1
|
Kwon K, Ahn J, Kim SN, Park HY, Bang C, Kim SI, Kim YH. Biomarkers in cerebrospinal fluid of persistent neuropathic pain after lumbar stenosis surgery. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2025:10.1007/s00586-025-08832-x. [PMID: 40381015 DOI: 10.1007/s00586-025-08832-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/22/2025] [Accepted: 03/29/2025] [Indexed: 05/19/2025]
Abstract
PURPOSE To analyze which CSF biomarkers affect persisting neuropathic pain (NP) after surgery. METHODS CSF samples were gathered from patients who had a dural tear injury during surgery or who underwent myelography, from July 2022 to July 2023. Patients were evaluated for pain and NP using LANSS, PDQ, DN4, ODI, and NRS questionnaires preoperatively, and at 1 month, 3 months, and 1 year postoperatively, and divided into three groups based on the presence of NP. CSF samples were processed and analyzed using ELISA to measure various biomarkers, including IL-6, Her1, MCP1, P-NFH, Apolipoprotein A1, MBP, IL-8, TNF- α, and BDNF. RESULTS Among 22 patients, 16 had preoperative pain; 8 resolved and 8 persisted postoperatively. Patients were categorized into Group A (no NP), Group B (resolved NP), and Group C (persistent NP). Comparisons revealed no significant differences according to preoperative NP presence. However, patients with persistent postoperative NP had significantly lower levels of Her1 (p = 0.019), IL-6 (p = 0.025), and MCP-1 (p = 0.039). Intergroup comparisons confirmed significantly lower IL-6 levels in group C compared to groups A and B. Her1 and MCP-1 levels showed marginal significance. Moderate correlation was found between IL-6 levels and NP improvement post-surgery (Spearman's rho: 0.56, p = 0.024; Kendall's tau: 0.48, p = 0.03), whereas IL-6 negatively correlated with persisting postoperative NP (r = -0.55, p = 0.008). CONCLUSION IL-6 concentration was significantly lower in patients with persisting NP after the surgery as well as other biomarkers such as Her1 and MCP-1. It can be suggested that such biomarkers associated with NP primarily impact the acute phase, while persistent NP requires distinct consideration and treatment approaches.
Collapse
Affiliation(s)
- Kihyun Kwon
- Seoul St. Mary's Hospital, Catholic University of Korea, Seoul, Republic of Korea
| | - Joonghyun Ahn
- Bucheon St. Mary's Hospital, Catholic University of Korea, Bucheon-Si, Republic of Korea
| | - So-Na Kim
- Seoul St. Mary's Hospital, Catholic University of Korea, Seoul, Republic of Korea
| | - Hyung-Youl Park
- Eunpyeong Hospital, Catholic University of Korea, Seoul, Republic of Korea
| | - Chungwon Bang
- Incheon St. Mary's Hospital, Catholic University of Korea, Incheon, Republic of Korea
| | - Sang-Il Kim
- Seoul St. Mary's Hospital, Catholic University of Korea, Seoul, Republic of Korea
| | - Young-Hoon Kim
- Seoul St. Mary's Hospital, Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Liu LL, Liu X, Zhao S, Li Z, Liu JX, Ma DY, Wang XL. Necroptosis of hippocampal neurons in paclitaxel chemotherapy-induced cognitive impairment mediates microglial activation via TLR4/MyD88 signaling pathway. Open Med (Wars) 2025; 20:20251182. [PMID: 40322465 PMCID: PMC12048903 DOI: 10.1515/med-2025-1182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 05/08/2025] Open
Abstract
Background Paclitaxel (PTX) chemotherapy frequently induces cognitive impairment, which is closely associated with two key pathological processes: necroptosis of hippocampal neurons and microglial polarization. Necroptotic neurons release damage-associated molecular patterns, triggering inflammatory responses. As the primary immune cells in the central nervous system, microglia can exhibit either pro-inflammatory or anti-inflammatory activity depending on their polarization state. However, the relationship between PTX-induced neuronal necroptosis and microglial activation remains unclear. Methods In this study, both in vivo and in vitro experiments were conducted. In vivo, an adult male C57BL/6N mouse model of PTX-induced cognitive impairment was established and divided into three groups: Veh (vehicle control), PTX (paclitaxel only), and P + N (paclitaxel with Nec-1 treatment). Necrostatin-1 (Nec-1), a specific inhibitor of RIPK1, was used to inhibit necroptosis. In vitro, HT22 cells were used to prepare necroptosis-conditioned medium, and BV-2 cells were treated with this medium. TAK-242, a TLR4 inhibitor, was used to explore the role of the TLR4/MyD88 signaling pathway. Immunofluorescence staining, western blot, and ELISA were employed to detect relevant markers and cytokines. Results The results demonstrated that PTX-induced necroptosis of hippocampal neurons activated microglia. Nec-1 effectively suppressed neuronal necroptosis and reduced M1 polarization of microglia. The TLR4/MyD88 signaling pathway was involved in microglial polarization induced by the necroptotic-conditioned medium of PTX-treated HT22 cells. TAK-242 significantly blocked the regulatory effect of PTX-induced neuronal necroptosis on BV-2 microglial polarization. Conclusion This study reveals that hippocampal neuron necroptosis activates microglia through the TLR4/MyD88 signaling pathway in PTX-induced cognitive impairment, promoting M1 polarization and neuroinflammation. Inhibiting necroptosis promotes M2 polarization and neuroprotection. These findings uncover a novel mechanism of PTX-induced cognitive impairment and suggest potential therapeutic targets.
