1
|
Leng SZ, Fang MJ, Wang YM, Lin ZJ, Li QY, Xu YN, Mai CL, Wan JY, Yu Y, Wei M, Li Y, Zheng YF, Zhang KL, Wang YJ, Zhou LJ, Tan Z, Zhang H. Elevated plasma CXCL12 leads to pain chronicity via positive feedback upregulation of CXCL12/CXCR4 axis in pain synapses. J Headache Pain 2024; 25:213. [PMID: 39627724 PMCID: PMC11616163 DOI: 10.1186/s10194-024-01917-w] [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: 07/09/2024] [Accepted: 11/16/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Chronic pain poses a clinical challenge due to its associated costly disability and treatment needs. Determining how pain transitions from acute to chronic is crucial for effective management. Upregulation of the chemokine C-X-C motif ligand 12 (CXCL12) in nociceptive pathway is associated with chronic pain. Our previous study has reported that elevated plasma CXCL12 mediates intracerebral neuroinflammation and the comorbidity of cognitive impairment in neuropathic pain, but whether it is also involved in the pathogenesis of pathologic pain has not been investigated. METHODS Intravenous or intrathecal injection (i.v. or i.t.) of recombinant mouse CXCL12, neutralizing antibody (anti-CXCL12) or AMD3100 [an antagonist of its receptor C-X-C chemokine receptor type 4 (CXCR4)] was used to investigate the role of CXCL12 signaling pathway in pain chronicity. Two behavioral tests were used to examine pain changes. ELISA, immunofluorescence staining, Western blot, quantitative Real Time-PCR and Cytokine array were applied to detect the expressions of different molecules. RESULTS We found that increased plasma CXCL12 was positively correlated with pain severity in both chronic pain patients and neuropathic pain model in mice with spared nerve injury (SNI). Neutralizing plasma CXCL12 mitigated SNI-induced hyperalgesia. A single i.v. injection of CXCL12 induced prolonged mechanical hyperalgesia and activation of the nociceptive pathway. Multiple intravenous CXCL12 caused persistent hypersensitivity, enhanced structural plasticity of nociceptors and up-regulation of the CXCL12/CXCR4 axis in the dorsal root ganglion (DRG) and spinal dorsal horn (SDH). However, intrathecal blocking of CXCL12/CXCR4 pathway by CXCL12 antibody or CXCR4 antagonist AMD3100 significantly alleviated CXCL12-induced pain hypersensitivity and pathological changes. CONCLUSIONS Our study provides strong evidence that a sustained increase in plasma CXCL12 contributes to neuropathic pain through a positive feedback loop that enhances nociceptor plasticity, and suggests that targeting CXCL12/CXCR4 axis in plasma or nociceptive pathways has potential value in regulating pain chronicity.
Collapse
Affiliation(s)
- Shi-Ze Leng
- Department of Anesthesiology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Mei-Jia Fang
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Yi-Min Wang
- Department of Anesthesiology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Zhen-Jia Lin
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Qian-Yi Li
- Department of Anesthesiology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Ya-Nan Xu
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Chun-Lin Mai
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Jun-Ya Wan
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Yangyinhui Yu
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Ming Wei
- Department of Anesthesiology and Pain Clinic, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Ying Li
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Yu-Fan Zheng
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Kai-Lang Zhang
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Ya-Juan Wang
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Li-Jun Zhou
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China.
| | - Zhi Tan
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China.
