1
|
Zhang X, Jin T, Wang H, Han S, Liang Y. Microglia in morphine tolerance: cellular and molecular mechanisms and therapeutic potential. Front Pharmacol 2024; 15:1499799. [PMID: 39669194 PMCID: PMC11635611 DOI: 10.3389/fphar.2024.1499799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024] Open
Abstract
Morphine has a crucial role in treating both moderate to severe pain and chronic pain. However, prolonged administration of morphine can lead to tolerance of analgesia, resulting in increased doses and poor treatment of pain. Many patients, such as those with terminal cancer, require high doses of morphine for long periods. Addressing morphine tolerance can help this group of patients to escape pain, and the mechanisms behind this need to be investigated. Microglia are the key cells involved in morphine tolerance and chronic morphine administration leads to microglia activation, which in turn leads to activation of internal microglia signalling pathways and protein transcription, ultimately leading to the release of inflammatory factors. Inhibiting the activation of microglia internal signalling pathways can reduce morphine tolerance. However, the exact mechanism of how morphine acts on microglia and ultimately leads to tolerance is unknown. This article discusses the mechanisms of morphine induced microglia activation, reviews the signalling pathways within microglia and the associated therapeutic targets and possible drugs, and provides possible directions for clinical prevention or retardation of morphine induced analgesic tolerance.
Collapse
Affiliation(s)
- Xiangning Zhang
- Department of Anesthesiology, Women and Children’s Hospital, Peking University People’s Hospital, Qingdao University, Qingdao, Shandong, China
- Clinical Medical College, Qingdao University, Qingdao, Shandong, China
| | - Tingting Jin
- Department of Anesthesiology, Women and Children’s Hospital, Peking University People’s Hospital, Qingdao University, Qingdao, Shandong, China
- Clinical Medical College, Qingdao University, Qingdao, Shandong, China
| | - Haixia Wang
- Department of Anesthesiology, Women and Children’s Hospital, Peking University People’s Hospital, Qingdao University, Qingdao, Shandong, China
- Clinical Medical College, Qingdao University, Qingdao, Shandong, China
| | - Shuai Han
- Department of Anesthesiology, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yongxin Liang
- Department of Anesthesiology, Women and Children’s Hospital, Peking University People’s Hospital, Qingdao University, Qingdao, Shandong, China
- Clinical Medical College, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
2
|
Ou C, Zhang K, Mu Y, Huang Z, Li X, Huang W, Wang Y, Zeng W, Ouyang H. YTHDF1 in periaqueductal gray inhibitory neurons contributes to morphine withdrawal responses in mice. BMC Med 2024; 22:406. [PMID: 39304892 PMCID: PMC11416010 DOI: 10.1186/s12916-024-03634-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Physical symptoms and aversion induced by opioid withdrawal strongly affect the management of opioid addiction. YTH N6-methyladenosine (m6A) RNA binding protein 1 (YTHDF1), an m6A-binding protein, from the periaqueductal gray (PAG) reportedly contributes to morphine tolerance and hyperalgesia. However, the role of YTHDF1 in morphine withdrawal remains unclear. METHODS A naloxone-precipitated morphine withdrawal model was established in C57/BL6 mice or transgenic mice. YTHDF1 was knocked down via adeno-associated virus transfection. Combined with the results of the single-cell RNA sequencing analysis, the changes in morphine withdrawal somatic signs and conditioned place aversion (CPA) scores were compared when YTHDF1 originating from different neurons in the ventrolateral periaqueductal gray (vlPAG) was knocked down. We further explored the role of inflammatory factors and transcription factors related to inflammatory response in morphine withdrawal. RESULTS Our results revealed that YTHDF1 expression was upregulated in the vlPAG of mice with morphine withdrawal and that the knockdown of vlPAG YTHDF1 attenuated morphine withdrawal-related somatic signs and aversion. The levels of NF-κB and p-NF-κB were reduced after the inhibition of YTHDF1 in the vlPAG. YTHDF1 from vlPAG inhibitory neurons, rather than excitatory neurons, facilitated morphine withdrawal responses. The inhibition of YTHDF1 in vlPAG somatostatin (Sst)-expressing neurons relieved somatic signs of morphine withdrawal and aversion, whereas the knockdown of YTHDF1 in cholecystokinin (Cck)-expressing or parvalbumin (PV)-expressing neurons did not change morphine withdrawal-induced responses. The activity of c-fos + neurons, the intensity of the calcium signal, the density of dendritic spines, and the frequency of mIPSCs in the vlPAG, which were increased in mice with morphine withdrawal, were decreased with the inhibition of YTHDF1 from vlPAG inhibitory neurons or Sst-expressing neurons. Knockdown of NF-κB in Sst-expressing neurons also alleviated morphine withdrawal-induced responses. CONCLUSIONS YTHDF1 originating from Sst-expressing neurons in the vlPAG is crucial for the modulation of morphine withdrawal responses, and the underlying mechanism might be related to the regulation of the expression and phosphorylation of NF-κB.