Collapse
Affiliation(s)
- Lan-Lan Liu
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, P.R. China
| | - Xin Liu
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, P.R. China
| | - Shuang Zhao
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, P.R. China
| | - Zhao Li
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, P.R. China
| | - Jia-Xin Liu
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, P.R. China
| | - Dong-Yang Ma
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, P.R. China
| | - Xiu-Li Wang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, P.R. China
| |
Collapse
|
3
|
Dong B, Li D, Song S, He N, Yue S, Yin S. MTOR Promotes Astrocyte Activation and Participates in Neuropathic Pain through an Upregulation of RIP3. Neurochem Res 2025; 50:93. [PMID: 39893345 PMCID: PMC11787194 DOI: 10.1007/s11064-025-04341-x] [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: 09/02/2024] [Revised: 11/27/2024] [Accepted: 01/16/2025] [Indexed: 02/04/2025]
Abstract
Neuropathic pain (NP), a chronic pain condition, is the result of abnormalities in both central and peripheral pain conduction pathways. Here, we investigated the underlying mechanisms associated with this effect. We found that following chronic constriction injury (CCI) surgery, there was an increase of mTOR in astrocytes and an activation of astrocytes within the spinal cord. Pharmacological inhibition of mTOR reversed CCI-induced hyperalgesia and neuroinflammation. Moreover, knockdown of astrocytic mTOR rescued the downregulation of spinal glutamate metabolism-related protein expression, underscoring the pivotal role of mTOR in modulating this pathway. Intriguingly, we observed that overexpression of mTOR, achieved via intrathecal administration of TSC2-shRNA, led to an upregulation of RIP3. Notably, pharmacological inhibition of RIP3, while ineffective in modulating mTOR activation, effectively eliminated the mTOR-induced astrocyte activation. Mechanistically, we found that mTOR controlled the expression of RIP3 in astrocytes through ITCH-mediated ubiquitination and an autophagy-dependent degradation. Taken together, our results reveal an unanticipated link between mTOR and RIP3 in promoting astrocyte activation, providing new avenues of investigation directed toward the management and treatment of NP.
Collapse
Affiliation(s)
- Bingru Dong
- Rehabilitation Center, Qilu Hospital of Shandong University, Jinan, 250000, Shandong, China
- Institute of Rehabilitation Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266000, Shandong, China
| | - Danyang Li
- Rehabilitation Center, Qilu Hospital of Shandong University, Jinan, 250000, Shandong, China
| | - Shasha Song
- Rehabilitation Center, Qilu Hospital of Shandong University, Jinan, 250000, Shandong, China
| | - Na He
- Rehabilitation Center, Qilu Hospital of Shandong University, Jinan, 250000, Shandong, China
| | - Shouwei Yue
- Rehabilitation Center, Qilu Hospital of Shandong University, Jinan, 250000, Shandong, China.
| | - Sen Yin
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250000, Shandong, China.
