note subtle, tissue-dependant alterations in several myeloid subpopulations following repeated dosing. These included a significant reduction in MHCII antigen presenting cells in the sciatic nerve and an increase in infiltrating cell types into the inguinal lymph nodes. Though repeated oxaliplatin administration had a systemic effect, we were unable to detect a pain-like behavioural phenotype in response to either cold or mechanical stimuli. Consequently, we cannot comment on whether the observed myeloid changes are associated with OIPN. Conclusions: Our discussion puts these results into the wider context of the field, advocating for greater transparency in reporting, alignment in experimental design and the introduction of more clinically relevant models. Only through joint concerted effort can we hope to increase our understanding of the underlying mechanisms of CIPN, including any immune contributions.
Collapse
Affiliation(s)
- Zoe Lee Hore
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| | - Sara Villa-Hernandez
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| | - Franziska Denk
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| |
Collapse
|
24
|
Hashiesh HM, Sharma C, Goyal SN, Sadek B, Jha NK, Kaabi JA, Ojha S. A focused review on CB2 receptor-selective pharmacological properties and therapeutic potential of β-caryophyllene, a dietary cannabinoid. Biomed Pharmacother 2021; 140:111639. [PMID: 34091179 DOI: 10.1016/j.biopha.2021.111639] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 02/06/2023] Open
Abstract
The endocannabinoid system (ECS), a conserved physiological system emerged as a novel pharmacological target for its significant role and potential therapeutic benefits ranging from neurological diseases to cancer. Among both, CB1 and CB2R types, CB2R have received attention for its pharmacological effects as antioxidant, anti-inflammatory, immunomodulatory and antiapoptotic that can be achieved without causing psychotropic adverse effects through CB1R. The ligands activate CB2R are of endogenous, synthetic and plant origin. In recent years, β-caryophyllene (BCP), a natural bicyclic sesquiterpene in cannabis as well as non-cannabis plants, has received attention due to its selective agonist property on CB2R. BCP has been well studied in a variety of pathological conditions mediating CB2R selective agonist property. The focus of the present manuscript is to represent the CB2R selective agonist mediated pharmacological mechanisms and therapeutic potential of BCP. The present narrative review summarizes insights into the CB2R-selective pharmacological properties and therapeutic potential of BCP such as cardioprotective, hepatoprotective, neuroprotective, nephroprotective, gastroprotective, chemopreventive, antioxidant, anti-inflammatory, and immunomodulator. The available evidences suggest that BCP, can be an important candidate of plant origin endowed with CB2R selective properties that may provide a pharmacological rationale for its pharmacotherapeutic application and pharmaceutical development like a drug. Additionally, given the wide availability in edible plants and dietary use, with safety, and no toxicity, BCP can be promoted as a nutraceutical and functional food for general health and well-being. Further, studies are needed to explore pharmacological and pharmaceutical opportunities for therapeutic and preventive applications of use of BCP in human diseases.
Collapse
Affiliation(s)
- Hebaallah Mamdouh Hashiesh
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Charu Sharma
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Sameer N Goyal
- Shri Vile Parle Kelvani Mandal's Institute of Pharmacy, Dhule 424001, Maharashtra, India
| | - Bassem Sadek
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, Uttar Pradesh 201310, India
| | - Juma Al Kaabi
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates; Zayed Bin Sultan Al Nahyan Center for Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates.
| |
Collapse
|
25
|
Uhelski ML, Li Y, Fonseca MM, Romero-Snadoval EA, Dougherty PM. Role of innate immunity in chemotherapy-induced peripheral neuropathy. Neurosci Lett 2021; 755:135941. [PMID: 33961945 DOI: 10.1016/j.neulet.2021.135941] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 01/18/2023]
Abstract
It has become increasingly clear that the innate immune system plays an essential role in the generation of many types of neuropathic pain including that which accompanies cancer treatment. In this article we review current findings of the role of the innate immune system in contributing to cancer treatment pain at the distal endings of peripheral nerve, in the nerve trunk, in the dorsal root ganglion and in the spinal dorsal horn.
Collapse
Affiliation(s)
- Megan L Uhelski
- The Department of Pain Medicine Research, The Division of Anesthesiology, Critical Care and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, United States
| | - Yan Li
- The Department of Pain Medicine Research, The Division of Anesthesiology, Critical Care and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, United States
| | - Miriam M Fonseca
- The Department of Anesthesiology, Wake Forest School of Medicine, United States
| | | | - Patrick M Dougherty
- The Department of Pain Medicine Research, The Division of Anesthesiology, Critical Care and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, United States.
| |
Collapse
|
26
|
Zhao YX, Yu XC, Gao JH, Yao MJ, Zhu B. Acupuncture for Paclitaxel-Induced Peripheral Neuropathy: A Review of Clinical and Basic Studies. J Pain Res 2021; 14:993-1005. [PMID: 33883931 PMCID: PMC8055287 DOI: 10.2147/jpr.s296150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/17/2021] [Indexed: 11/23/2022] Open
Abstract
Paclitaxel-induced peripheral neuropathy (PIPN) is a common and intractable side effect of the conventional chemotherapeutic agent paclitaxel. Acupuncture has been reported as an effective alternative therapy in treatment of PIPN in both basic studies and clinical trials. However, there is a lack of comprehensive surveys to summarize the action of acupuncture in management of PIPN. In this review, we briefly demonstrate the basic pathology of PIPN, which includes the activation of ion channels, mitochondrial dysfunction, disruption of axonal transport and also neuro-inflammatory involvement. Meanwhile, we review both the clinical and basic studies as an emphasis to give a general overview of the therapeutic effect of acupuncture against PIPN. Finally, we summarize the current known mechanisms underlying the action of acupuncture against PIPN mainly at peripheral and spinal levels, which include various neurotransmitters, multiple receptors, different types of enzymes and molecules. In conclusion, acupuncture could be considered as a potential alternative therapy in treatment of PIPN, and further clinical and experimental studies are called for in the future.
Collapse
Affiliation(s)
- Yu-Xue Zhao
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Xiao-Chun Yu
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Jun-Hong Gao
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Ming-Jiang Yao
- Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, People's Republic of China.,Key Laboratory of Pharmacology of Chinese Materia Medica, Beijing, 100091, People's Republic of China
| | - Bing Zhu
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| |
Collapse
|
27
|
Duffy SS, Hayes JP, Fiore NT, Moalem-Taylor G. The cannabinoid system and microglia in health and disease. Neuropharmacology 2021; 190:108555. [PMID: 33845074 DOI: 10.1016/j.neuropharm.2021.108555] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/13/2022]
Abstract
Recent years have yielded significant advances in our understanding of microglia, the immune cells of the central nervous system (CNS). Microglia are key players in CNS development, immune surveillance, and the maintenance of proper neuronal function throughout life. In the healthy brain, homeostatic microglia have a unique molecular signature. In neurological diseases, microglia become activated and adopt distinct transcriptomic signatures, including disease-associated microglia (DAM) implicated in neurodegenerative disorders. Homeostatic microglia synthesise the endogenous cannabinoids 2-arachidonoylglycerol and anandamide and express the cannabinoid receptors CB1 and CB2 at constitutively low levels. Upon activation, microglia significantly increase their synthesis of endocannabinoids and upregulate their expression of CB2 receptors, which promote a protective microglial phenotype by enhancing their production of neuroprotective factors and reducing their production of pro-inflammatory factors. Here, we summarise the effects of the microglial cannabinoid system in the CNS demyelinating disease multiple sclerosis, the neurodegenerative diseases Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis, chronic inflammatory and neuropathic pain, and psychiatric disorders including depression, anxiety and schizophrenia. We discuss the therapeutic potential of cannabinoids in regulating microglial activity and highlight the need to further investigate their specific microglia-dependent immunomodulatory effects.
Collapse
Affiliation(s)
- Samuel S Duffy
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, UNSW Sydney, NSW, 2052, Australia
| | - Jessica P Hayes
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, UNSW Sydney, NSW, 2052, Australia
| | - Nathan T Fiore
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, UNSW Sydney, NSW, 2052, Australia
| | - Gila Moalem-Taylor
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, UNSW Sydney, NSW, 2052, Australia.
| |
Collapse
|
28
|
Hore ZL, Villa-Hernandez S, Denk F. Probing the peripheral immune response in mouse models of oxaliplatin-induced peripheral neuropathy highlights their limited translatability. Wellcome Open Res 2021; 6:68. [PMID: 34250264 PMCID: PMC8243229 DOI: 10.12688/wellcomeopenres.16635.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2021] [Indexed: 04/03/2024] Open
Abstract
Background: Chemotherapy-induced peripheral neuropathy (CIPN) is a disabling side effect of various chemotherapeutic agents, including oxaliplatin. It is highly prevalent amongst cancer patients, causing sensory abnormalities and pain. Unfortunately, as the underlying mechanisms remain poorly understood, effective therapeutics are lacking. Neuro-immune interactions have been highlighted as potential contributors to the development and maintenance of CIPN, however, whether this is the case in oxaliplatin-induced peripheral neuropathy (OIPN) is yet to be fully established. Methods: In this study we used flow cytometry to examine the peripheral immune response of male C57BL/6 mice following both single and repeated oxaliplatin administration. In animals exposed to repeated dosing, we also undertook mechanical and thermal behavioural assays to investigate how oxaliplatin alters phenotype, and conducted RT-qPCR experiments on bone marrow derived macrophages in order to further inspect the effects of oxaliplatin on immune cells. Results: In contrast to other reports, we failed to observe substantial changes in overall leukocyte, lymphocyte or myeloid cell numbers in dorsal root ganglia, sciatic nerves or inguinal lymph nodes. We did however note subtle, tissue-dependant alterations in several myeloid subpopulations following repeated dosing. These included a significant reduction in MHCII antigen presenting cells in the sciatic nerve and an increase in infiltrating cell types into the inguinal lymph nodes. Though repeated oxaliplatin administration had a systemic effect, we were unable to detect a pain-like behavioural phenotype in response to either cold or mechanical stimuli. Consequently, we cannot comment on whether the observed myeloid changes are associated with OIPN. Conclusions: Our discussion puts these results into the wider context of the field, advocating for greater transparency in reporting, alignment in experimental design and the introduction of more clinically relevant models. Only through joint concerted effort can we hope to increase our understanding of the underlying mechanisms of CIPN, including any immune contributions.