Collapse
Affiliation(s)
- Chaopeng Ou
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Kun Zhang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yanyu Mu
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Zhenzhen Huang
- Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xile Li
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Wan Huang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yan Wang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Weian Zeng
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Handong Ouyang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
3
|
Ouyang H, Zhang J, Chi D, Zhang K, Huang Y, Huang J, Huang W, Bai X. The YTHDF1-TRAF6 pathway regulates the neuroinflammatory response and contributes to morphine tolerance and hyperalgesia in the periaqueductal gray. J Neuroinflammation 2022; 19:310. [PMID: 36550542 PMCID: PMC9784087 DOI: 10.1186/s12974-022-02672-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Long-term use of opioids such as morphine has negative side effects, such as morphine analgesic tolerance and morphine-induced hyperalgesia (MIH). These side effects limit the clinical use and analgesic efficacy of morphine. Elucidation of the mechanisms and identification of feasible and effective methods or treatment targets to solve this clinical phenomenon are important. Here, we discovered that YTHDF1 and TNF receptor-associated factor 6 (TRAF6) are crucial for morphine analgesic tolerance and MIH. The m6A reader YTHDF1 positively regulated the translation of TRAF6 mRNA, and chronic morphine treatments enhanced the m6A modification of TRAF6 mRNA. TRAF6 protein expression was drastically reduced by YTHDF1 knockdown, although TRAF6 mRNA levels were unaffected. By reducing inflammatory markers such as IL-1β, IL-6, TNF-α and NF-κB, targeted reduction of YTHDF1 or suppression of TRAF6 activity in ventrolateral periaqueductal gray (vlPAG) slows the development of morphine analgesic tolerance and MIH. Our findings provide new insights into the mechanism of morphine analgesic tolerance and MIH indicating that YTHDF1 regulates inflammatory factors such as IL-1β, IL-6, TNF-α and NF-κB by enhancing TRAF6 protein expression.
Collapse
Affiliation(s)
- Handong Ouyang
- grid.488530.20000 0004 1803 6191Department of Anesthesiology, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangzhou, China
| | - Jianxing Zhang
- grid.488530.20000 0004 1803 6191Department of Anesthesiology, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangzhou, China
| | - Dongmei Chi
- grid.488530.20000 0004 1803 6191Department of Anesthesiology, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangzhou, China
| | - Kun Zhang
- grid.488530.20000 0004 1803 6191Department of Anesthesiology, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangzhou, China
| | - Yongtian Huang
- grid.488530.20000 0004 1803 6191Department of Anesthesiology, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangzhou, China
| | - Jingxiu Huang
- grid.488530.20000 0004 1803 6191Department of Anesthesiology, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangzhou, China
| | - Wan Huang
- grid.488530.20000 0004 1803 6191Department of Anesthesiology, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangzhou, China
| | - Xiaohui Bai
- grid.488530.20000 0004 1803 6191Department of Anesthesiology, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangzhou, China ,grid.412536.70000 0004 1791 7851Department of Anesthesiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yangjiang Road West, Guangzhou, China
| |
Collapse
|
4
|
Chang CL, Cai Z, Hsu SYT. Gel-forming antagonist provides a lasting effect on CGRP-induced vasodilation. Front Pharmacol 2022; 13:1040951. [PMID: 36569288 PMCID: PMC9772450 DOI: 10.3389/fphar.2022.1040951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/14/2022] [Indexed: 12/14/2022] Open
Abstract
Migraine affects ∼15% of the adult population, and the standard treatment includes the use of triptans, ergotamines, and analgesics. Recently, CGRP and its receptor, the CLR/RAMP1 receptor complex, have been targeted for migraine treatment due to their critical roles in mediating migraine headaches. The effort has led to the approval of several anti-CGRP antibodies for chronic migraine treatment. However, many patients still suffer continuous struggles with migraine, perhaps due to the limited ability of anti-CGRP therapeutics to fully reduce CGRP levels or reach target cells. An alternative anti-CGRP strategy may help address the medical need of patients who do not respond to existing therapeutics. By serendipity, we have recently found that several chimeric adrenomedullin/adrenomedullin 2 peptides are potent CLR/RAMP receptor antagonists and self-assemble to form liquid gels. Among these analogs, the ADE651 analog, which potently inhibits CLR/RAMP1 receptor signaling, forms gels at a 6-20% level. Screening of ADE651 variants indicated that residues at the junctional region of this chimeric peptide are important for gaining the gel-forming capability. Gel-formation significantly slowed the passage of ADE651 molecules through Centricon filters. Consistently, subcutaneous injection of ADE651 gel in rats led to the sustained presence of ADE651 in circulation for >1 week. In addition, analysis of vascular blood flow in rat hindlimbs showed ADE651 significantly reduces CGRP-induced vasodilation. Because gel-forming antagonists could have direct and sustained access to target cells, ADE651 and related antagonists for CLR/RAMP receptors may represent promising candidates for targeting CGRP- and/or adrenomedullin-mediated headaches in migraine patients.