| |
Collapse
|
4
|
Gao P, Peng F, Liu J, Wu W, Zhao G, Liu C, Cao H, Li Y, Qiu F, Zhang W. Lidocaine Enhanced Antitumor Efficacy and Relieved Chemotherapy-Induced Hyperalgesia in Mice with Metastatic Gastric Cancer. Int J Mol Sci 2025; 26:828. [PMID: 39859541 PMCID: PMC11766172 DOI: 10.3390/ijms26020828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
With the widespread use of lidocaine for pain control in cancer therapy, its antitumor activity has attracted considerable attention in recent years. This paper provides a simple strategy of combining lidocaine with chemotherapy drugs for cancer therapy, aiming to relieve chemotherapy-induced pain and achieve stronger antitumor efficacy. However, there is still a lack of substantial pre-clinical evidence for the efficacy and related mechanisms of such combinations, obstructing their potential clinical application. In this study, we propose intraperitoneal chemotherapy (IPC) against gastric cancer (GC) as an ideal scenario to evaluate the efficacy of a lidocaine/paclitaxel combination. Firstly, we used human GC cells MKN-45-luc to investigate the antitumor activity and related mechanisms of the lidocaine/paclitaxel combination in vitro. Then, we used C57BL/6 mice with intraperitoneal drug suffusion to evaluate the efficacy of lidocaine to suppress paclitaxel-induced hyperalgesia and related mechanisms. Lastly, in BALB/c tumor-bearing nude mice we evaluated the synergistic antitumor activity and pain-relieving effect of the lidocaine/paclitaxel combination. Our results showed enhanced antitumor activity for the lidocaine/paclitaxel combination, which induced apoptosis, inhibited migration, and the invasion of GC cells in a synergistic manner. In animal models, the lidocaine/paclitaxel combination effectively inhibited growth and peritoneal metastasis of the tumor, resulting in prolonged survival time. Meanwhile, lidocaine showed considerable anti-inflammatory activity alongside its anesthetic effect, which, in combination, effectively relieved hyperalgesia induced by paclitaxel. These results suggested that intraperitoneal suffusion with lidocaine/paclitaxel could be a pain-free IPC formulation with enhanced antitumor activity, which could provide a promising treatment for GC with peritoneal metastasis.
Collapse
Affiliation(s)
- Peiwen Gao
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (P.G.); (J.L.); (W.W.)
| | - Fei Peng
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| | - Jing Liu
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (P.G.); (J.L.); (W.W.)
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| | - Weiwei Wu
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (P.G.); (J.L.); (W.W.)
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| | - Guoyan Zhao
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| | - Congyan Liu
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| | - Hangxue Cao
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| | - Yuncheng Li
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| | - Feng Qiu
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (P.G.); (J.L.); (W.W.)
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| | - Wensheng Zhang
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (P.G.); (J.L.); (W.W.)
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (F.P.); (G.Z.); (C.L.); (H.C.); (Y.L.)
| |
Collapse
|
5
|
Liu Y, Hao S, Hao H, Zheng G, Bing J, Kang L, Li J, Zhao H, Hao H. Construction of a Novel Necroptosis-Related Signature in Rat DRG for Neuropathic Pain. J Inflamm Res 2025; 18:147-165. [PMID: 39802520 PMCID: PMC11720641 DOI: 10.2147/jir.s494286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/27/2024] [Indexed: 01/16/2025] Open
Abstract
Background Recent studies have shown necroptosis may play a role in the development of inflammation-associated pain. However, research on the correlation between necroptosis-related genes and neuropathic pain in the dorsal root ganglia (DRG) is limited. This study aims to identify a gene signature related to necroptosis in DRG that can predict neuropathic pain. Methods The mRNA expression profiles associated with neuropathic pain (GSE24982 and GSE30691) were acquired from the Gene Expression Omnibus (GEO) database. The Least Absolute Shrinkage and Selection Operator (Lasso) and Support Vector Machine-Recursive Feature Elimination (SVM-RFE) regressions were performed in GSE24982 database to constructed the necroptosis-related diferentially expressed genes (NRDEGs) signature related to neuropathic pain. Nomogram, Receiver Operating Characteristic (ROC), GSE30691 database analysis and basic experiments were used to verify the accuracy of the signature. Go and KEGG analysis, interaction network and immune infiltration were used to analyze the biological function of the signature. Results A predictive signature targeting rat DRG for neuropathic pain through a variety of methods to verify the accuracy was developed based on 3 NRDEGs (TLR4, CAPN2, RIPK3). Significantly enriched KEGG and GO pathways, drug target prediction and non-coding RNAs related to the signature holded promise for advancing our understanding of potential avenues for treatment and the mechanisms underlying neuropathic pain. Immune infiltration analysis revealed which types of immune cells related to the NRDEGs signature played an important role in the occurrence and development of neuropathic pain. Basic experiments provided crucial evidence that the 3 NRDEGs in DRG served as important regulators of neuropathic pain. Conclusion The prediction signature based on 3 key NRDEGs showed promise in predicting the presence of neuropathic pain, which may open up new avenues for the development of novel therapies for neuropathic pain.