Collapse
Affiliation(s)
- Zoe Lee Hore
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| | - Sara Villa-Hernandez
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| | - Franziska Denk
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| |
Collapse
|
29
|
Paniagua N, Sánchez-Robles EM, Bagues A, Martín-Fontelles MI, Goicoechea C, Girón R. Behavior and electrophysiology studies of the peripheral neuropathy induced by individual and co-administration of paclitaxel and oxaliplatin in rat. Life Sci 2021; 277:119397. [PMID: 33794249 DOI: 10.1016/j.lfs.2021.119397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 11/26/2022]
Abstract
AIMS Antitumor agents, as taxanes and platinum compounds, induce peripheral neuropathies which can hamper their use for cancer treatment. The study of chemotherapy-induced neuropathies in humans is difficult because of ethical reasons, differences among administration protocols and intrinsic characteristics of patients. The aim of the present study is to compare the neuropathic signs induced by individual or combined administration of paclitaxel and oxaliplatin. MAIN METHODS Oxaliplatin and paclitaxel were administered individually and combined to induce peripheral neuropathy in rats, sensory neuropathic signs were assessed in the hind limbs and orofacial area. The in vitro skin-saphenous nerve preparation was used to record the axonal activity of Aδ sensory neurons. KEY FINDINGS Animals treated with the combination developed mechanical allodynia in the paws and muscular hyperalgesia in the orofacial area, which was similar to that in animals treated with monotherapy, the latter also developed cold allodynia in the paws. Aδ-fibers of the rats treated with the combination were hyperexcited and presented hypersensitivity to pressure stimulation of the innervated skin, also similar to that recorded in the fibers of the animals treated with monotherapy. SIGNIFICANCE Our work objectively demonstrates that the combination of a platinum compound with a taxane does not worsen the development of sensorial neuropathies in rats, which is an interesting data to take into account when the combination of antitumor drugs is necessary. Co-administration of antitumor drugs is more effective in cancer treatment without increasing the risk of the disabling neuropathic side effects.
Collapse
Affiliation(s)
- N Paniagua
- Área de Farmacología, Nutrición y Bromatología, Dpto. C.C. Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Unidad Asociada CSIC-IQM, High Performance Research Group in Experimental Pharmacology (PHARMAKOM), Alcorcón, Spain
| | - E M Sánchez-Robles
- Área de Farmacología, Nutrición y Bromatología, Dpto. C.C. Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Unidad Asociada CSIC-IQM, High Performance Research Group in Experimental Pharmacology (PHARMAKOM), Alcorcón, Spain
| | - A Bagues
- Área de Farmacología, Nutrición y Bromatología, Dpto. C.C. Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Unidad Asociada CSIC-IQM, High Performance Research Group in Experimental Pharmacology (PHARMAKOM), Alcorcón, Spain.
| | - M I Martín-Fontelles
- Área de Farmacología, Nutrición y Bromatología, Dpto. C.C. Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Unidad Asociada CSIC-IQM, High Performance Research Group in Experimental Pharmacology (PHARMAKOM), Alcorcón, Spain
| | - C Goicoechea
- Área de Farmacología, Nutrición y Bromatología, Dpto. C.C. Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Unidad Asociada CSIC-IQM, High Performance Research Group in Experimental Pharmacology (PHARMAKOM), Alcorcón, Spain
| | - R Girón
- Área de Farmacología, Nutrición y Bromatología, Dpto. C.C. Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Unidad Asociada CSIC-IQM, High Performance Research Group in Experimental Pharmacology (PHARMAKOM), Alcorcón, Spain
| |
Collapse
|
30
|
Toma W, Caillaud M, Patel NH, Tran TH, Donvito G, Roberts J, Bagdas D, Jackson A, Lichtman A, Gewirtz DA, Makriyannis A, Malamas MS, Imad Damaj M. N-acylethanolamine-hydrolysing acid amidase: A new potential target to treat paclitaxel-induced neuropathy. Eur J Pain 2021; 25:1367-1380. [PMID: 33675555 DOI: 10.1002/ejp.1758] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 03/01/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Although paclitaxel is an effective chemotherapeutic agent used to treat multiple types of cancer (e.g. breast, ovarian, neck and lung), it also elicits paclitaxel-induced peripheral neuropathy (PIPN), which represents a major dose-limiting side effect of this drug. METHODS As the endogenously produced N-acylethanolamine, palmitoylethanolamide (PEA), reverses paclitaxel-induced mechanical hypersensitivity in mice, the main goals of this study were to examine if paclitaxel affects levels of endogenous PEA in the spinal cord of mice and whether exogenous administration of PEA provides protection from the occurrence of paclitaxel-induced mechanical hypersensitivity. We further examined whether inhibition of N-acylethanolamine-hydrolysing acid amidase (NAAA), a hydrolytic PEA enzyme, would offer protection in mouse model of PIPN. RESULTS Paclitaxel reduced PEA levels in the spinal cord, suggesting that dysregulation of this lipid signalling system may contribute to PIPN. Consistent with this idea, repeated administration of PEA partially prevented the paclitaxel-induced mechanical hypersensitivity. We next evaluated whether the selective NAAA inhibitor, AM9053, would prevent paclitaxel-induced mechanical hypersensitivity in mice. Acute administration of AM9053 dose-dependently reversed mechanical hypersensitivity through a PPAR-α mechanism, whereas repeated administration of AM9053 fully prevented the development of PIPN, without any evidence of tolerance. Moreover, AM9053 produced a conditioned place preference in paclitaxel-treated mice, but not in control mice. This pattern of findings suggests a lack of intrinsic rewarding effects, but a reduction in the pain aversiveness induced by paclitaxel. Finally, AM9053 did not alter paclitaxel-induced cytotoxicity in lung tumour cells. CONCLUSIONS Collectively, these studies suggest that NAAA represents a promising target to treat and prevent PIPN. SIGNIFICANCE The present study demonstrates that the chemotherapeutic paclitaxel alters PEA levels in the spinal cord, whereas repeated exogenous PEA administration moderately alleviates PIPN in mice. Additionally, targeting NAAA, PEA's hydrolysing enzyme with a selective compound AM9053 reverses and prevents the PIPN via the PPAR-α mechanism. Overall, the data suggest that selective NAAA inhibitors denote promising future therapeutics to mitigate and prevent PIPN.
Collapse
Affiliation(s)
- Wisam Toma
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Martial Caillaud
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Nipa H Patel
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Tammy H Tran
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Giulia Donvito
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Jane Roberts
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Deniz Bagdas
- Department of Psychiatry, Yale University School of Medicine, Yale Tobacco Center of Regulatory Science, New Haven, CT, USA
| | - Asti Jackson
- Department of Psychiatry, Yale University School of Medicine, Yale Tobacco Center of Regulatory Science, New Haven, CT, USA
| | - Aron Lichtman
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.,Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - David A Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Alexandros Makriyannis
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, USA
| | - Michael S Malamas
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, USA
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.,Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
31
|
CAILLAUD M, PATEL NH, WHITE A, WOOD M, Contreras KM, TOMA W, Alkhlaif Y, ROBERTS JL, Tran TH, JACKSON AB, POKLIS J, GEWIRTZ DA, DAMAJ MI. Targeting Peroxisome Proliferator-Activated Receptor-α (PPAR- α) to reduce paclitaxel-induced peripheral neuropathy. Brain Behav Immun 2021; 93:172-185. [PMID: 33434562 PMCID: PMC8226373 DOI: 10.1016/j.bbi.2021.01.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND AND PURPOSE Paclitaxel, a widely used anti-cancer drug, is frequently associated with prolonged and severe peripheral neuropathies (PIPN), associated with neuroinflammation. Currently, PIPN effective treatments are lacking. Peroxisome Proliferator-Activated Receptor-α (PPAR-⍺) can modulate inflammatory responses. Thus, the use of PPAR-⍺ agonists, such as fibrates (fenofibrate and choline-fenofibrate), currently used in dyslipidemia treatment, could represent an interesting therapeutic approach in PIPN. EXPERIMENTAL APPROACH Our studies tested the efficacy of fenofibrate (150 mg/kg, daily, i.p.) and choline fenofibrate (60 mg/kg daily, p.o.) in reversing and preventing the development of PIPN (paclitaxel: 8 mg/kg, i.p., every other day for 4 days) in male and female C57BL/6J mice. Mechanical and cold hypersensitivity, conditioned place preference, sensory nerve action potential (SNAP), as well as the expression of PPAR-⍺, TNF-⍺, IL-1β and IL-6 mRNA were evaluated. KEY RESULTS While fenofibrate treatment partially reversed and prevented the development of mechanical hypersensitivity, this was completely reversed and prevented by choline-fenofibrate. Both fibrates were able to completely reverse and prevent cold hypersensitivity induced by paclitaxel. The reduction of SNAP amplitude induced by paclitaxel was also reversed by both fenofibrate and choline-fenofibrate. Our results indicate that suppression of paclitaxel-induced hypersensitivity by fibrates involves the regulation of PPAR-⍺ expression and decrease neuroinflammation in DRG. Finally, the co-treatment of Paclitaxel and fenofibric acid (fibrates active metabolite) was tested on different cancer cell lines, no decrease in the antitumoral effect of paclitaxel was observed. CONCLUSIONS AND IMPLICATIONS Taken together, our results show for the first time the therapeutic potential (prevention and reversal) of fibrates in PIPN and opens to a potential pharmacological repurposing of these drugs.