Collapse
Affiliation(s)
- Chia Lin Chang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Chang Gung University, Taoyuan, Taiwan
| | - Zheqing Cai
- CL Laboratory LLC, Gaithersburg, MD, United States
| | - Sheau Yu Teddy Hsu
- Adepthera LLC, San Jose, CA, United States,*Correspondence: Sheau Yu Teddy Hsu,
| |
Collapse
|
5
|
Wang H, Luo J, Chen X, Hu H, Li S, Zhang Y, Shi C. Clinical Observation of the Effects of Oral Opioid on Inflammatory Cytokines and Gut Microbiota in Patients with Moderate to Severe Cancer Pain: A Retrospective Cohort Study. Pain Ther 2022; 11:667-681. [PMID: 35435623 PMCID: PMC9098777 DOI: 10.1007/s40122-022-00386-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 03/31/2022] [Indexed: 10/31/2022] Open
Abstract
Introduction Methods Results Conclusion
Collapse
|
6
|
Rees T, Hendrikse E, Hay D, Walker C. Beyond CGRP: The calcitonin peptide family as targets for migraine and pain. Br J Pharmacol 2022; 179:381-399. [PMID: 34187083 PMCID: PMC9441195 DOI: 10.1111/bph.15605] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/20/2021] [Accepted: 06/10/2021] [Indexed: 02/03/2023] Open
Abstract
The CGRP system has emerged as a key pharmacological target for the treatment of migraine. However, some individuals who suffer from migraine have low or no response to anti-CGRP or other treatments, suggesting the need for additional clinical targets. CGRP belongs to the calcitonin family of peptides, which includes calcitonin, amylin, adrenomedullin and adrenomedullin 2. These peptides display a range of pro-nociceptive and anti-nociceptive actions, in primary headache conditions such as migraine. Calcitonin family peptides also show expression at sites relevant to migraine and pain. This suggests that calcitonin family peptides and their receptors, beyond CGRP, may be therapeutically useful in the treatment of migraine and other pain disorders. This review considers the localisation of the calcitonin family in peripheral pain pathways and discusses how they may contribute to migraine and pain. LINKED ARTICLES: This article is part of a themed issue on Advances in Migraine and Headache Therapy (BJP 75th Anniversary). To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.3/issuetoc.