| | - Hui Zhang
- Department of Anesthesiology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
| |
Collapse
|
2
|
Duan C, Zhu Y, Zhang Z, Wu T, Shen M, Xu J, Gao W, Pan J, Wei L, Su H, Shi C. Esketamine inhibits the c-Jun N-terminal kinase pathway in the spinal dorsal horn to relieve bone cancer pain in rats. Mol Pain 2024; 20:17448069241239231. [PMID: 38417838 PMCID: PMC10938627 DOI: 10.1177/17448069241239231] [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: 11/08/2023] [Revised: 02/04/2024] [Accepted: 02/25/2024] [Indexed: 03/01/2024] Open
Abstract
Cancer-induced bone pain (CIBP) is one of the most common and feared symptoms in patients with advanced tumors. The X-C motif chemokine ligand 12 (CXCL12) and the CXCR4 receptor have been associated with glial cell activation in bone cancer pain. Moreover, mitogen-activated protein kinases (MAPKs), as downstream CXCL12/CXCR4 signals, and c-Jun, as activator protein AP-1 components, contribute to the development of various types of pain. However, the specific CIBP mechanisms remain unknown. Esketamine is a non-selective N-methyl-d-aspartic acid receptor (NMDA) inhibitor commonly used as an analgesic in the clinic, but its analgesic mechanism in bone cancer pain remains unclear. We used a tumor cell implantation (TCI) model and explored that CXCL12/CXCR4, p-MAPKs, and p-c-Jun were stably up-regulated in the spinal cord. Immunofluorescence images showed activated microglia in the spinal cord on day 14 after TCI and co-expression of CXCL12/CXCR4, p-MAPKs (p-JNK, p-ERK, p-p38 MAPK), and p-c-Jun in microglia. Intrathecal injection of the CXCR4 inhibitor AMD3100 reduced JNK and c-Jun phosphorylations, and intrathecal injection of the JNK inhibitor SP600125 and esketamine also alleviated TCI-induced pain and reduced the expression of p-JNK and p-c-Jun in microglia. Overall, our data suggest that the CXCL12/CXCR4-JNK-c-Jun signaling pathway of microglia in the spinal cord mediates neuronal sensitization and pain hypersensitivity in cancer-induced bone pain and that esketamine exerts its analgesic effect by inhibiting the JNK-c-Jun pathway.
Collapse
Affiliation(s)
- Chenxia Duan
- Department of Anesthesiology, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yi Zhu
- Department of Anesthesiology, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Zhuoliang Zhang
- Department of Anesthesiology, Suzhou Municipal Hospital, Xuzhou Medical University, Suzhou, China
| | - Tiantian Wu
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Mengwei Shen
- Department of Anesthesiology, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jinfu Xu
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Wenxin Gao
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Jianhua Pan
- Department of Anesthesiology, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Lei Wei
- Department of Anesthesiology, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Huibin Su
- Department of Anesthesiology, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Chenghuan Shi
- Department of Anesthesiology, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| |
Collapse
|
3
|
Hu Y, Liu J, Zhuang R, Zhang C, Lin F, Wang J, Peng S, Zhang W. Progress in Pathological and Therapeutic Research of HIV-Related Neuropathic Pain. Cell Mol Neurobiol 2023; 43:3343-3373. [PMID: 37470889 PMCID: PMC11410024 DOI: 10.1007/s10571-023-01389-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023]
Abstract
HIV-related neuropathic pain (HRNP) is a neurodegeneration that gradually develops during the long-term course of acquired immune deficiency syndrome (AIDS) and manifests as abnormal sock/sleeve-like symmetrical pain and nociceptive hyperalgesia in the extremities, which seriously reduces patient quality of life. To date, the pathogenesis of HRNP is not completely clear. There is a lack of effective clinical treatment for HRNP and it is becoming a challenge and hot spot for medical research. In this study, we conducted a systematic review of the progress of HRNP research in recent years including (1) the etiology, classification and clinical symptoms of HRNP, (2) the establishment of HRNP pathological models, (3) the pathological mechanisms underlying HRNP from three aspects: molecules, signaling pathways and cells, (4) the therapeutic strategies for HRNP, and (5) the limitations of recent HRNP research and the future research directions and prospects of HRNP. This detailed review provides new and systematic insight into the pathological mechanism of HRNP, which establishes a theoretical basis for the future exploitation of novel target drugs. HIV infection, antiretroviral therapy and opioid abuse contribute to the etiology of HRNP with symmetrical pain in both hands and feet, allodynia and hyperalgesia. The pathogenesis involves changes in cytokine expression, activation of signaling pathways and neuronal cell states. The therapy for HRNP should be patient-centered, integrating pharmacologic and nonpharmacologic treatments into multimodal intervention.