Collapse
Affiliation(s)
- Yang Liu
- Department of Pathology, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Shikang Hao
- The First Clinical Medical School, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Hongyu Hao
- Department of Neurology, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Guona Zheng
- Department of Pathology, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Jie Bing
- Department of Pathology, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Lin Kang
- Department of Pathology, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Jia Li
- Outpatient Department, Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Huanfen Zhao
- Department of Pathology, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Han Hao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Center of Innovative Drug Research and Evaluation, Hebei Medical University, Shijiazhuang, People’s Republic of China
| |
Collapse
|
6
|
Tang W, Li X, Liu H, Xu C, Deng S. The role of macrophages in chronic pain. Cytokine 2025; 185:156813. [PMID: 39577336 DOI: 10.1016/j.cyto.2024.156813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024]
Abstract
Chronic pain typically lasts or recurs for more than three months and is an unpleasant sensory and emotional experience, including neuropathic pain, long-term tissue damage, tumors, and viral or bacterial infections.The unpleasantness associated with pain affects the basic life of patients and has become a truly global problem. Macrophages, a powerful immune effector cell whose functional plasticity leads to polarization into different subtypes and opposite effects in different environments, are also indispensable in the development of pain.In recent years, there has been an increasing number of studies on the effects of macrophages on pain, and there are multiple pathways that regulate macrophage polarization, including lipopolysaccharide induction and IL-4/IL-13 stimulation.In addition, pathways involving macrophages and macrophage polarization have been found to have an exacerbating or mitigating role in the progression of chronic pain, with M1 macrophages generally exacerbating pain progression and M2 macrophages mitigating pain progression.Therefore, modulating macrophage polarization holds great promise as an intervention in chronic pain. In this paper, we synthesize multiple macrophage pathways as well as mechanisms affecting their pain processes in the context of different types of chronic pain, providing new avenues for chronic pain relief.
Collapse
Affiliation(s)
- Weikang Tang
- School of Medicine, Tarim University, Alaer, 843300 Xinjiang, China
| | - Xuan Li
- School of Medicine, Tarim University, Alaer, 843300 Xinjiang, China
| | - Huixia Liu
- School of Medicine, Tarim University, Alaer, 843300 Xinjiang, China
| | - Chunyan Xu
- School of Medicine, Tarim University, Alaer, 843300 Xinjiang, China
| | - Siyao Deng
- School of Medicine, Tarim University, Alaer, 843300 Xinjiang, China..
| |
Collapse
|
7
|
Wang H, Wang X, Wang L, Wang H, Zhang Y. Plant‐Derived Phytochemicals and Their Nanoformulations for Inducing Programed Cell Death in Cancer. ADVANCED THERAPEUTICS 2024; 7. [DOI: 10.1002/adtp.202400197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Indexed: 01/05/2025]
Abstract
AbstractPhytochemicals are a diverse class of compounds found in various plant‐based foods and beverages that have displayed the capacity to exert powerful anticancer effects through the induction of programed cell death (PCD) in malignancies. PCD is a sophisticated process that maintains in upholding tissue homeostasis and eliminating injured or neoplastic cells. Phytochemicals have shown the potential to induce PCD in malignant cells through various mechanisms, including modulation of cell signaling pathways, regulation of reactive oxygen species (ROS), and interaction with critical targets in cells such as DNA. Moreover, recent studies have suggested that nanomaterials loaded with phytochemicals may enhance cell death in tumors, which can also stimulate antitumor immunity. In this review, a comprehensive overview of the current understanding of the anticancer effects of phytochemicals and their potential as a promising approach to cancer therapy, is provided. The impacts of phytochemicals such as resveratrol, curcumin, apigenin, quercetin, and some approved plant‐derived drugs, such as taxanes on the regulation of some types of PCD, including apoptosis, pyroptosis, anoikis, autophagic cell death, ferroptosis, and necroptosis, are discussed. The underlying mechanisms and the potential of nanomaterials loaded with phytochemicals to enhance PCD in tumors are also explained.