Collapse
Affiliation(s)
- T.A. Rees
- School of Biological Science, University of Auckland, Auckland, NZ.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - E.R Hendrikse
- School of Biological Science, University of Auckland, Auckland, NZ
| | - D.L. Hay
- School of Biological Science, University of Auckland, Auckland, NZ.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.,Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand.,Corresponding author(s): Christopher S Walker, , Debbie L. Hay,
| | - C.S Walker
- School of Biological Science, University of Auckland, Auckland, NZ.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.,Corresponding author(s): Christopher S Walker, , Debbie L. Hay,
| |
Collapse
|
7
|
Morphine promotes microglial activation by upregulating the EGFR/ERK signaling pathway. PLoS One 2021; 16:e0256870. [PMID: 34520454 PMCID: PMC8439491 DOI: 10.1371/journal.pone.0256870] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/17/2021] [Indexed: 11/20/2022] Open
Abstract
Although they represent the cornerstone of analgesic therapy, opioids, such as morphine, are limited in efficacy by drug tolerance, hyperalgesia and other side effects. Activation of microglia and the consequent production of proinflammatory cytokines play a key pathogenic role in morphine tolerance, but the exact mechanisms are not well understood. This study aimed to investigate the regulatory mechanism of epidermal growth factor receptor (EGFR) on microglial activation induced by morphine in mouse microglial BV-2 cells. In this research, BV-2 cells were stimulated with morphine or pretreated with AG1478 (an inhibitor of EGFR). Expression levels of cluster of differentiation molecule 11b (CD11b), EGFR, and phospho-EGFR were detected by immunofluorescence staining. Cell signaling was assayed by Western blot. The migration ability of BV-2 cells was tested by Transwell assay. The production of interleukin-1beta (IL-1β) and tumor necrosis factor-alpha (TNF-α) in the cell supernatant was determined by ELISA. We observed that the expression of CD11b induced by morphine was increased in a dose- and time- dependent manner in BV-2 cells. Phosphorylation levels of EGFR and ERK1/2, migration of BV-2 cells, and production of IL-1β and TNFα were markedly enhanced by morphine treatment. The activation, migration, and production of proinflammatory cytokines in BV-2 cells were inhibited by blocking the EGFR signaling pathway with AG1478. The present study demonstrated that the EGFR/ERK signaling pathway may represent a novel pharmacological strategy to suppress morphine tolerance through attenuation of microglial activation.
Collapse
|
8
|
Tu H, Chu H, Guan S, Hao F, Xu N, Zhao Z, Liang Y. The role of the M1/M2 microglia in the process from cancer pain to morphine tolerance. Tissue Cell 2020; 68:101438. [PMID: 33220596 DOI: 10.1016/j.tice.2020.101438] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 12/29/2022]
Abstract
Cancer pain, especially bone cancer pain, is a pain state often caused by inflammation or dysfunctional nerves. Moreover, in the management of cancer pain, opioid especially morphine is widely used, however, it also brings severe side effects such as morphine tolerance to the patient (Deandrea et al., 2008). A growing body of literatures demonstrated that neuroinflammation is mediated by microglia. As the macrophages like immune cells, microglia play an important role in the pathogenesis of cancer pain and morphine tolerance. Microglia acquire different activation states to regulate the function of these cells. As to M1 phenotype, microglia release pro-inflammatory cytokines and neurotoxic molecules that promote inflammation and cytotoxic reactions. Conversely, when microglia represent M2 phenotypes secreting anti-inflammatory cytokines and nutrient factors that promote the function of repair, regeneration and restore homeostasis. A better understanding of microglia activation in cancer pain and morphine tolerance is crucial for the development of hypothesized neuroprotective drugs. Targeting microglia different polarization states by the inhibition of their deleterious pro-inflammatory neurotoxicity and/or enhancing their beneficial anti-inflammatory protective function seems to be an effective treatment for cancer pain and morphine tolerance.
Collapse
Affiliation(s)
- Houan Tu
- Department of Anesthesiology, Women's and Children's Hospital Affiliated to Qingdao University, 6 Tongfu Road, Qingdao, Shandong 266034, China
| | - Haichen Chu
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, 59 Hai Er Road, Qingdao, Shandong 266061, China
| | - Sen Guan
- Department of Anesthesiology, Women's and Children's Hospital Affiliated to Qingdao University, 6 Tongfu Road, Qingdao, Shandong 266034, China
| | - Fengxi Hao
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, 59 Hai Er Road, Qingdao, Shandong 266061, China
| | - Na Xu
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, 59 Hai Er Road, Qingdao, Shandong 266061, China
| | - Zhiping Zhao
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, 59 Hai Er Road, Qingdao, Shandong 266061, China
| | - Yongxin Liang
- Department of Anesthesiology, Women's and Children's Hospital Affiliated to Qingdao University, 6 Tongfu Road, Qingdao, Shandong 266034, China.