Collapse
Affiliation(s)
- YanLing Hu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - JinHong Liu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Renjie Zhuang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Chen Zhang
- Department of Biological Sciences, University of Denver, Denver, CO, 80210, USA
| | - Fei Lin
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Jun Wang
- Department of Orthopedics, Rongjun Hospital, Jiaxing, Zhejiang, China
| | - Sha Peng
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Wenping Zhang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
Jonart LM, Ostergaard J, Brooks A, Fitzpatrick G, Chen L, Gordon PM. CXCR4 antagonists disrupt leukaemia-meningeal cell adhesion and attenuate chemoresistance. Br J Haematol 2023; 201:459-469. [PMID: 36535585 PMCID: PMC10121760 DOI: 10.1111/bjh.18607] [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: 09/19/2022] [Revised: 11/28/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
The effective prophylaxis and treatment of central nervous system (CNS) involvement in acute lymphoblastic leukaemia (ALL) remains a significant clinical challenge. Developing novel and more effective CNS-directed therapies has been hampered, in part, by our limited understanding of the leukaemia niche in the CNS relative to the bone marrow. Accordingly, defining the molecular and cellular components critical for the establishment and maintenance of the CNS leukaemia niche may lead to new therapeutic opportunities. In prior work we showed that direct intercellular interactions between leukaemia and meningeal cells enhance leukaemia chemoresistance in the CNS. Herein, we show that the CXCR4/CXCL12 chemokine axis contributes to leukaemia-meningeal cell adhesion. Importantly, clinically tested CXCR4 antagonists, which are likely to cross the blood-brain and blood-cerebral spinal fluid barriers and penetrate the CNS, effectively disrupted leukaemia-meningeal cell adhesion. Moreover, by disrupting these intercellular interactions, CXCR4 antagonists attenuated leukaemia chemoresistance in leukaemia-meningeal cell co-culture experiments and enhanced the efficacy of cytarabine in targeting leukaemia cells in the meninges in vivo. This work identifies the CXCR4/CXCL12 axis as an important regulator of intercellular interactions within the CNS leukaemia niche and supports further testing of the therapeutic efficacy of CXCR4 antagonists in overcoming CNS niche-mediated chemoresistance.
Collapse
Affiliation(s)
- Leslie M Jonart
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jason Ostergaard
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Athena Brooks
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Garrett Fitzpatrick
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Liam Chen
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Peter M Gordon
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
5
|
Huang A, Ji L, Huang Y, Yu Q, Li Y. miR-185-5p alleviates CCI-induced neuropathic pain by repressing NLRP3 inflammasome through dual targeting MyD88 and CXCR4. Int Immunopharmacol 2022; 104:108508. [PMID: 34999395 DOI: 10.1016/j.intimp.2021.108508] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/28/2021] [Accepted: 12/28/2021] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) are important modulators in the evolvement and progression of neuropathic pain (NP). According to reports, miR-185-5p contributes to various diseases and inflammatory responses. However, it is not clear whether miR-185-5p mediates neuroinflammation and NP following chronic constrictive injury (CCI). The CCI model was constructed in rats to induce NP. Paw withdrawal threshold (PWT) and paw withdrawal latency (PWL) were employed to evaluate pain threshold in CCI rats. The expression of miR-185-5p, GFAP, Iba1, Caspase-3-positive cells, terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL)-labeled apoptotic neurons, inflammatory mediators, including interleukin (IL)-6, IL-1β and tumor necrosis factor-α (TNF-α) in lumbar portion (L4-L6) of CCI rats were determined. Furthermore, the targets of miR-185-5p were predicted by the Starbase, and the binding association between miR-185-5p and MyD88, miR-185-5p and CXCR4 was verified by the dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. As