Collapse
Affiliation(s)
- Martial CAILLAUD
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA.,Corresponding authors:,
| | - Nipa H. PATEL
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Alyssa WHITE
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Mackinsey WOOD
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Katherine M. Contreras
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Wisam TOMA
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Yasmin Alkhlaif
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Jane L. ROBERTS
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Tammy H. Tran
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Asti B. JACKSON
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - Justin POKLIS
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - David A. GEWIRTZ
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA
| | - M. Imad DAMAJ
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA.,Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, USA.,Corresponding authors:,
| |
Collapse
|
32
|
Fumagalli G, Monza L, Cavaletti G, Rigolio R, Meregalli C. Neuroinflammatory Process Involved in Different Preclinical Models of Chemotherapy-Induced Peripheral Neuropathy. Front Immunol 2021; 11:626687. [PMID: 33613570 PMCID: PMC7890072 DOI: 10.3389/fimmu.2020.626687] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Peripheral neuropathies are characterized by nerves damage and axonal loss, and they could be classified in hereditary or acquired forms. Acquired peripheral neuropathies are associated with several causes, including toxic agent exposure, among which the antineoplastic compounds are responsible for the so called Chemotherapy-Induced Peripheral Neuropathy (CIPN). Several clinical features are related to the use of anticancer drugs which exert their action by affecting different mechanisms and structures of the peripheral nervous system: the axons (axonopathy) or the dorsal root ganglia (DRG) neurons cell body (neuronopathy/ganglionopathy). In addition, antineoplastic treatments may affect the blood brain barrier integrity, leading to cognitive impairment that may be severe and long-lasting. CIPN may affect patient quality of life leading to modification or discontinuation of the anticancer therapy. Although the mechanisms of the damage are not completely understood, several hypotheses have been proposed, among which neuroinflammation is now emerging to be relevant in CIPN pathophysiology. In this review, we consider different aspects of neuro-immune interactions in several CIPN preclinical studies which suggest a critical connection between chemotherapeutic agents and neurotoxicity. The features of the neuroinflammatory processes may be different depending on the type of drug (platinum derivatives, taxanes, vinca alkaloids and proteasome inhibitors). In particular, recent studies have demonstrated an involvement of the immune response (both innate and adaptive) and the stimulation and secretion of mediators (cytokines and chemokines) that may be responsible for the painful symptoms, whereas glial cells such as satellite and Schwann cells might contribute to the maintenance of the neuroinflammatory process in DRG and axons respectively. Moreover, neuroinflammatory components have also been shown in the spinal cord with microglia and astrocytes playing an important role in CIPN development. Taking together, better understanding of these aspects would permit the development of possible strategies in order to improve the management of CIPN.
Collapse
Affiliation(s)
- Giulia Fumagalli
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, Monza, Italy
| | - Laura Monza
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, Monza, Italy
| | - Guido Cavaletti
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, Monza, Italy
| | - Roberta Rigolio
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, Monza, Italy
| | - Cristina Meregalli
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
33
|
Meregalli C, Monza L, Chiorazzi A, Scali C, Guarnieri C, Fumagalli G, Alberti P, Pozzi E, Canta A, Ballarini E, Rodriguez-Menendez V, Oggioni N, Cavaletti G, Marmiroli P. Human Intravenous Immunoglobulin Alleviates Neuropathic Symptoms in a Rat Model of Paclitaxel-Induced Peripheral Neurotoxicity. Int J Mol Sci 2021; 22:ijms22031058. [PMID: 33494384 PMCID: PMC7865319 DOI: 10.3390/ijms22031058] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 01/07/2023] Open
Abstract
The onset of chemotherapy-induced peripheral neurotoxicity (CIPN) is a leading cause of the dose reduction or discontinuation of cancer treatment due to sensory symptoms. Paclitaxel (PTX) can cause painful peripheral neuropathy, with a negative impact on cancer survivors' quality of life. While recent studies have shown that neuroinflammation is involved in PTX-induced peripheral neurotoxicity (PIPN), the pathophysiology of this disabling side effect remains largely unclear and no effective therapies are available. Therefore, here we investigated the effects of human intravenous immunoglobulin (IVIg) on a PIPN rat model. PTX-treated rats showed mechanical allodynia and neurophysiological alterations consistent with a severe sensory axonal polyneuropathy. In addition, morphological evaluation showed a reduction of intra-epidermal nerve fiber (IENF) density and evidenced axonopathy with macrophage infiltration, which was more prominent in the distal segment of caudal nerves. Three weeks after the last PTX injection, mechanical allodynia was still present in PTX-treated rats, while the full recovery in the group of animals co-treated with IVIg was observed. At the pathological level, this behavioral result was paralleled by prevention of the reduction in IENF density induced by PTX in IVIg co-treated rats. These results suggest that the immunomodulating effect of IVIg co-treatment can alleviate PIPN neurotoxic manifestations, probably through a partial reduction of neuroinflammation.
Collapse
Affiliation(s)
- Cristina Meregalli
- Experimental Neurology Unit, School of Medicine and Surgery, and NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, 20900 Monza, Italy; (C.M.); (L.M.); (A.C.); (G.F.); (P.A.); (E.P.); (A.C.); (E.B.); (V.R.-M.); (N.O.); (P.M.)
| | - Laura Monza
- Experimental Neurology Unit, School of Medicine and Surgery, and NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, 20900 Monza, Italy; (C.M.); (L.M.); (A.C.); (G.F.); (P.A.); (E.P.); (A.C.); (E.B.); (V.R.-M.); (N.O.); (P.M.)
| | - Alessia Chiorazzi
- Experimental Neurology Unit, School of Medicine and Surgery, and NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, 20900 Monza, Italy; (C.M.); (L.M.); (A.C.); (G.F.); (P.A.); (E.P.); (A.C.); (E.B.); (V.R.-M.); (N.O.); (P.M.)
| | - Carla Scali
- Global Medical and R&D Department, Kedrion S.p.A., Località Ai Conti, Castelvecchio Pascoli, 55051 Lucca, Italy; (C.S.); (C.G.)
| | - Chiara Guarnieri
- Global Medical and R&D Department, Kedrion S.p.A., Località Ai Conti, Castelvecchio Pascoli, 55051 Lucca, Italy; (C.S.); (C.G.)
| | - Giulia Fumagalli
- Experimental Neurology Unit, School of Medicine and Surgery, and NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, 20900 Monza, Italy; (C.M.); (L.M.); (A.C.); (G.F.); (P.A.); (E.P.); (A.C.); (E.B.); (V.R.-M.); (N.O.); (P.M.)
| | - Paola Alberti
- Experimental Neurology Unit, School of Medicine and Surgery, and NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, 20900 Monza, Italy; (C.M.); (L.M.); (A.C.); (G.F.); (P.A.); (E.P.); (A.C.); (E.B.); (V.R.-M.); (N.O.); (P.M.)
| | - Eleonora Pozzi
- Experimental Neurology Unit, School of Medicine and Surgery, and NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, 20900 Monza, Italy; (C.M.); (L.M.); (A.C.); (G.F.); (P.A.); (E.P.); (A.C.); (E.B.); (V.R.-M.); (N.O.); (P.M.)
| | - Annalisa Canta
- Experimental Neurology Unit, School of Medicine and Surgery, and NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, 20900 Monza, Italy; (C.M.); (L.M.); (A.C.); (G.F.); (P.A.); (E.P.); (A.C.); (E.B.); (V.R.-M.); (N.O.); (P.M.)
| | - Elisa Ballarini
- Experimental Neurology Unit, School of Medicine and Surgery, and NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, 20900 Monza, Italy; (C.M.); (L.M.); (A.C.); (G.F.); (P.A.); (E.P.); (A.C.); (E.B.); (V.R.-M.); (N.O.); (P.M.)
| | - Virginia Rodriguez-Menendez
- Experimental Neurology Unit, School of Medicine and Surgery, and NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, 20900 Monza, Italy; (C.M.); (L.M.); (A.C.); (G.F.); (P.A.); (E.P.); (A.C.); (E.B.); (V.R.-M.); (N.O.); (P.M.)
| | - Norberto Oggioni
- Experimental Neurology Unit, School of Medicine and Surgery, and NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, 20900 Monza, Italy; (C.M.); (L.M.); (A.C.); (G.F.); (P.A.); (E.P.); (A.C.); (E.B.); (V.R.-M.); (N.O.); (P.M.)
| | - Guido Cavaletti
- Experimental Neurology Unit, School of Medicine and Surgery, and NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, 20900 Monza, Italy; (C.M.); (L.M.); (A.C.); (G.F.); (P.A.); (E.P.); (A.C.); (E.B.); (V.R.-M.); (N.O.); (P.M.)