| |
Collapse
|
9
|
Beylerli OA, Azizova ST, Konovalov NA, Akhmedov AD, Gareev IF, Belogurov AA. [Non-coding RNAs as therapeutic targets in spinal cord injury]. ZHURNAL VOPROSY NEĬROKHIRURGII IMENI N. N. BURDENKO 2020; 84:104-110. [PMID: 32759933 DOI: 10.17116/neiro202084031104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Spinal cord injury (SCI) may be followed by persistent motor dysfunction and somatosensory disturbances that negatively influences the quality of life of patients and creates a significant economic burden. Analysis of secondary biological processes associated with changes in genetic expression is becoming increasingly important every day in understanding the pathophysiology of spinal cord injury. The results of international sequencing of the human genome were analyzed in 2004. These data revealed about 20,000 protein-coding genes covering near 2% of the total genomic sequence. The vast majority of gene transcripts are actually characterized as non-coding RNAs (ncRNAs). These RNA clusters do not encode functional proteins and ensure post-transcriptional regulation of gene expression. The clusters may be small (approximately 20 nucleotides) known as miRNAs or the transcripts can enroll over 200 nucleotides defined as long non-coding RNAs (lncRNAs). Some modern studies describe transient expression of microRNA in case of spinal cord injury. These RNAs are associated with inflammation and apoptosis, functional recovery and regeneration. Large-scale genomic analysis has demonstrated the existence of multiple lncRNAs whose expression is associated with some processes of spinal cord injury. lncRNA can be divided into two categories depending on the position in relation to the coding genes: intergenic and intragenic. Intergenic lncRNAs is currently the most studied class. Intragenic lncRNAs can be subdivided depending on the overlap of the coding genes (antisense, intron, etc.). According to recent studies, long non-coding RNAs are abundantly present in the tissues of central nervous system and may be crucial in the pathogenesis of certain diseases of nervous system. At the cellular level, it has been shown that lncRNAs regulate the expression of protein-coding RNAs. Moreover, these molecules are involved into such processes as neuronal death, demyelination and glia activation. This review is devoted to the role of ncRNAs in the pathogenesis of spinal cord injury and their potential use as targets for the treatment of consequences of spinal cord injury.
Collapse
Affiliation(s)
- O A Beylerli
- Bashkir State Medical University of the Ministry of Health of the Russian Federation, Ufa, Russia
| | - Sh T Azizova
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | | | - I F Gareev
- Bashkir State Medical University of the Ministry of Health of the Russian Federation, Ufa, Russia
| | - A A Belogurov
- Shemyakin-Ovcinnicov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
10
|
Du ER, Fan RP, Rong LL, Xie Z, Xu CS. Regulatory mechanisms and therapeutic potential of microglial inhibitors in neuropathic pain and morphine tolerance. J Zhejiang Univ Sci B 2020; 21:204-217. [PMID: 32133798 PMCID: PMC7086010 DOI: 10.1631/jzus.b1900425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/24/2019] [Indexed: 12/30/2022]
Abstract
Microglia are important cells involved in the regulation of neuropathic pain (NPP) and morphine tolerance. Information on their plasticity and polarity has been elucidated after determining their physiological structure, but there is still much to learn about the role of this type of cell in NPP and morphine tolerance. Microglia mediate multiple functions in health and disease by controlling damage in the central nervous system (CNS) and endogenous immune responses to disease. Microglial activation can result in altered opioid system activity, and NPP is characterized by resistance to morphine. Here we investigate the regulatory mechanisms of microglia and review the potential of microglial inhibitors for modulating NPP and morphine tolerance. Targeted inhibition of glial activation is a clinically promising approach to the treatment of NPP and the prevention of morphine tolerance. Finally, we suggest directions for future research on microglial inhibitors.
Collapse
Affiliation(s)
- Er-rong Du
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang 330006, China
| | - Rong-ping Fan
- Department of Fourth Clinical Medicine, School of Medicine, Nanchang University, Nanchang 330006, China
| | - Li-lou Rong
- Department of Fourth Clinical Medicine, School of Medicine, Nanchang University, Nanchang 330006, China
| | - Zhen Xie
- Department of First Clinical Medicine, School of Medicine, Nanchang University, Nanchang 330006, China
| | - Chang-shui Xu
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang 330006, China
- Key Laboratory of Autonomic Nervous Function and Disease of Jiangxi Province, Nanchang 330006, China
| |
Collapse
|
11
|
Abstract
Adrenomedullin, a peptide with multiple physiological functions in nervous system injury and disease, has aroused the interest of researchers. This review summarizes the role of adrenomedullin in neuropathological disorders, including pathological pain, brain injury and nerve regeneration, and their treatment. As a newly characterized pronociceptive mediator, adrenomedullin has been shown to act as an upstream factor in the transmission of noxious information for various types of pathological pain including acute and chronic inflammatory pain, cancer pain, neuropathic pain induced by spinal nerve injury and diabetic neuropathy. Initiation of glia-neuron signaling networks in the peripheral and central nervous system by adrenomedullin is involved in the formation and maintenance of morphine tolerance. Adrenomedullin has been shown to exert a facilitated or neuroprotective effect against brain injury including hemorrhagic or ischemic stroke and traumatic brain injury. Additionally, adrenomedullin can serve as a regulator to promote nerve regeneration in pathological conditions. Therefore, adrenomedullin is an important participant in nervous system diseases.