shown by the data, miR-185-5p was distinctly reduced in L4-L6 spinal cord tissues of rats after CCI. Up-regulating miR-185-5p alleviated mechanical and thermal hyperalgesia, inactivated microglia and astrocytes accumulation, and abated the contents of IL-1β, IL-6 and TNF-α in L4-L6 spinal cord tissues of CCI rats. Bioinformatics analysis suggested that MyD88 and CXCR4 were potential target genes of miR-185-5p. Increasing miR-185-5p expression notably impeded the expression of MyD88, CXCR4 and NLRP3 inflammasome in BV2 microglia, while attenuating miR-185-5p expression exerted the opposite effects. Notably, down-regulating MyD88 and CXCR4 significantly enhanced the miR-185-5p-mediated anti-inflammatory effects, and reversed miR-185-5p inhibitor-mediated proinflammatory effects. Additionally, up-regulating miR-185-5p repressed BV2-induced neuronal apoptosis and increased neuronal viability. In conclusion, this study suggested that miR-185-5p chokes CCI-induced NP and neuroinflammation by targeting MyD88 and CXCR4, indicating that miR-186-5p is an underlying therapeutic target for NP.
Collapse
Affiliation(s)
- Airu Huang
- Department of Rehabilitation Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, PR China
| | - Ling Ji
- Department of Rehabilitation Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, PR China
| | - Yilong Huang
- Gastrointestinal surgery, Pidu District People's Hospital, Chengdu, Sichuan 611730, PR China
| | - Qian Yu
- Department of Rehabilitation Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, PR China.
| | - Yufeng Li
- Department of Rehabilitation Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, PR China.
| |
Collapse
|
6
|
Upregulating miR-130a-5p relieves astrocyte over activation-induced neuropathic pain through targeting C-X-C motif chemokine receptor 12/C-X-C motif chemokine receptor 4 axis. Neuroreport 2021; 32:135-143. [PMID: 33395188 DOI: 10.1097/wnr.0000000000001573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVES This study intends to explore the role and specific mechanism of miR-130a-5p in neuropathic pain through regulating the C-X-C motif chemokine receptor 12 (CXCL12)-C-X-C motif chemokine receptor 4 (CXCR4) pathway. METHODS First, mouse neuropathic pain model was constructed by spinal nerve ligation. MiR-130a-5p mimics were used to upregulate miR-130a-5p in vivo. The behaviour and pain scores of the spinal cord injury (SCI) mice were assessed. In addition, astrocytic activation as well as inflammatory response in the spinal lesions was determined. RESULTS The results manifested miR-130a-5p was notably downregulated in neuropathic pain model and reached the lowest point at 3 days after injury. Besides, tail vein injection of miR-130a-5p mimics inhibited the activation and inflammatory response of astrocytes, thus alleviating chronic constriction injury-induced neuropathic pain. Moreover, miR-130a-5p inactivated CXCR4 and its downstream Rac1, nuclear factor-κB (NF-κB) and extracellular regulated protein kinases signalling pathways by attenuating CXCL12. CONCLUSION MiR-130a-5p inactivated astrocytes by targeting CXCL12/CXCR4, thus alleviating SCI-induced neuropathic pain.
Collapse
|
7
|
Kawabata A, Tsubota M, Sekiguchi F, Tsujita R. [HMGB1 as a target for prevention of chemotherapy-induced peripheral neuropathy]. Nihon Yakurigaku Zasshi 2019; 154:236-240. [PMID: 31735750 DOI: 10.1254/fpj.154.236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) considerably impairs cancer patients' QOL, and may lead to discontinuation of drug treatment of cancer. Currently, there is no effective strategy against CIPN. Therefore, it is an urgent issue to develop clinically available drugs that prevent or treat CIPN. We have shown that high mobility group box 1 (HMGB1), a damage-associated molecular pattern (DAMP) molecule, plays an essential role in the development of CIPN. Most interestingly, thrombomodulin α, approved as a medicine for treatment of disseminated intravascular coagulation (DIC) in Japan, causes thrombin-dependent degradation of extracellular HMGB1 that is released in response to chemotherapeutics, and prevents CIPN. Thus, we expect that targeting HMGB1 or its receptors would lead to prevention of CIPN in cancer patients in near future.