- Correspondence:
| | - Paola Marmiroli
- Experimental Neurology Unit, School of Medicine and Surgery, and NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, 20900 Monza, Italy; (C.M.); (L.M.); (A.C.); (G.F.); (P.A.); (E.P.); (A.C.); (E.B.); (V.R.-M.); (N.O.); (P.M.)
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| |
Collapse
|
34
|
Selective activation of metabotropic glutamate receptor 7 blocks paclitaxel-induced acute neuropathic pain and suppresses spinal glial reactivity in rats. Psychopharmacology (Berl) 2021; 238:107-119. [PMID: 33089875 DOI: 10.1007/s00213-020-05662-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 09/10/2020] [Indexed: 12/30/2022]
Abstract
RATIONALE Paclitaxel-induced acute pain syndrome (P-APS), characterized by deep muscle aches and arthralgia, occurs in more than 70% of patients who receive paclitaxel. P-APS can be debilitating for patients and lead to reductions and discontinuation of potentially curable therapy. Despite being relatively common in clinical practice, no clear treatment exists for P-APS and the underlying mechanisms remain poorly defined. Regulation of glutamatergic transmission by metabotropic glutamate receptors (mGluRs) has received growing attention with respect to its role in neuropathic pain. To our knowledge, no study has been conducted on alterations and functions of group III mGluR7 signaling in P-APS. OBJECTIVES In the present study, we determined whether a single administration of paclitaxel induces glutamatergic alterations and whether mGluR7 activation blocks paclitaxel-induced neuropathic pain by suppressing glial reactivity in the spinal cord. RESULTS A single paclitaxel injection dose-dependently induced acute mechanical and thermal hypersensitivity, and was associated with increased glutamate level accompanied by reduction in mGluR7 expression in the spinal cord. Selective activation of mGluR7 by its positive allosteric modulator, AMN082, blocked the development of paclitaxel-induced acute mechanical and thermal hypersensitivity, without affecting the normal pain behavior of control rats. Moreover, activation of mGluR7 by AMN082 inhibited glial reactivity and decreased pro-inflammatory cytokine release during P-APS. Abortion of spinal glial reaction to paclitaxel alleviated paclitaxel-induced acute mechanical and thermal hypersensitivity. CONCLUSIONS There results support the hypothesis that spinal mGluR7 signaling plays an important role in P-APS; Selective activation of mGluR7 by its positive allosteric modulator, AMN082, blocks P-APS in part by reducing spinal glial reactivity and neuroinflammatory process.
Collapse
|
35
|
Caillaud M, Patel NH, Toma W, White A, Thompson D, Mann J, Tran TH, Roberts JL, Poklis JL, Bigbee JW, Fang X, Gewirtz DA, Damaj MI. A Fenofibrate Diet Prevents Paclitaxel-Induced Peripheral Neuropathy in Mice. Cancers (Basel) 2020; 13:cancers13010069. [PMID: 33383736 PMCID: PMC7795224 DOI: 10.3390/cancers13010069] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Paclitaxel, a drug used in the treatment of malignancies such as lung, ovarian and breast cancer, often produces severe side effects, among which is peripheral neuropathy. This neuropathy involves diffuse or localized pain, notably burning pain, cold and mechanical hyperexcitability. Recently, fenofibrate, a Food and Drug Administration (FDA)-approved drug for the treatment of dyslipidemia, has been shown to reduce the severity of symptoms in other forms of peripheral neuropathy. In the current work, we tested whether fenofibrate could reverse mechanical and cold hypersensitivity and improve motivation and the reduction in nerve conduction in a mouse model of paclitaxel-induced neuropathy. Our behavioral, histological and molecular assessments indicate that fenofibrate prevents the development of paclitaxel-induced neuropathy. Taken together, our studies support the therapeutic potential of fenofibrate in the prevention of paclitaxel-induced neuropathy and suggest the possible repurposing of this drug for this purpose in the clinic. Abstract Background: Paclitaxel-induced peripheral neuropathy (PIPN) is a major adverse effect of this chemotherapeutic agent that is used in the treatment of a number of solid malignancies. PIPN leads notably to burning pain, cold and mechanical allodynia. PIPN is thought to be a consequence of alterations of mitochondrial function, hyperexcitability of neurons, nerve fiber loss, oxidative stress and neuroinflammation in dorsal root ganglia (DRG) and spinal cord (SC). Therefore, reducing neuroinflammation could potentially attenuate neuropathy symptoms. Peroxisome proliferator-activated receptor-α (PPAR-α) nuclear receptors that modulate inflammatory responses can be targeted by non-selective agonists, such as fenofibrate, which is used in the treatment of dyslipidemia. Methods: Our studies tested the efficacy of a fenofibrate diet (0.2% and 0.4%) in preventing the development of PIPN. Paclitaxel (8 mg/kg) was administered via 4 intraperitoneal (i.p.) injections in C57BL/6J mice (both male and female). Mechanical and cold hypersensitivity, wheel running activity, sensory nerve action potential (SNAP), sciatic nerve histology, intra-epidermal fibers, as well as the expression of PPAR-α and neuroinflammation were evaluated in DRG and SC. Results: Fenofibrate in the diet partially prevented the development of mechanical hypersensitivity but completely prevented cold hypersensitivity and the decrease in wheel running activity induced by paclitaxel. The reduction in SNAP amplitude induced by paclitaxel was also prevented by fenofibrate. Our results indicate that suppression of paclitaxel-induced pain by fenofibrate involves the regulation of PPAR-α expression through reduction in neuroinflammation. Finally, co-administration of paclitaxel and the active metabolite of fenofibrate (fenofibric acid) did not interfere with the suppression of tumor cell growth or clonogenicity by paclitaxel in ovarian and breast cancer cell lines. Conclusions: Taken together, our results show the therapeutic potential of fenofibrate in the prevention of PIPN development.
Collapse
Affiliation(s)
- Martial Caillaud
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23284, USA; (W.T.); (A.W.); (D.T.); (J.M.); (J.L.R.); (J.L.P.)
- Correspondence: (M.C.); (M.I.D.)
| | - Nipa H. Patel
- Departments of Pharmacology and Toxicology and Medicine and Massey Cancer Center, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23284, USA; (N.H.P.); (T.H.T.); (D.A.G.)
| | - Wisam Toma
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23284, USA; (W.T.); (A.W.); (D.T.); (J.M.); (J.L.R.); (J.L.P.)
| | - Alyssa White
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23284, USA; (W.T.); (A.W.); (D.T.); (J.M.); (J.L.R.); (J.L.P.)
| | - Danielle Thompson
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23284, USA; (W.T.); (A.W.); (D.T.); (J.M.); (J.L.R.); (J.L.P.)
| | - Jared Mann
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23284, USA; (W.T.); (A.W.); (D.T.); (J.M.); (J.L.R.); (J.L.P.)
| | - Tammy H. Tran
- Departments of Pharmacology and Toxicology and Medicine and Massey Cancer Center, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23284, USA; (N.H.P.); (T.H.T.); (D.A.G.)
| | - Jane L. Roberts
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23284, USA; (W.T.); (A.W.); (D.T.); (J.M.); (J.L.R.); (J.L.P.)
| | - Justin L. Poklis
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23284, USA; (W.T.); (A.W.); (D.T.); (J.M.); (J.L.R.); (J.L.P.)
| | - John W. Bigbee
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Xianjun Fang
- Department of Biochemistry & Molecular Biology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - David A. Gewirtz
- Departments of Pharmacology and Toxicology and Medicine and Massey Cancer Center, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23284, USA; (N.H.P.); (T.H.T.); (D.A.G.)
| | - M. Imad Damaj
- Department of Pharmacology and Toxicology and Translational Research Initiative for Pain and Neuropathy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23284, USA; (W.T.); (A.W.); (D.T.); (J.M.); (J.L.R.); (J.L.P.)