Collapse
Affiliation(s)
- Haoyu Wang
- Medical College Xijing University Xi'an Shaanxi 710123 China
- Department of Orthopedics The Second Affiliated Hospital Xi'an Jiaotong University Xi'an Shaanxi 710004 China
| | - Xiaoyang Wang
- Department of Orthopedics The Second Affiliated Hospital Xi'an Jiaotong University Xi'an Shaanxi 710004 China
| | - Long Wang
- Medical College Xijing University Xi'an Shaanxi 710123 China
| | - Haifan Wang
- Department of Orthopedics The Second Affiliated Hospital Xi'an Jiaotong University Xi'an Shaanxi 710004 China
| | - Yuxing Zhang
- Medical College Xijing University Xi'an Shaanxi 710123 China
| |
Collapse
|
8
|
Ege E, Briggi D, Vu P, Cheng J, Lin F, Xu J. Targeting dorsal root ganglia for chemotherapy-induced peripheral neuropathy: from bench to bedside. Ther Adv Neurol Disord 2024; 17:17562864241252718. [PMID: 39318973 PMCID: PMC11421407 DOI: 10.1177/17562864241252718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a debilitating condition affecting an increasing number of cancer survivors worldwide. However, insights into its pathophysiology and availability of effective therapies remain lacking. Dorsal root ganglia (DRG) have been studied as a key component of chemotherapeutic drug toxicity and a potential therapeutic target for CIPN treatment. This comprehensive review aims to synthesize, summarize, and correlate the results of both preclinical and clinical studies relevant to the pathophysiology and management of CIPN in relation to the DRG. Design: Review. A thorough literature search was conducted using the terms 'dorsal root ganglion' and 'chemotherapy-induced peripheral neuropathy', along with appropriate variations. Searched databases included PubMed, EMBASE, Medline, Cochrane Library, Wiley Library, and Web of Science. Inclusion criteria targeted all English language, peer-reviewed original research from the inception of these databases to the present year. Review articles, book chapters, and other nonoriginal publications were excluded. Of 134 relevant studies identified, the majority were preclinical studies elucidating how various chemotherapeutic agents, especially taxanes, disrupt neurotransmission, inflammatory processes, and apoptotic pathways within sensory neurons of DRG. Not only do these effects correlate with the presentation of CIPN, but their disruption has also been shown to reduce CIPN symptoms in preclinical models. However, clinical studies addressing DRG interventions are very limited in number and scope at this time. These results reveal various pathways within DRG that may be effective targets for CIPN treatment. While limited, clinical studies do offer promise in the utility of DRG neuromodulation in managing painful CIPN. In the future, clinical trials are needed to assess interventions aimed at these neuronal and nonneuronal pathological targets to better treat this complex condition.
Collapse
Affiliation(s)
- Eliana Ege
- Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| | - Daniel Briggi
- Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| | - Peter Vu
- Department of Physical Medicine and Rehabilitation, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jianguo Cheng
- Department of Pain Management, Cleveland Clinic, Cleveland, OH, USA
- Department of Neuroscience, Cleveland Clinic, Cleveland, OH, USA
| | - Feng Lin
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, USA
| | - Jijun Xu
- Department of Pain Management and Inflammation and Immunity, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
9
|
Ollodart J, Steele LR, Romero-Sandoval EA, Strowd RE, Shiozawa Y. Contributions of neuroimmune interactions to chemotherapy-induced peripheral neuropathy development and its prevention/therapy. Biochem Pharmacol 2024; 222:116070. [PMID: 38387528 PMCID: PMC10964384 DOI: 10.1016/j.bcp.2024.116070] [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: 09/06/2023] [Revised: 12/04/2023] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a debilitating sequela that is difficult for both clinicians and cancer patients to manage. Precise mechanisms of CIPN remain elusive and current clinically prescribed therapies for CIPN have limited efficacy. Recent studies have begun investigating the interactions between the peripheral and central nervous systems and the immune system. Understanding these neuroimmune interactions may shift the paradigm of elucidating CIPN mechanisms. Although the contribution of immune cells to CIPN pathogenesis represents a promising area of research, its fully defined mechanisms have not yet been established. Therefore, in this review, we will discuss (i) current shortcoming of CIPN treatments, (ii) the roles of neuroimmune interactions in CIPN development and (iii) potential neuroimmune interaction-targeting treatment strategies for CIPN. Interestingly, monocytes/macrophages in dorsal root ganglia; microglia and astrocytes in spinal cord; mast cells in skin; and Schwann cell near peripheral nerves have been identified as inducers of CIPN behaviors, whereas T cells have been found to contribute to CIPN resolution. Additionally, nerve-resident immune cells have been targeted as prevention and/or therapy for CIPN using traditional herbal medicines, small molecule inhibitors, and intravenous immunoglobulins in a preclinical setting. Overall, unveiling neuroimmune interactions associated with CIPN may ultimately reduce cancer mortality and improve cancer patients' quality of life.