Collapse
Affiliation(s)
- Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University
| | - Maho Tsubota
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University
| | - Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University
| | - Ryuichi Tsujita
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University.,Asahi Kasei Pharma Corporation
| |
Collapse
|
8
|
Caroleo MC, Brizzi A, De Rosa M, Pandey A, Gallelli L, Badolato M, Carullo G, Cione E. Targeting Neuropathic Pain: Pathobiology, Current Treatment and Peptidomimetics as a New Therapeutic Opportunity. Curr Med Chem 2019; 27:1469-1500. [PMID: 31142248 DOI: 10.2174/0929867326666190530121133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 01/25/2019] [Accepted: 02/15/2019] [Indexed: 12/18/2022]
Abstract
There is a huge need for pharmaceutical agents for the treatment of chronic Neuropathic Pain (NP), a complex condition where patients can suffer from either hyperalgesia or allodynia originating from central or peripheral nerve injuries. To date, the therapeutic guidelines include the use of tricyclic antidepressants, serotonin-noradrenaline reuptake inhibitors and anticonvulsants, beside the use of natural compounds and non-pharmacological options. Unfortunately, these drugs suffer from limited efficacy and serious dose-dependent adverse effects. In the last decades, the heptapeptide SP1-7, the major bioactive metabolite produced by Substance P (SP) cleavage, has been extensively investigated as a potential target for the development of novel peptidomimetic molecules to treat NP. Although the physiological effects of this SP fragment have been studied in detail, the mechanism behind its action is not fully clarified and the target for SP1-7 has not been identified yet. Nevertheless, specific binding sites for the heptapeptide have been found in brain and spinal cord of both mouse and rats. Several Structure-Affinity Relationship (SAR) studies on SP1-7 and some of its synthetic analogues have been carried out aiming to developing more metabolically stable and effective small molecule SP1-7-related amides that could be used as research tools for a better understanding of the SP1-7 system and, in a longer perspective, as potential therapeutic agents for future treatment of NP.
Collapse
Affiliation(s)
- Maria Cristina Caroleo
- Department of Pharmacy and Health and Nutrition Sciences, University of Calabria, Edificio Polifunzionale, 87026 Rende (CS), Italy
| | - Antonella Brizzi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Polo Scientifico San Miniato, Via A. Moro 2, 53100 Siena, Italy
| | - Maria De Rosa
- Drug Discovery Unit, Ri.MED Foundation, Palermo 90133, Italy
| | - Ankur Pandey
- Department of Chemistry and Center of Advanced Studies in Chemistry, Punjab University, Chandigarh, India
| | - Luca Gallelli
- Department of Health Science, School of Medicine, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Mariateresa Badolato
- Department of Pharmacy and Health and Nutrition Sciences, University of Calabria, Edificio Polifunzionale, 87026 Rende (CS), Italy
| | - Gabriele Carullo
- Department of Pharmacy and Health and Nutrition Sciences, University of Calabria, Edificio Polifunzionale, 87026 Rende (CS), Italy
| | - Erika Cione
- Department of Pharmacy and Health and Nutrition Sciences, University of Calabria, Edificio Polifunzionale, 87026 Rende (CS), Italy
| |
Collapse
|
9
|
De Clercq E. Mozobil® (Plerixafor, AMD3100), 10 years after its approval by the US Food and Drug Administration. Antivir Chem Chemother 2019; 27:2040206619829382. [PMID: 30776910 PMCID: PMC6379795 DOI: 10.1177/2040206619829382] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AMD3100 (plerixafor, Mozobil®) was first identified as an anti-HIV agent
specifically active against the T4-lymphotropic HIV strains, as it selectively
blocked the CXCR4 receptor. Through interference with the interaction of CXCR4
with its natural ligand, SDF-1 (also named CXCL12), it also mobilized the
CD34+stem cells from the bone marrow into the peripheral blood
stream. In December 2008, AMD3100 was formally approved by the US FDA for
autologous transplantation in patients with Non-Hodgkin’s Lymphoma or multiple
myeloma. It may be beneficially used in various other malignant diseases as well
as hereditary immunological disorders such as WHIM syndrome, and
physiopathological processes such as hepatopulmonary syndrome.