- Correspondence: (M.C.); (M.I.D.)
| |
Collapse
|
36
|
Zhang R, Gan Y, Li J, Feng Y. Vagus Nerve Stimulation Transiently Mitigates Chemotherapy-Induced Peripheral Neuropathy in Rats. J Pain Res 2020; 13:3457-3465. [PMID: 33376391 PMCID: PMC7764936 DOI: 10.2147/jpr.s281190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/10/2020] [Indexed: 11/23/2022] Open
Abstract
Background Chemotherapy-induced peripheral neuropathy is a severe side effect of chemotherapeutic agents. Vagus nerve stimulation attenuates neuroinflammation by activating the cholinergic anti-inflammatory pathway and thus may attenuate CIPN. Methods Adult male Sprague-Dawley rats received intraperitoneal paclitaxel injection (2 mg/kg) every other day for a total of 4 injections. Three weeks later, the left cervical vagus nerve was exposed under general anesthesia, and the rats randomly received 20-min stimulation (1 V, 2 ms, 5 Hz, 30 s ON/5 min OFF) or sham stimulation. Heat and mechanical pain sensitivity was evaluated using Hargreaves and von Frey tests before and after treatment (n=12 per group per time point). Additionally, rats receiving paclitaxel or saline but no surgery were included. Expression of representative pro- and anti-inflammatory cytokines in dorsal root ganglia was assessed by Western blotting assays and immunohistochemistry. Results Paclitaxel significantly reduced the sensitivity for heat (withdrawal latency: paclitaxel 6.16 ± 0.54 s vs saline 9.93 ± 0.78 s, p<0.001) and mechanical pain (withdrawal frequency: paclitaxel 32.22 ± 15.51% vs saline 3.33 ± 4.92%, p<0.001). Compared with sham-stimulated rats, rats receiving vagus nerve stimulation had significantly higher sensitivity for heat (withdrawal latency: VNS 10.28 ± 1.15 s vs sham 6.27 ± 0.56 s, p<0.001) and mechanical pain (withdrawal frequency: VNS 10.00 ± 9.54% vs Sham 31.67 ± 18.99%, p=0.003) on +1 day, but not 7 days later (withdrawal latency: VNS 6.97 ± 1.13 s vs Sham 6.23 ± 0.79 s, p=0.080; withdrawal frequency: VNS 21.67 ± 11.93% vs Sham 23.33 ± 7.79%, p=0.689). Western blotting assays and immunohistochemistry revealed that interleukin-10 level was elevated in the dorsal root ganglia of rats receiving vagus nerve stimulation while no apparent changes in NF-κB or TNF-α levels were observed. Conclusion Vagus nerve stimulation could transiently attenuate paclitaxel-induced hyperalgesia in rats. Future studies are needed to investigate whether stimulation with different protocols could achieve durable effects.
Collapse
Affiliation(s)
- Ran Zhang
- Department of Anesthesiology, Peking University People's Hospital, Beijing, People's Republic of China
| | - Yu Gan
- Department of Anesthesiology, Peking University People's Hospital, Beijing, People's Republic of China
| | - Jun Li
- Department of Pain Medicine, Peking University People's Hospital, Beijing, People's Republic of China
| | - Yi Feng
- Department of Anesthesiology, Peking University People's Hospital, Beijing, People's Republic of China.,Department of Pain Medicine, Peking University People's Hospital, Beijing, People's Republic of China
| |
Collapse
|
37
|
Khasabova IA, Golovko MY, Golovko SA, Simone DA, Khasabov SG. Intrathecal administration of Resolvin D1 and E1 decreases hyperalgesia in mice with bone cancer pain: Involvement of endocannabinoid signaling. Prostaglandins Other Lipid Mediat 2020; 151:106479. [PMID: 32745525 PMCID: PMC7669692 DOI: 10.1016/j.prostaglandins.2020.106479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 07/07/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023]
Abstract
Pain produced by bone cancer is often severe and difficult to treat. Here we examined effects of Resolvin D1 (RvD1) or E1 (RvE1), antinociceptive products of ω-3 polyunsaturated fatty acids, on cancer-induced mechanical allodynia and heat hyperalgesia. Experiments were performed using a mouse model of bone cancer produced by implantation of osteolytic ficrosarcoma into and around the calcaneus bone. Mechanical allodynia and heat hyperalgesia in the tumor-bearing paw were assessed by measuring withdrawal responses to a von Frey monofilament and to radiant heat applied on the plantar hind paw. RvD1, RvE1, and cannabinoid receptor antagonists were injected intrathecally. Spinal content of endocannabinoids was evaluated using UPLC-MS/MS analysis. RvD1 and RvE1 had similar antinociceptive potencies. ED50s for RvD1 and RvE1 in reducing mechanical allodynia were 0.2 pg (0.53 fmol) and 0.6 pg (1.71 fmol), respectively, and were 0.3 pg (0.8 fmol) and 0.2 pg (0.57 fmol) for reducing heat hyperalgesia. Comparisons of dose-response relationships showed equal efficacy for reducing mechanical allodynia, however, efficacy for reducing heat hyperalgesia was greater for of RvD1. Using UPLC-MS/MS we determined that RvD1, but not RvE1, increased levels of the endocannabinoids Anandamide and 2-Arachidonoylglycerol in the spinal cord. Importantly, Resolvins did not alter acute nociception or motor function in naïve mice. Our data indicate, that RvD1 and RvE1 produce potent antiallodynia and antihyperalgesia in a model of bone cancer pain. RvD1 also triggers spinal upregulation of endocannabinoids that produce additional antinociception predominantly through CB2 receptors.
Collapse
Affiliation(s)
- Iryna A Khasabova
- Department of Diagnostic and Biological Sciences, University of Minnesota, School of Dentistry, Minneapolis, MN, USA
| | - Mikhail Y Golovko
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Svetlana A Golovko
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Donald A Simone
- Department of Diagnostic and Biological Sciences, University of Minnesota, School of Dentistry, Minneapolis, MN, USA
| | - Sergey G Khasabov
- Department of Diagnostic and Biological Sciences, University of Minnesota, School of Dentistry, Minneapolis, MN, USA.
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW The prevalence of cancer pain will continue to rise as pain is common among the survivorship and general cancer population. As interest in cannabis and cannabinoids for medicinal use including pain management continues to rise, there is growing need to update and review the current state of evidence for their use. The literature was searched for articles in English with key words cannabis, cannabinoids, and cancer pain. The sources of articles were PubMed, Embase, and open Google search. RECENT FINDINGS In a double-blind randomized placebo-controlled trial including a 3-week treatment period of nabiximol for advanced cancer patients with pain refractory to optimized opiate therapy, improvements in average pain were seen in the intention to treat population (P = 0.0854) and per- protocol population (P = 0.0378). SUMMARY To date, preclinical data has demonstrated evidence to suggest promising potential for cancer pain and the urgent need to translate this into clinical practice. Unfortunately, due to limited data, for adults with advanced cancer being treated with opiate therapy, the addition of cannabis or cannabinoids is not currently supported to address cancer pain effectively.
Collapse
|
39
|
Javid H, Rezayof A, Ghasemzadeh Z, Sardari M. The involvement of ventral hippocampal microglial cells, but not cannabinoid CB1 receptors, in morphine-induced analgesia in rats. Acta Neurol Belg 2020; 120:1077-1084. [PMID: 31006075 DOI: 10.1007/s13760-019-01144-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 04/12/2019] [Indexed: 12/12/2022]
Abstract
It is well known that glial cells are involved in pain processing. The purpose of the present study was to investigate the possible involvement of the ventral hippocampal (VH) glial cells in morphine-induced analgesia. A tail-flick apparatus was used to measure pain sensitivity in male Wistar rats that were bilaterally cannulated in the VH by stereotaxic surgery. The results showed that intraperitoneal (i.p.) administration of morphine (2.5-7.5 mg/kg) induced analgesia in a time-dependent manner. The blockade of the VH glial cell activation by bilateral microinjection of a glial inhibitor, minocycline (5-15 µg/rat) into the VH with an ineffective dose of morphine (2.5 mg/kg, i.p) significantly increased morphine analgesia. Considering that the endocannabinoid system via CB1 receptors play a crucial role in pain modulation, we also assessed the possible role of the VH cannabinoid CB1 receptors in the functional interaction between minocycline and morphine in acute pain. Our results indicated that intra-VH injection of the cannabinoid CB1 receptor agonist, arachidonylcyclopropylamide (ACPA; 4-12 ng/rat) had no effect on minocycline-induced potentiation of morphine analgesia. It should be considered that intra-VH microinjection of minocycline or ACPA by itself had no effect on tail-flick latency. Our findings suggest that the activation of the VH microglial cells may be involved in mediating pain sensation, because the inhibition of these cells by intra-VH injection of minocycline could potentiate morphine-induced analgesia. Although endocannabinoids have a regulatory role in glia function, the activation of CB1 receptors could not affect the potentiative effect of minocycline on morphine analgesia.
Collapse
Affiliation(s)
- Hanieh Javid
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, 4155-6455, Tehran, Iran
| | - Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, 4155-6455, Tehran, Iran.
| | - Zahra Ghasemzadeh
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, 4155-6455, Tehran, Iran
| | - Maryam Sardari
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, 4155-6455, Tehran, Iran
| |
Collapse
|
40
|
da Costa R, Passos GF, Quintão NL, Fernandes ES, Maia JRL, Campos MM, Calixto JB. Taxane-induced neurotoxicity: Pathophysiology and therapeutic perspectives. Br J Pharmacol 2020; 177:3127-3146. [PMID: 32352155 PMCID: PMC7312267 DOI: 10.1111/bph.15086] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/17/2020] [Accepted: 04/25/2020] [Indexed: 12/28/2022] Open
Abstract
Taxane-derived drugs are antineoplastic agents used for the treatment of highly common malignancies. Paclitaxel and docetaxel are the most commonly used taxanes; however, other drugs and formulations have been used, such as cabazitaxel and nab-paclitaxel. Taxane treatment is associated with neurotoxicity, a well-known and relevant side effect, very prevalent amongst patients undergoing chemotherapy. Painful peripheral neuropathy is the most dose-limiting side effect of taxanes, affecting up to 97% of paclitaxel-treated patients. Central neurotoxicity is an emerging side effect of taxanes and it is characterized by cognitive impairment and encephalopathy. Besides impairing compliance to chemotherapy treatment, taxane-induced neurotoxicity (TIN) can adversely affect the patient's life quality on a long-term basis. Despite the clinical relevance, not many reviews have comprehensively addressed taxane-induced neurotoxicity when they are used therapeutically. This article provides an up-to-date review on the pathophysiology of TIN and the novel potential therapies to prevent or treat this side effect.