Collapse
Affiliation(s)
- Feng-Jiao Li
- College of Life Sciences, Laboratory of Neuroendocrinology, Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian Province, China
| | - Si-Ru Zheng
- College of Life Sciences, Laboratory of Neuroendocrinology, Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian Province, China
| | - Dong-Mei Wang
- College of Life Sciences, Laboratory of Neuroendocrinology, Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian Province, China
| |
Collapse
|
12
|
Chang CL, Hsu SYT. Development of chimeric and bifunctional antagonists for CLR/RAMP receptors. PLoS One 2019; 14:e0216996. [PMID: 31150417 PMCID: PMC6544337 DOI: 10.1371/journal.pone.0216996] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 05/02/2019] [Indexed: 11/26/2022] Open
Abstract
CGRP, adrenomedullin (ADM), and adrenomedullin 2 (ADM2) family peptides are
important neuropeptides and hormones for the regulation of neurotransmission,
vasotone, cardiovascular morphogenesis, vascular integrity, and feto‒placental
development. These peptides signal through CLR/RAMP1, 2 and 3 receptor
complexes. CLR/RAMP1, or CGRP receptor, antagonists have been developed for the
treatment of migraine headache and osteoarthritis pain; whereas CLR/RAMP2, or
ADM receptor, antagonists are being developed for the treatment of tumor
growth/metastasis. Based on the finding that an acylated chimeric ADM/ADM2
analog potently stimulates CLR/RAMP1 and 2 signaling, we hypothesized that the
binding domain of this analog could have potent inhibitory activity on CLR/RAMP
receptors. Consistent with this hypothesis, we showed that acylated truncated
ADM/ADM2 analogs of 27–31 residues exhibit potent antagonistic activity toward
CLR/RAMP1 and 2. On the other hand, nonacylated analogs have minimal activity.
Further truncation at the junctional region of these chimeric analogs led to the
generation of CLR/RAMP1-selective antagonists. A 17-amino-acid analog
(Antagonist 2–4) showed 100-fold selectivity for CLR/RAMP1 and was >100-fold
more potent than the classic CGRP receptor antagonist CGRP8-37. In addition, we
showed (1) a lysine residue in the Antagonist 2–4 is important for enhancing the
antagonistic activity, (2) an analog consisted of an ADM sequence motif and a
12-amino-acid binding domain of CGRP exhibits potent CLR/RAMP1-inhibitory
activity, and (3) a chimeric analog consisted of a somatostatin analog and an
ADM antagonist exhibits dual activities on somatostatin and CLR/RAMP receptors.
Because the blockage of CLR/RAMP signaling prevents migraine pain and suppresses
tumor growth/metastasis, further studies of these analogs, which presumably have
better access to the tumor microenvironment and nerve endings at the trigeminal
ganglion and synovial joints as compared to antibody-based therapies, may lead
to the development of better anti-CGRP therapy and alternative antiangiogenesis
therapy. Likewise, the use of bifunctional somatostatin-ADM antagonist analogs
could be a promising strategy for the treatment of high-grade neuroendocrine
tumors by targeting an antiangiogenesis agent to the neuroendocrine tumor
microenvironment.
Collapse
Affiliation(s)
- Chia Lin Chang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital
Linkou Medical Center, Chang Gung University, Kweishan, Taoyuan,
Taiwan
| | | |
Collapse
|
13
|
Li Z, Ho IHT, Li X, Xu D, Wu WKK, Chan MTV, Li S, Liu X. Long non-coding RNAs in the spinal cord injury: Novel spotlight. J Cell Mol Med 2019; 23:4883-4890. [PMID: 31140726 PMCID: PMC6653026 DOI: 10.1111/jcmm.14422] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 01/05/2023] Open
Abstract
Spinal cord injury (SCI) may lead to persistent locomotor dysfunction and somatosensory disorders, which adversely affect the quality of life of patients and cause a significant economic burden to the society. The efficacies of current therapeutic interventions are still far from satisfaction as the secondary damages resulting from the complex and progressive molecular alterations after SCI are not properly addressed. Recent studies revealed that long non‐coding RNAs (lncRNAs) are abundant in the brain and might play critical roles in several nervous system disorders. At the cellular level, lncRNAs have been shown to regulate the expression of protein‐coding RNAs and hence participate in neuronal death, demyelination and glia activation. Notably, SCI is characterized by these biological processes, suggesting that lncRNAs could be novel modulators in the pathogenesis of SCI. This review describes recent progresses in the lncRNA transcriptome analyses and their molecular functions in regulating SCI progression.