Collapse
|
10
|
Guo X, Deng G, Liu J, Zou P, Du F, Liu F, Chen AT, Hu R, Li M, Zhang S, Tang Z, Han L, Liu J, Sheth KN, Chen Q, Gou X, Zhou J. Thrombin-Responsive, Brain-Targeting Nanoparticles for Improved Stroke Therapy. ACS NANO 2018; 12:8723-8732. [PMID: 30107729 DOI: 10.1021/acsnano.8b04787] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Current treatments for ischemic stroke are insufficient. The lack of effective pharmacological approaches can be mainly attributed to the difficulty in overcoming the blood-brain barrier. Here, we report a simple strategy to synthesize protease-responsive, brain-targeting nanoparticles for the improved treatment of stroke. The resulting nanoparticles respond to proteases enriched in the ischemic microenvironment, including thrombin or matrix metalloproteinase-9, by shrinking or expanding their size. Targeted delivery was achieved using surface conjugation of ligands that bind to proteins that were identified to enrich in the ischemic brain using protein arrays. By screening a variety of formulations, we found that AMD3100-conjugated, size-shrinkable nanoparticles (ASNPs) exhibited the greatest delivery efficiency. The brain targeting effect is mainly mediated by AMD3100, which interacts with CXCR4 that is enriched in the ischemic brain tissue. We showed that ASNPs significantly enhanced the efficacy of glyburide, a promising stroke therapeutic drug whose efficacy is limited by its toxicity. Due to their high efficiency in penetrating the ischemic brain and low toxicity, we anticipate that ASNPs have the potential to be translated into clinical applications for the improved treatment of stroke patients.
Collapse
Affiliation(s)
| | - Gang Deng
- Department of Neurosurgery , Renmin Hospital of Wuhan University , Wuhan , Hubei 430060 , China
| | | | | | | | | | | | | | | | - Shenqi Zhang
- Department of Neurosurgery , Renmin Hospital of Wuhan University , Wuhan , Hubei 430060 , China
| | - Zhishu Tang
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine , Xi'an Medical University , Xi'an , Shannxi 710021 , China
| | | | - Jie Liu
- Department of Biomedical Engineering, School of Engineering , Sun Yat-sen University , Guangzhou , Guangdong 510006 , China
| | | | - Qianxue Chen
- Department of Neurosurgery , Renmin Hospital of Wuhan University , Wuhan , Hubei 430060 , China
| | - Xingchun Gou
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine , Xi'an Medical University , Xi'an , Shannxi 710021 , China
| | | |
Collapse
|
11
|
Pan Z, Shan Q, Gu P, Wang XM, Tai LW, Sun M, Luo X, Sun L, Cheung CW. miRNA-23a/CXCR4 regulates neuropathic pain via directly targeting TXNIP/NLRP3 inflammasome axis. J Neuroinflammation 2018; 15:29. [PMID: 29386025 PMCID: PMC5791181 DOI: 10.1186/s12974-018-1073-0] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 01/19/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Chemokine CXC receptor 4 (CXCR4) in spinal glial cells has been implicated in neuropathic pain. However, the regulatory cascades of CXCR4 in neuropathic pain remain elusive. Here, we investigated the functional regulatory role of miRNAs in the pain process and its interplay with CXCR4 and its downstream signaling. METHODS miRNAs and CXCR4 and its downstream signaling molecules were measured in the spinal cords of mice with sciatic nerve injury via partial sciatic nerve ligation (pSNL). Immunoblotting, immunofluorescence, immunoprecipitation, and mammal two-hybrid and behavioral tests were used to explore the downstream CXCR4-dependent