Collapse
Affiliation(s)
- Jenna Ollodart
- Department of Cancer Biology, Wake Forest University School of Medicine, and Atrium Health Wake Forest Baptist Comprehensive Cancer, Winston-Salem, NC, USA
| | - Laiton R Steele
- Department of Cancer Biology, Wake Forest University School of Medicine, and Atrium Health Wake Forest Baptist Comprehensive Cancer, Winston-Salem, NC, USA
| | | | - Roy E Strowd
- Department of Cancer Biology, Wake Forest University School of Medicine, and Atrium Health Wake Forest Baptist Comprehensive Cancer, Winston-Salem, NC, USA
| | - Yusuke Shiozawa
- Department of Cancer Biology, Wake Forest University School of Medicine, and Atrium Health Wake Forest Baptist Comprehensive Cancer, Winston-Salem, NC, USA.
| |
Collapse
|
10
|
Wang L, Zhang Y, Huang M, Yuan Y, Liu X. RIP3 in Necroptosis: Underlying Contributions to Traumatic Brain Injury. Neurochem Res 2024; 49:245-257. [PMID: 37743445 DOI: 10.1007/s11064-023-04038-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/07/2023] [Accepted: 09/20/2023] [Indexed: 09/26/2023]
Abstract
Traumatic brain injury (TBI) is a global public safety issue that poses a threat to death, characterized by high fatality rates, severe injuries and low recovery rates. There is growing evidence that necroptosis regulates the pathophysiological processes of a variety of diseases, particularly those affecting the central nervous system. Thus, moderate necroptosis inhibition may be helpful in the management of TBI. Receptor-interacting protein kinase (RIP) 3 is a key mediator in the necroptosis, and its absence helps restore the microenvironment at the injured site and improve cognitive impairment after TBI. In this report, we review different domains of RIP3, multiple analyses of necroptosis, and associations between necroptosis and TBI, RIP3, RIP1, and mixed lineage kinase domain-like. Next, we elucidate the potential involvement of RIP3 in TBI and highlight how RIP3 deficiency enhances neuronal function.
Collapse
Affiliation(s)
- Lvxia Wang
- School of Life and Environmental Sciences, Shaoxing University, Zhejiang, China
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China
| | - Yong Zhang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China
| | - Min Huang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China
| | - Yiling Yuan
- Department of Biosciences, Durham University, Durham, UK
| | - Xuehong Liu
- School of Life and Environmental Sciences, Shaoxing University, Zhejiang, China.
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China.
| |
Collapse
|
11
|
Basu A, Yang JY, Tsirukis VE, Loiacono A, Koch G, Khwaja IA, Krishnamurthy M, Fazio N, White E, Jha A, Shah S, Takmil C, Bagdas D, Demirer A, Master A, Natke E, Honkanen R, Huang L, Rigas B. Phosphosulindac (OXT-328) prevents and reverses chemotherapy induced peripheral neuropathy in mice. Front Neurosci 2024; 17:1240372. [PMID: 38347876 PMCID: PMC10860339 DOI: 10.3389/fnins.2023.1240372] [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: 06/14/2023] [Accepted: 10/19/2023] [Indexed: 02/15/2024] Open
Abstract
Background Chemotherapy-induced peripheral neuropathy (CIPN), a side effect of chemotherapy, is particularly difficult to treat. We explored whether phosphosulindac (PS), a modified NSAID, could treat CIPN. Methods CIPN was induced in male C57BL/6 J mice by paclitaxel, vincristine or oxaliplatin. Mechanical allodynia was measured with the von Frey test and cold allodynia with the acetone test. To determine the preventive effect of PS, it was administered 2 days before the induction of CIPN. Mouse Lewis lung carcinoma xenografts were used to determine if PS altered the chemotherapeutic efficacy of paclitaxel. Cultured cell lines were used to evaluate the effect of PS on neuroinflammation. Results Treatment with each of the three chemotherapeutic agents used to induce CIPN lowered the mechanical allodynia scores by 56 to 85% depending on the specific agent. PS gel was applied topically 3x/day for 16-22 days to the hind paws of mice with CIPN. This effect was dose-dependent. Unlike vehicle, PS returned mechanical allodynia scores back to pre-CIPN levels. PS had a similar effect on paclitaxel-induced CIPN cold allodynia. Sulindac, a metabolite of PS, had no effect on CIPN. PS significantly prevented CIPN compared to vehicle. Given concomitantly with paclitaxel to mice with lung cancer xenografts, PS relieved CIPN without affecting the anticancer effect of paclitaxel. The enantiomers of PS were equally efficacious against CIPN, suggesting the therapeutic suitability of the racemate PS. There were no apparent side effects of PS. PS suppressed the levels of IL-6, IL-10, CXCL1, and CXCL2 induced by paclitaxel in a neuroblastoma cell line, and macrophage activation to the M1 proinflammatory phenotype. Conclusion Topically applied PS demonstrated broad therapeutic and preventive efficacy against CIPN, preserved the anticancer effect of paclitaxel, and was safe. Its anti-CIPN effect appears to be mediated, in part, by suppression of neuroinflammation. These data support further evaluation of topical PS for the control of CIPN.