Collapse
Affiliation(s)
- Zheng Li
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Idy H T Ho
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Xingye Li
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital, Fourth Clinical College of Peking University, Jishuitan Orthopaedic College of Tsinghua University, Beijing, China
| | - Derong Xu
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - William K K Wu
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong.,State Key Laboratory of Digestive Diseases, Faculty of Medicine, Institute of Digestive Diseases and LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Matthew T V Chan
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Shugang Li
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaodong Liu
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
14
|
Eidson LN, Murphy AZ. Inflammatory mediators of opioid tolerance: Implications for dependency and addiction. Peptides 2019; 115:51-58. [PMID: 30890355 PMCID: PMC6863079 DOI: 10.1016/j.peptides.2019.01.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/24/2019] [Accepted: 01/30/2019] [Indexed: 12/27/2022]
Abstract
Each year, over 50 million Americans suffer from persistent pain, including debilitating headaches, joint pain, and severe back pain. Although morphine is amongst the most effective analgesics available for the management of severe pain, prolonged morphine treatment results in decreased analgesic efficacy (i.e., tolerance). Despite significant headway in the field, the mechanisms underlying the development of morphine tolerance are not well understood. The midbrain ventrolateral periaqueductal gray (vlPAG) is a primary neural substrate for the analgesic effects of morphine, as well as for the development of morphine tolerance. A growing body of literature indicates that activated glia (i.e., microglia and astrocytes) facilitate pain transmission and oppose morphine analgesia, making these cells important potential targets in the treatment of chronic pain. Morphine affects glia by binding to the innate immune receptor toll-like receptor 4 (TLR4), leading to the release of proinflammatory cytokines and opposition of morphine analgesia. Despite the established role of the vlPAG as an integral locus for the development of morphine tolerance, most studies have examined the role of glia activation within the spinal cord. Additionally, the role of TLR4 in the development of tolerance has not been elucidated. This review attempts to summarize what is known regarding the role of vlPAG glia and TLR4 in the development of morphine tolerance. These data, together, provide information about the mechanism by which central nervous system glia regulate morphine tolerance, and identify a potential therapeutic target for the enhancement of analgesic efficacy in the clinical treatment of chronic pain.
Collapse
Affiliation(s)
- Lori N Eidson
- Department of Physiology, Emory University, Atlanta, GA, 30322, United States
| | - Anne Z Murphy
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30308, United States.
| |
Collapse
|
15
|
Liu DQ, Zhou YQ, Gao F. Targeting Cytokines for Morphine Tolerance: A Narrative Review. Curr Neuropharmacol 2019; 17:366-376. [PMID: 29189168 PMCID: PMC6482476 DOI: 10.2174/1570159x15666171128144441] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/06/2017] [Accepted: 11/23/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Despite its various side effects, morphine has been widely used in clinics for decades due to its powerful analgesic effect. Morphine tolerance is one of the major side effects, hindering its long-term usage for pain therapy. Currently, the thorough cellular and molecular mechanisms underlying morphine tolerance remain largely uncertain. METHODS We searched the PubMed database with Medical subject headings (MeSH) including 'morphine tolerance', 'cytokines', 'interleukin 1', 'interleukin 1 beta', 'interleukin 6', 'tumor necrosis factor alpha', 'interleukin 10', 'chemokines'. Manual searching was carried out by reviewing the reference lists of relevant studies obtained from the primary search. The searches covered the period from inception to November 1, 2017. RESULTS The expression levels of certain chemokines and pro-inflammatory cytokines were significantly increased in animal models of morphine tolerance. Cytokines and cytokine receptor antagonist showed potent effect of alleviating the development of morphine tolerance. CONCLUSION Cytokines play a fundamental role in the development of morphine tolerance. Therapeutics targeting cytokines may become alternative strategies for the management of morphine tolerance.