signaling pathway. RESULTS CXCR4 expression increased in spinal glial cells of mice with pSNL-induced neuropathic pain. Blocking CXCR4 alleviated the pain behavior; contrarily, overexpressing CXCR4 induced pain hypersensitivity. MicroRNA-23a-3p (miR-23a) directly bounds to 3' UTR of CXCR4 mRNA. pSNL-induced neuropathic pain significantly reduced mRNA expression of miR-23a. Overexpression of miR-23a by intrathecal injection of miR-23a mimics or lentivirus reduced spinal CXCR4 and prevented pSNL-induced neuropathic pain. In contrast, knockdown of miR-23a by intrathecal injection of miR-23a inhibitor or lentivirus induced pain-like behavior, which was reduced by CXCR4 inhibition. Additionally, miR-23a knockdown or CXCR4 overexpression in naïve mice could increase the thioredoxin-interacting protein (TXNIP), which was associated with induction of NOD-like receptor protein 3 (NLRP3) inflammasome. Indeed, CXCR4 and TXNIP were co-expressed. The mammal two-hybrid assay revealed the direct interaction between CXCR4 and TXNIP, which was increased in the spinal cord of pSNL mice. In particular, inhibition of TXNIP reversed pain behavior elicited by pSNL, miR-23a knockdown, or CXCR4 overexpression. Moreover, miR-23a overexpression or CXCR4 knockdown inhibited the increase of TXNIP and NLRP3 inflammasome in pSNL mice. CONCLUSIONS miR-23a, by directly targeting CXCR4, regulates neuropathic pain via TXNIP/NLRP3 inflammasome axis in spinal glial cells. Epigenetic interventions against miR-23a, CXCR4, or TXNIP may potentially serve as novel therapeutic avenues in treating peripheral nerve injury-induced nociceptive hypersensitivity.
Collapse
Affiliation(s)
- Zhiqiang Pan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221002, China. .,Laboratory and Clinical Research Institute for Pain, Department of Anesthesiology, The University of Hong Kong, Hong Kong SAR, China. .,Department of Anaesthesiology, Queen Mary Hospital, The University of Hong Kong, Rm 424, 4/F, Block K, 102 Pokfulam, Hong Kong, China.
| | - Qun Shan
- Laboratory and Clinical Research Institute for Pain, Department of Anesthesiology, The University of Hong Kong, Hong Kong SAR, China.,School of Life Science, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, People's Republic of China
| | - Pan Gu
- Laboratory and Clinical Research Institute for Pain, Department of Anesthesiology, The University of Hong Kong, Hong Kong SAR, China
| | - Xiao Min Wang
- Laboratory and Clinical Research Institute for Pain, Department of Anesthesiology, The University of Hong Kong, Hong Kong SAR, China
| | - Lydia Wai Tai
- Laboratory and Clinical Research Institute for Pain, Department of Anesthesiology, The University of Hong Kong, Hong Kong SAR, China
| | - Menglan Sun
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221002, China
| | - Xin Luo
- Laboratory and Clinical Research Institute for Pain, Department of Anesthesiology, The University of Hong Kong, Hong Kong SAR, China
| | - Liting Sun
- Laboratory and Clinical Research Institute for Pain, Department of Anesthesiology, The University of Hong Kong, Hong Kong SAR, China
| | - Chi Wai Cheung
- Laboratory and Clinical Research Institute for Pain, Department of Anesthesiology, The University of Hong Kong, Hong Kong SAR, China. .,Research Centre of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, Hong Kong SAR, China. .,Department of Anaesthesiology, Queen Mary Hospital, The University of Hong Kong, Rm 424, 4/F, Block K, 102 Pokfulam, Hong Kong, China.
| |
Collapse
|