Collapse
Affiliation(s)
- Robson da Costa
- Faculdade de FarmáciaUniversidade Federal do Rio de JaneiroRio de JaneiroRJBrazil
| | - Giselle F. Passos
- Faculdade de FarmáciaUniversidade Federal do Rio de JaneiroRio de JaneiroRJBrazil
| | - Nara L.M. Quintão
- Programa de Pós‐graduação em Ciências FarmacêuticasUniversidade do Vale do ItajaíItajaíSCBrazil
| | - Elizabeth S. Fernandes
- Instituto Pelé Pequeno PríncipeCuritibaPRBrazil
- Programa de Pós‐graduação em Biotecnologia Aplicada à Saúde da Criança e do AdolescenteFaculdades Pequeno PríncipeCuritibaPRBrazil
| | | | - Maria Martha Campos
- Escola de Ciências da Saúde e da VidaPontifícia Universidade Católica do Rio Grande do SulPorto AlegreRSBrazil
| | - João B. Calixto
- Centro de Inovação e Ensaios Pré‐clínicos ‐ CIEnPFlorianópolisSCBrazil
| |
Collapse
|
41
|
Saika F, Matsuzaki S, Kobayashi D, Ideguchi Y, Nakamura TY, Kishioka S, Kiguchi N. Chemogenetic Regulation of CX3CR1-Expressing Microglia Using Gi-DREADD Exerts Sex-Dependent Anti-Allodynic Effects in Mouse Models of Neuropathic Pain. Front Pharmacol 2020; 11:925. [PMID: 32636748 PMCID: PMC7318895 DOI: 10.3389/fphar.2020.00925] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/08/2020] [Indexed: 12/25/2022] Open
Abstract
Despite growing evidence suggesting that spinal microglia play an important role in the molecular mechanism underlying experimental neuropathic pain (NP) in male rodents, evidence regarding the sex-dependent role of these microglia in NP is insufficient. In this study, we evaluated the effects of microglial regulation on NP using Gi-designer receptors exclusively activated by designer drugs (Gi-DREADD) driven by the microglia-specific Cx3cr1 promoter. For the Cre-dependent expression of human Gi-coupled M4 muscarinic receptors (hM4Di) in CX3C chemokine receptor 1-expressing (CX3CR1+) cells, R26-LSL-hM4Di-DREADD mice were crossed with CX3CR1-Cre mice. Mouse models of NP were generated by partial sciatic nerve ligation (PSL) and treatment with anti-cancer agent paclitaxel (PTX) or oxaliplatin (OXA), and mechanical allodynia was evaluated using the von Frey test. Immunohistochemistry revealed that hM4Di was specifically expressed on Iba1+ microglia, but not on astrocytes or neurons in the spinal dorsal horn of CX3CR1-hM4Di mice. PSL-induced mechanical allodynia was significantly attenuated by systemic (intraperitoneal, i.p.) administration of 10 mg/kg of clozapine N-oxide (CNO), a hM4Di-selective ligand, in male CX3CR1-hM4Di mice. The mechanical threshold in naive CX3CR1-hM4Di mice was not altered by i.p. administration of CNO. Consistently, local (intrathecal, i.t.) administration of CNO (20 nmol) significantly relieved PSL-induced mechanical allodynia in male CX3CR1-hM4Di mice. However, neither i.p. nor i.t. administration of CNO affected PSL-induced mechanical allodynia in female CX3CR1-hM4Di mice. Both i.p. and i.t. administration of CNO relieved PTX-induced mechanical allodynia in male CX3CR1-hM4Di mice, and a limited effect of i.p. CNO was observed in female CX3CR1-hM4Di mice. Unlike PTX-induced allodynia, OXA-induced mechanical allodynia was slightly improved, but not significantly relieved, by i.p. administration of CNO in both male and female CX3CR1-hM4Di mice. These results suggest that spinal microglia can be regulated by Gi-DREADD and support the notion that CX3CR1+ spinal microglia play sex-dependent roles in nerve injury-induced NP; however, their roles may vary among different models of NP.
Collapse
Affiliation(s)
- Fumihiro Saika
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| | - Shinsuke Matsuzaki
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| | - Daichi Kobayashi
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan.,Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yuya Ideguchi
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| | - Tomoe Y Nakamura
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| | - Shiroh Kishioka
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan.,Faculty of Wakayama Health Care Sciences, Takarazuka University of Medical and Health Care, Wakayama, Japan
| | - Norikazu Kiguchi
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
42
|
Staff NP, Fehrenbacher JC, Caillaud M, Damaj MI, Segal RA, Rieger S. Pathogenesis of paclitaxel-induced peripheral neuropathy: A current review of in vitro and in vivo findings using rodent and human model systems. Exp Neurol 2020; 324:113121. [PMID: 31758983 PMCID: PMC6993945 DOI: 10.1016/j.expneurol.2019.113121] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/29/2019] [Accepted: 11/19/2019] [Indexed: 12/22/2022]
Abstract
Paclitaxel (Brand name Taxol) is widely used in the treatment of common cancers like breast, ovarian and lung cancer. Although highly effective in blocking tumor progression, paclitaxel also causes peripheral neuropathy as a side effect in 60-70% of chemotherapy patients. Recent efforts by numerous labs have aimed at defining the underlying mechanisms of paclitaxel-induced peripheral neuropathy (PIPN). In vitro models using rodent dorsal root ganglion neurons, human induced pluripotent stem cells, and rodent in vivo models have revealed a number of molecular pathways affected by paclitaxel within axons of sensory neurons and within other cell types, such as the immune system and peripheral glia, as well skin. These studies revealed that paclitaxel induces altered calcium signaling, neuropeptide and growth factor release, mitochondrial damage and reactive oxygen species formation, and can activate ion channels that mediate responses to extracellular cues. Recent studies also suggest a role for the matrix-metalloproteinase 13 (MMP-13) in mediating neuropathy. These diverse changes may be secondary to paclitaxel-induced microtubule transport impairment. Human genetic studies, although still limited, also highlight the involvement of cytoskeletal changes in PIPN. Newly identified molecular targets resulting from these studies could provide the basis for the development of therapies with which to either prevent or reverse paclitaxel-induced peripheral neuropathy in chemotherapy patients.
Collapse
Affiliation(s)
- Nathan P Staff
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jill C Fehrenbacher
- Department of Pharmacology and Toxicology, University School of Medicine, Indianapolis, IN 46202, USA
| | - Martial Caillaud
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, USA
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, USA
| | - Rosalind A Segal
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sandra Rieger
- Department of Biology, University of Miami, Coral Gables, FL 33146, USA.
| |
Collapse
|
43
|
Zhao YX, Yao MJ, Liu Q, Xin JJ, Gao JH, Yu XC. Electroacupuncture Treatment Attenuates Paclitaxel-Induced Neuropathic Pain in Rats via Inhibiting Spinal Glia and the TLR4/NF-κB Pathway. J Pain Res 2020; 13:239-250. [PMID: 32099448 PMCID: PMC7005725 DOI: 10.2147/jpr.s241101] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 01/14/2020] [Indexed: 12/13/2022] Open
Abstract
Background and Purpose Neuropathic pain is a major side-effect of paclitaxel (PTX) chemotherapy. Although the precise mechanisms responsible for this pain are unclear, the activation of neuroglia and upregulation of the TLR4/NF-κB pathway are known to be involved. In this study, we determined whether electroacupuncture (EA) could limit mechanical hypersensitivity resulting from the chemotherapeutic drug PTX in rats, and investigated the potential mechanisms involved. Methods Rats intraperitoneally received a cumulative dose of 8 mg/kg PTX (2 mg/kg per day) or vehicle control on alternate days (day 0, 2, 4 and 6). EA treatment (10 Hz, 1 mA) was applied at bilateral ST36 acupoints in rats once every other day on days 0–14. For sham EA, needles were inserted at ST36 acupoints without electrical stimulation. Mechanical allodynia was measured by mechanical withdrawal latency (MWL) of paws to a mechanical stimulus every 2 days. Protein expression of TLR4 and NF-κB p65, as well as TMEM119 and GFAP (indicators of microglia and astrocytes, respectively) in spinal cord was quantified by Western blot analysis. Levels of inflammatory cytokines IL-1β and TNF-α in spinal cord and serum were detected by ELISA. Results Mechanical allodynia induced by PTX in both paws (right and left) of rats was significantly attenuated by EA but not sham EA treatment. In addition, EA, but not sham EA, inhibited the activation of both microglia (TMEM119) and astrocytes (GFAP) in lumbar spinal cord. Moreover, Western blot analysis revealed that protein expression of TLR4 and NF-κB in spinal cord was suppressed by EA but not sham EA treatment. PTX significantly increased inflammatory cytokines in spinal cord and serum, which were ameliorated by EA treatment but not by sham EA. Conclusion These results indicate that EA treatment attenuates PTX-induced mechanical allodynia. The putative mechanism corroborating this finding could be related to the suppression of activated microglia and astrocytes in spinal cord, as well as the inhibition of the activated TLR4/NF-κB signaling pathway by EA treatment.