Collapse
Affiliation(s)
- Aryah Basu
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Jennifer Y. Yang
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Vasiliki E. Tsirukis
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Andrew Loiacono
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Gina Koch
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Ishan A. Khwaja
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Mahila Krishnamurthy
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Nicholas Fazio
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Emily White
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Aayushi Jha
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Shrila Shah
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Cameron Takmil
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Deniz Bagdas
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States
| | - Aylin Demirer
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, United States
| | - Adam Master
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Ernest Natke
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Robert Honkanen
- Departments of Ophthalmology, Stony Brook University, Stony Brook, NY, United States
| | - Liqun Huang
- Medicon Pharmaceuticals, Inc, Setauket, NY, United States
| | - Basil Rigas
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
12
|
Li L, Li T, Qu X, Sun G, Fu Q, Han G. Stress/cell death pathways, neuroinflammation, and neuropathic pain. Immunol Rev 2024; 321:33-51. [PMID: 37688390 DOI: 10.1111/imr.13275] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/14/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023]
Abstract
Neuropathic pain is a common and debilitating modality of chronic pain induced by a lesion or disease of the somatosensory nervous system. Albeit the elucidation of numerous pathophysiological mechanisms and the development of potential treatment compounds, safe and reliable therapies of neuropathic pain remain poor. Multiple stress/cell death pathways have been shown to be implicated in neuroinflammation during neuropathic pain. Here, we summarize the current knowledge of stress/cell death pathways and present an overview of the roles and molecular mechanisms of stress/cell death pathways in neuroinflammation during neuropathic pain, covering intrinsic and extrinsic apoptosis, autophagy, mitophagy, ferroptosis, pyroptosis, necroptosis, and phagoptosis. Small molecule compounds that modulate stress/cell death pathways in alleviating neuropathic pain are discussed mainly based on preclinical neuropathic pain models. These findings will contribute to in-depth understanding of the pathological processes during neuropathic pain as well as bridge the gap between basic and translational research to uncover new neuroprotective interventions.
Collapse
Affiliation(s)
- Lu Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xinyu Qu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Guangwei Sun
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qi Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Guang Han
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
13
|
Bacalhau C, Costa-Pereira JT, Tavares I. Preclinical research in paclitaxel-induced neuropathic pain: a systematic review. Front Vet Sci 2023; 10:1264668. [PMID: 38188718 PMCID: PMC10766764 DOI: 10.3389/fvets.2023.1264668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/21/2023] [Indexed: 01/09/2024] Open
Abstract
Introduction Chemotherapy-induced peripheral neuropathy (CIPN) is a common consequence of cancer treatment and pain is a frequent complaint of the patients. Paclitaxel, a cytostatic drug, generates a well-described peripheral nerve injury and neuroinflammation, which may be experimentally mimicked in animal models. We conducted a systematic review analyzing the experimental design, reporting and mechanisms underlying paclitaxel-induced neuropathy in the included studies to establish the perspectives of translation of the current literature in models of CIPN. Methods We elected studies published in Pubmed and Scopus between 1 January 2018 and 3 December 2022. Results According to a defined mesh of keywords searched, and after applying exclusion and inclusion criteria, 70 original studies were included and analyzed in detail. Most studies used male Sprague-Dawley rats to induce paclitaxel-induced neuropathy, used low doses of paclitaxel, and the analyzed studies mainly focused at 14-28 days of CIPN. Mechanical nociceptive tests were preferred in the behavioral evaluation. The mechanisms under study were mainly neuroinflammation of peripheral nerves. The overall methodological quality was considered moderate, and the risk of bias was unclear. Discussion Despite the ample preclinical research in paclitaxel-induced neuropathy, this systematic review alerts to some flaws in the experimental design along with limitations in reporting, e.g., lack of representation of both sexes in experimental work and the lack of reporting of the ARRIVE guidelines. This may limit the reproducibility of preclinical studies in CIPN. In addition, the clinical features of CIPN should be considered when designing animal experiments, such as sex and age of the CIPN patients. In this way the experimental studies aiming to establish the mechanisms of CIPN may allow the development of new drugs to treat CIPN and translation in the research of CIPN could be improved.