Collapse
Affiliation(s)
- Yu-Xue Zhao
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, People's Republic of China
| | - Ming-Jiang Yao
- Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing 100091, People's Republic of China.,Key Laboratory of Pharmacology of Chinese Materia Medica, Beijing 100091, People's Republic of China
| | - Qun Liu
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, People's Republic of China
| | - Juan-Juan Xin
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, People's Republic of China
| | - Jun-Hong Gao
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, People's Republic of China
| | - Xiao-Chun Yu
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, People's Republic of China
| |
Collapse
|
44
|
Wu Z, Li L, Xie F, Xu G, Dang D, Yang Q. Enhancing KCNQ Channel Activity Improves Neurobehavioral Recovery after Spinal Cord Injury. J Pharmacol Exp Ther 2020; 373:72-80. [PMID: 31969383 DOI: 10.1124/jpet.119.264010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/17/2020] [Indexed: 01/08/2023] Open
Abstract
Spinal cord injury (SCI) usually leads to acute neuronal death and delayed secondary degeneration, resulting in sensory dysfunction, paralysis, and chronic pain. Excessive excitation is one of the critical factors leading to secondary neural damage initiated by various insults. KCNQ/Kv7 channels are highly expressed in spinal neurons and axons and play an important role in controlling their excitability. Enhancing KCNQ channel activity by using its specific opener retigabine could thus be a plausible treatment strategy to reduce the pathology after SCI. We produced contusive SCI at T10 in adult male rats, which then received 10 consecutive days' treatment with retigabine or vehicle starting 3 hours or 3 days after contusion. Two different concentrations and two different delivery methods were applied. Delivery of retigabine via Alzet osmotic pumps, but not intraperitoneal injections 3 hours after contusion, promoted recovery of locomotor function. Remarkably, retigabine delivery in both methods significantly attenuated the development of mechanical stimuli-induced hyperreflexia and spontaneous pain; however, no significant difference in the thermal threshold was observed. Although retigabine delivered 3 days after contusion significantly attenuated the development of mechanical hypersensitivity and spontaneous pain, the locomotor function is not improved by the delayed treatments. Finally, we found that early application of retigabine attenuates the inflammatory activity in the spinal cord and increases the survival of white matter after SCI. Our results suggest that decreasing neuronal excitability by targeting KCNQ/Kv7 channels at acute stage aids the recovery of locomotor function and attenuates the development of neuropathic pain after SCI. SIGNIFICANCE STATEMENT: Several pharmacological interventions have been proposed for spinal cord injury (SCI) treatment, but none have been shown to be both effective and safe in clinical trials. Necrotic neuronal death and chronic pain are often the cost of pathological neural excitation after SCI. We show that early, brief application of retigabine could aid locomotor and sensory neurobehavioral recovery after SCI, supporting the use of this drug in the clinic to promote motor and sensory function in patients with SCI.
Collapse
Affiliation(s)
- Zizhen Wu
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (Z.W.); Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, Texas (L.L., F.X.); Department of Critical Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (F.X.); and Department of Neuroscience, Cell Biology and Anatomy at University of Texas Medical Branch, Galveston, Texas (G.X., D.D., Q.Y.)
| | - Lin Li
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (Z.W.); Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, Texas (L.L., F.X.); Department of Critical Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (F.X.); and Department of Neuroscience, Cell Biology and Anatomy at University of Texas Medical Branch, Galveston, Texas (G.X., D.D., Q.Y.)
| | - Fuhua Xie
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (Z.W.); Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, Texas (L.L., F.X.); Department of Critical Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (F.X.); and Department of Neuroscience, Cell Biology and Anatomy at University of Texas Medical Branch, Galveston, Texas (G.X., D.D., Q.Y.)
| | - Guoying Xu
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (Z.W.); Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, Texas (L.L., F.X.); Department of Critical Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (F.X.); and Department of Neuroscience, Cell Biology and Anatomy at University of Texas Medical Branch, Galveston, Texas (G.X., D.D., Q.Y.)
| | - Danny Dang
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (Z.W.); Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, Texas (L.L., F.X.); Department of Critical Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (F.X.); and Department of Neuroscience, Cell Biology and Anatomy at University of Texas Medical Branch, Galveston, Texas (G.X., D.D., Q.Y.)
| | - Qing Yang
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (Z.W.); Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, Texas (L.L., F.X.); Department of Critical Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (F.X.); and Department of Neuroscience, Cell Biology and Anatomy at University of Texas Medical Branch, Galveston, Texas (G.X., D.D., Q.Y.)
| |
Collapse
|
45
|
Slivicki RA, Mali SS, Hohmann AG. Voluntary exercise reduces both chemotherapy-induced neuropathic nociception and deficits in hippocampal cellular proliferation in a mouse model of paclitaxel-induced peripheral neuropathy. NEUROBIOLOGY OF PAIN 2019; 6:100035. [PMID: 31528755 PMCID: PMC6739464 DOI: 10.1016/j.ynpai.2019.100035] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/21/2019] [Accepted: 08/25/2019] [Indexed: 12/18/2022]
Abstract
Paclitaxel treatment did not alter voluntary running activity. Voluntary running reduced mechanical and cold allodynia induced by paclitaxel. Voluntary running reduced paclitaxel-induced deficits in hippocampal cellular proliferation.
Chemotherapy-induced peripheral neuropathy (CIPN) is a common dose-limiting side-effect of all major chemotherapeutic agents. Here, we explored efficacy of voluntary exercise as a nonpharmacological strategy for suppressing two distinct adverse side effects of chemotherapy treatment. We evaluated whether voluntary running would suppress both neuropathic pain and deficits in hippocampal cell proliferation in a mouse model of CIPN induced by the taxane chemotherapeutic agent paclitaxel. Mice were given free access to running wheels or were housed without running wheels during one of three different intervention phases: 1) during the onset (i.e. development phase) of paclitaxel-induced neuropathy, 2) prior to dosing with paclitaxel or its vehicle, or 3) following the establishment (i.e. maintenance phase) of paclitaxel-induced neuropathy. Paclitaxel treatment did not alter running wheel behavior relative to vehicle-treated animals in any study. Animals that engaged in voluntary running during the development phase of paclitaxel-induced neuropathy failed to display mechanical or cold hypersensitivities relative to sedentary control animals that did not have access to running wheels. A prior history of voluntary running delayed the onset of, but did not fully prevent, development of paclitaxel-induced neuropathic pain behavior. Voluntary running reduced already established mechanical and cold allodynia induced by paclitaxel. Importantly, voluntary running did not alter mechanical or cold responsivity in vehicle-treated animals, suggesting that the observed antinociceptive effect of exercise was dependent upon the presence of the pathological pain state. In the same animals evaluated for nociceptive responding, paclitaxel also reduced cellular proliferation but not cellular survival in the dentate gyrus of the hippocampus, as measured by immunohistochemistry for Ki67 and BrdU expression, respectively. Voluntary running abrogated paclitaxel-induced reductions in cellular proliferation to levels observed in vehicle-treated mice and also increased BrdU expression levels irrespective of chemotherapy treatment. Our studies support the hypothesis that voluntary exercise may be beneficial in suppressing both neuropathic pain and markers of hippocampal cellular function that are impacted by toxic challenge with chemotherapeutic agents.
Collapse
Affiliation(s)
- Richard A. Slivicki
- Program in Neuroscience, Indiana University, Bloomington, IN, United States
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Sonali S. Mali
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Andrea G. Hohmann
- Program in Neuroscience, Indiana University, Bloomington, IN, United States
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
- Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States
- Corresponding author at: Department of Psychological and Brain Sciences, Indiana University, 1101 E 10th Street, Bloomington, IN 47405-7007, United States.
| |
Collapse
|
46
|
Toma W, Kyte SL, Bagdas D, Jackson A, Meade JA, Rahman F, Chen ZJ, Del Fabbro E, Cantwell L, Kulkarni A, Thakur GA, Papke RL, Bigbee JW, Gewirtz DA, Damaj MI. The α7 nicotinic receptor silent agonist R-47 prevents and reverses paclitaxel-induced peripheral neuropathy in mice without tolerance or altering nicotine reward and withdrawal. Exp Neurol 2019; 320:113010. [PMID: 31299179 DOI: 10.1016/j.expneurol.2019.113010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/05/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022]
Abstract
Various antitumor drugs, including paclitaxel, frequently cause chemotherapy-induced peripheral neuropathy (CIPN) that can be sustained even after therapy has been completed. The current work was designed to evaluate R-47, an α7 nAChR silent agonist, in our mouse model of CIPN. R-47 was administered to male C57BL/6J mice prior to and during paclitaxel treatment. Additionally, we tested if R-47 would alter nicotine's reward and withdrawal effects. The H460 and A549 non-small cell lung cancer (NSCLC) cell lines were exposed to R-47 for 24-72 h, and tumor-bearing NSG mice received R-47 prior to and during paclitaxel treatment. R-47 prevents and reverses paclitaxel-induced mechanical hypersensitivity in mice in an α7 nAChR-dependent manner. No tolerance develops following repeated administration of R-47, and the drug lacks intrinsic rewarding effects. Additionally, R-47 neither changes the rewarding effect of nicotine in the Conditioned Place Preference test nor enhances mecamylamine-precipitated withdrawal. Furthermore, R-47 prevents paclitaxel-mediated loss of intraepidermal nerve fibers and morphological alterations of microglia in the spinal cord. Moreover, R-47 does not increase NSCLC cell viability, colony formation, or proliferation, and does not interfere with paclitaxel-induced growth arrest, DNA fragmentation, or apoptosis. Most importantly, R-47 does not increase the growth of A549 tumors or interfere with the antitumor activity of paclitaxel in tumor-bearing mice. These studies suggest that R-47 could be a viable and efficacious approach for the prevention and treatment of CIPN that would not interfere with the antitumor activity of paclitaxel or promote lung tumor growth.