Collapse
Affiliation(s)
- Carolina Bacalhau
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - José Tiago Costa-Pereira
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Porto, Portugal
- I3S-Institute of Investigation and Innovation in Health, University of Porto, Porto, Portugal
- Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal
| | - Isaura Tavares
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Porto, Portugal
- I3S-Institute of Investigation and Innovation in Health, University of Porto, Porto, Portugal
| |
Collapse
|
14
|
Duan Y, Li Q, Zhou Y, Chen S, Li Y, Zang Y. Activation of the TNF-α-Necroptosis Pathway in Parvalbumin-Expressing Interneurons of the Anterior Cingulate Cortex Contributes to Neuropathic Pain. Int J Mol Sci 2023; 24:15454. [PMID: 37895135 PMCID: PMC10607712 DOI: 10.3390/ijms242015454] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
The hyperexcitability of the anterior cingulate cortex (ACC) has been implicated in the development of chronic pain. As one of the key causes of ACC hyperexcitation, disinhibition of the ACC may be closely related to the dysfunction of inhibitory parvalbumin (PV)-expressing interneurons (PV-INs). However, the molecular mechanism underlying the ACC PV-INs injury remains unclear. The present study demonstrates that spared sciatic nerve injury (SNI) induces an imbalance in the excitation and inhibition (E/I) of the ACC. To test whether tumor necrosis factor-α (TNF-α) upregulation in the ACC after SNI activates necroptosis and participates in PV-INs damage, we performed a differential analysis of transcriptome sequencing using data from neuropathic pain models and found that the expression of genes key to the TNF-α-necroptosis pathway were upregulated. TNF-α immunoreactivity (IR) signals in the ACCs of SNI rats were co-located with p-RIP3- and PV-IR, or p-MLKL- and PV-IR signals. We then systematically detected the expression and cell localization of necroptosis-related proteins, including kinase RIP1, RIP3, MLKL, and their phosphorylated states, in the ACC of SNI rats. Except for RIP1 and MLKL, the levels of these proteins were significantly elevated in the contralateral ACC and mainly expressed in PV-INs. Blocking the ACC TNF-α-necroptosis pathway by microinjecting TNF-α neutralizing antibody or using an siRNA knockdown to block expression of MLKL in the ACC alleviated SNI-induced pain hypersensitivity and inhibited the upregulation of TNF-α and p-MLKL. Targeting TNF-α-triggered necroptosis within ACC PV-INs may help to correct PV-INs injury and E/I imbalance in the ACC in neuropathic pain.
Collapse
Affiliation(s)
- Yiwen Duan
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Pain Research Center, Department of Physiology, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou 510080, China; (Y.D.); (Q.L.); (Y.Z.); (Y.L.)
| | - Qiaoyun Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Pain Research Center, Department of Physiology, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou 510080, China; (Y.D.); (Q.L.); (Y.Z.); (Y.L.)
| | - Yaohui Zhou
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Pain Research Center, Department of Physiology, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou 510080, China; (Y.D.); (Q.L.); (Y.Z.); (Y.L.)
| | - Shaoxia Chen
- State Key Laboratory of Oncology in South China, Department of Anesthesiology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China;
| | - Yongyong Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Pain Research Center, Department of Physiology, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou 510080, China; (Y.D.); (Q.L.); (Y.Z.); (Y.L.)
| | - Ying Zang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Pain Research Center, Department of Physiology, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou 510080, China; (Y.D.); (Q.L.); (Y.Z.); (Y.L.)
| |
Collapse
|
15
|
Zhu M, Huang H. The Underlying Mechanisms of Sleep Deprivation Exacerbating Neuropathic Pain. Nat Sci Sleep 2023; 15:579-591. [PMID: 37533626 PMCID: PMC10392808 DOI: 10.2147/nss.s414174] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023] Open
Abstract
Pain disrupts sleep, and sleep deprivation or interference can alter pain perception in animals and humans, for example by increasing sensitivity to pain. However, the mechanism by which sleep affects neuropathic pain remains unclear. In this review, we discuss the available evidence from the epidemiologic, clinical, and human, as well as laboratory studies. In previous studies, we have found that sleep deprivation affects various injurious systems, including opioids, dopaminergic, immune, orexins, hypothalamic-pituitary-adrenal axis, and adenosine. At the same time, these systems play a crucial role in neuropathic pain regulation. In the complex interactions between these neurobiological systems, there may be potential regulatory pathways through which sleep deprivation amplifies neuropathic pain. Because of the impact sleep problems and neuropathic pain can have on the patients' quality of life, studying the link between sleep and neuropathic pain is important for neuropathic pain prevention and public health.
Collapse
Affiliation(s)
- Manmin Zhu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Guizhou, People’s Republic of China
| | - Hao Huang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Guizhou, People’s Republic of China
| |
Collapse
|