Collapse
Affiliation(s)
- Wisam Toma
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States of America.
| | - S Lauren Kyte
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Rockville, MD, United States of America
| | - Deniz Bagdas
- Department of Psychiatry, Yale University School of Medicine, Yale Tobacco Center of Regulatory Science, New Haven, CT, United States of America
| | - Asti Jackson
- Department of Psychiatry, Yale University School of Medicine, Yale Tobacco Center of Regulatory Science, New Haven, CT, United States of America
| | - Julie A Meade
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Faria Rahman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Zhi-Jian Chen
- Department of Neurology, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Egidio Del Fabbro
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States of America; Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Lucas Cantwell
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States of America
| | - Abhijit Kulkarni
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States of America
| | - Ganesh A Thakur
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States of America
| | - Roger L Papke
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, United States of America
| | - John W Bigbee
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States of America
| | - David A Gewirtz
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States of America
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States of America; Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, Richmond, VA, United States of America
| |
Collapse
|
47
|
Chemokine Signaling in Chemotherapy-Induced Neuropathic Pain. Int J Mol Sci 2019; 20:ijms20122904. [PMID: 31197114 PMCID: PMC6627296 DOI: 10.3390/ijms20122904] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 12/17/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a side effect of chemotherapics such as taxanes, vinca alkaloids, and platinum compounds. In recent years, several reports have indicated the involvement of different molecular mechanisms in CIPN. The pathways described so far are diverse and target various components of the peripheral Nervous System (PNS). Among the contributors to neuropathic pain, inflammation has been indicated as a powerful driver of CIPN. Several pieces of evidence have demonstrated a chemotherapy-induced increase in peripheral pro-inflammatory cytokines and a strong correlation with peripheral neuropathy. At present, there are not adequate strategies to prevent CIPN, although there are drugs for treating CIPN, such as duloxetine, that have displayed a moderate effect on CIPN. In this review, we focus on the players involved in CIPN with a particular emphasis on chemokine signaling.
Collapse
|
48
|
Blanton HL, Brelsfoard J, DeTurk N, Pruitt K, Narasimhan M, Morgan DJ, Guindon J. Cannabinoids: Current and Future Options to Treat Chronic and Chemotherapy-Induced Neuropathic Pain. Drugs 2019; 79:969-995. [PMID: 31127530 PMCID: PMC8310464 DOI: 10.1007/s40265-019-01132-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Increases in cancer diagnosis have tremendous negative impacts on patients and their families, and major societal and economic costs. The beneficial effect of chemotherapeutic agents on tumor suppression comes with major unwanted side effects such as weight and hair loss, nausea and vomiting, and neuropathic pain. Chemotherapy-induced peripheral neuropathy (CIPN), which can include both painful and non-painful symptoms, can persist 6 months or longer after the patient's last chemotherapeutic treatment. These peripheral sensory and motor deficits are poorly treated by our current analgesics with limited effectiveness. Therefore, the development of novel treatment strategies is an important preclinical research focus and an urgent need for patients. Approaches to prevent CIPN have yielded disappointing results since these compounds may interfere with the anti-tumor properties of chemotherapeutic agents. Nevertheless, the first (serotonin noradrenaline reuptake inhibitors [SNRIs], anticonvulsants, tricyclic antidepressants) and second (5% lidocaine patches, 8% capsaicin patches and weak opioids such as tramadol) lines of treatment for CIPN have shown some efficacy. The clinical challenge of CIPN management in cancer patients and the need to target novel therapies with long-term efficacy in alleviating CIPN are an ongoing focus of research. The endogenous cannabinoid system has shown great promise and efficacy in alleviating CIPN in preclinical and clinical studies. In this review, we will discuss the mechanisms through which the platinum, taxane, and vinca alkaloid classes of chemotherapeutics may produce CIPN and the potential therapeutic effect of drugs targeting the endocannabinoid system in preclinical and clinical studies, in addition to cannabinoid compounds diffuse mechanisms of action in alleviation of CIPN.
Collapse
Affiliation(s)
- Henry L Blanton
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Jennifer Brelsfoard
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Nathan DeTurk
- Department of Anesthesiology and Department of Pharmacology, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Kevin Pruitt
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6592, Lubbock, TX, 79430, USA
| | - Madhusudhanan Narasimhan
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Daniel J Morgan
- Department of Anesthesiology and Department of Pharmacology, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Josée Guindon
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA.
| |
Collapse
|
49
|
Banister SD, Krishna Kumar K, Kumar V, Kobilka BK, Malhotra SV. Selective modulation of the cannabinoid type 1 (CB 1) receptor as an emerging platform for the treatment of neuropathic pain. MEDCHEMCOMM 2019; 10:647-659. [PMID: 31191856 PMCID: PMC6533890 DOI: 10.1039/c8md00595h] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/12/2019] [Indexed: 12/27/2022]
Abstract
Neuropathic pain is caused by a lesion or dysfunction in the nervous system, and it may arise from illness, be drug-induced or caused by toxin exposure. Since the discovery of two G-protein-coupled cannabinoid receptors (CB1 and CB2) nearly three decades ago, there has been a rapid expansion in our understanding of cannabinoid pharmacology. This is currently one of the most active fields of neuropharmacology, and interest has emerged in developing cannabinoids and other small molecule modulators of CB1 and CB2 as therapeutics for neuropathic pain. This short review article provides an overview of the chemotypes currently under investigation for the development of novel neuropathic pain treatments targeting CB1 receptors.
Collapse
Affiliation(s)
- Samuel D Banister
- Department of Radiation Oncology , Stanford University School of Medicine , Stanford , CA 94305 , USA .
| | - Kaavya Krishna Kumar
- Department of Molecular and Cellular Physiology , Stanford University School of Medicine , Stanford , CA 94305 , USA
| | - Vineet Kumar
- Department of Radiation Oncology , Stanford University School of Medicine , Stanford , CA 94305 , USA .
| | - Brian K Kobilka
- Department of Molecular and Cellular Physiology , Stanford University School of Medicine , Stanford , CA 94305 , USA
| | - Sanjay V Malhotra
- Department of Radiation Oncology , Stanford University School of Medicine , Stanford , CA 94305 , USA .
| |
Collapse
|
50
|
Yan X, Li F, Maixner DW, Yadav R, Gao M, Ali MW, Hooks SB, Weng HR. Interleukin-1beta released by microglia initiates the enhanced glutamatergic activity in the spinal dorsal horn during paclitaxel-associated acute pain syndrome. Glia 2019; 67:482-497. [PMID: 30578561 DOI: 10.1002/glia.23557] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 10/08/2018] [Accepted: 10/08/2018] [Indexed: 01/30/2023]
Abstract
Patients receiving paclitaxel for cancer treatment often develop an acute pain syndrome (paclitaxel-associated acute pain syndrome, P-APS), which occurs immediately after paclitaxel treatment. Mechanisms underlying P-APS remain largely unknown. We recently reported that rodents receiving paclitaxel develop acute pain and activation of spinal microglial toll like receptor 4 (TLR4) by paclitaxel penetrating into the spinal cord is a critical event in the genesis of P-APS. Our current study dissected cellular and molecular mechanisms underlying the P-APS. We demonstrated that bath-perfusion of paclitaxel, at a concentration similar to that found in the cerebral spinal fluid in animals receiving i.v. paclitaxel (2 mg/kg), resulted in increased calcium activity in microglia instantly, and in astrocytes with 6 min delay. TLR4 activation in microglia by paclitaxel caused microglia to rapidly release interleukin-1β (IL-1β) but not tumor necrosis factor α, IL-6, or interferon-γ. IL-1β release from microglia depended on capthepsin B. IL-1β acted on astrocytes, leading to elevated calcium activity and suppressed glutamate uptake. IL-1β also acted on neurons to increase presynaptic glutamate release and postsynaptic AMPA receptor activity in the spinal dorsal horn. Knockout of IL-1 receptors prevented the development of acute pain induced by paclitaxel in mice. Our study indicates that IL-1β is a crucial molecule used by microglia to alter functions in astrocytes and neurons upon activation of TLR4 in the genesis of P-APS, and targeting the signaling pathways regulating the production and function of IL-1β from microglia is a potential avenue for the development of analgesics for the treatment of P-APS.
Collapse
Affiliation(s)
- Xisheng Yan
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, Athens, Georgia
- Department of Cardiovascular Medicine, Wuhan Third Hospital, Wuhan, China
| | - Fen Li
- Department of Neurology, Wuhan Third Hospital, Wuhan, China
| | - Dylan W Maixner
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, Athens, Georgia
| | - Ruchi Yadav
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, Athens, Georgia
| | - Mei Gao
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, Athens, Georgia
| | - Mourad Wagdy Ali
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, Athens, Georgia
| | - Shelley B Hooks
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, Athens, Georgia
| | - Han-Rong Weng
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, Athens, Georgia
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, Georgia
| |
Collapse
|