Collapse
Affiliation(s)
| | | | - Feng Gao
- Address correspondence to this author at the Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, China; Tel: +86 27 83662853; E-mail:
| |
Collapse
|
16
|
Hu W, He T, Huo Y, Hong Y. Involvement of Adrenomedullin in Bone Cancer Pain in Rats. INT J PHARMACOL 2018. [DOI: 10.3923/ijp.2018.601.608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
17
|
Ferrero H, Larrayoz IM, Gil-Bea FJ, Martínez A, Ramírez MJ. Adrenomedullin, a Novel Target for Neurodegenerative Diseases. Mol Neurobiol 2018; 55:8799-8814. [PMID: 29600350 DOI: 10.1007/s12035-018-1031-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/22/2018] [Indexed: 01/18/2023]
Abstract
Neurodegenerative diseases represent a heterogeneous group of disorders whose common characteristic is the progressive degeneration of neuronal structure and function. Although much knowledge has been accumulated on the pathophysiology of neurodegenerative diseases over the years, more efforts are needed to understand the processes that underlie these diseases and hence to propose new treatments. Adrenomedullin (AM) is a multifunctional peptide involved in vasodilation, hormone secretion, antimicrobial defense, cellular growth, and angiogenesis. In neurons, AM and related peptides are associated with some structural and functional cytoskeletal proteins that interfere with microtubule dynamics. Furthermore, AM may intervene in neuronal dysfunction through other mechanisms such as immune and inflammatory response, apoptosis, or calcium dyshomeostasis. Alterations in AM expression have been described in neurodegenerative processes such as Alzheimer's disease or vascular dementia. This review addresses the current state of knowledge on AM and its possible implication in neurodegenerative diseases.
Collapse
Affiliation(s)
- Hilda Ferrero
- Department of Pharmacology and Toxicology, and IdiSNA, Navarra Institute for Health Research, University of Navarra, Pamplona, Spain
| | - Ignacio M Larrayoz
- Biomarkers and Molecular Signaling, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Francisco J Gil-Bea
- Department of Pharmacology and Toxicology, and IdiSNA, Navarra Institute for Health Research, University of Navarra, Pamplona, Spain
- Neuroscience Area, Biodonostia Health Research Institute, CIBERNED, San Sebastian, Spain
| | - Alfredo Martínez
- Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - María J Ramírez
- Department of Pharmacology and Toxicology, and IdiSNA, Navarra Institute for Health Research, University of Navarra, Pamplona, Spain.
| |
Collapse
|
18
|
Zhang Y, Wang K, Lin M, Li Q, Hong Y. Inhibition of morphine tolerance by MrgC receptor via modulation of interleukin-1β and matrix metalloproteinase 9 in dorsal root ganglia in rats. Eur J Pharmacol 2017; 815:10-17. [PMID: 28993160 DOI: 10.1016/j.ejphar.2017.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 09/30/2017] [Accepted: 10/05/2017] [Indexed: 11/27/2022]
Abstract
Opiate tolerance is a critical issue in pain management. Previous studies show that activation of Mas-related gene (Mrg) C receptor can modulate the development of morphine tolerance. This study was designed to investigate the underlying mechanism(s). Intrathecal (i.t.) administration of morphine (20µg) increased the expression of interleukin-1β (IL-1β) and matrix metalloproteinase-9 (MMP-9) in small- and medium-sized neurons in dorsal root ganglia (DRG). Co-administration of bovine adrenal medulla 8-22 (BAM8-22), a selective MrgC receptor agonist, via i.t. route inhibited the increase of IL-1β and MMP-9 in the DRG. Exposure of DRG cultures to morphine (3.3μM) for 3 or 5 days, but not for 1 day, induced an increase in MMP-9 mRNA expression. The treatment with BAM8-22 (10nM) for 20, 40 or 60min abolished chronic (5 days) morphine-induced increase of MMP-9 mRNA in the cultured DRG. The treatment with BAM8-22 for 1h inhibited chronic morphine-induced increase of MMP-9 and IL-1β mRNA in DRG but these effects were abolished by MrgC receptor antibody. The treatment with BAM8-22 for 24 and 72h respectively inhibited and enhanced morphine-induced expression of MMP-9 and IL-1β mRNA in the cultured DRG. The BAM8-22-induced inhibition and enhancement were abolished by MrgC receptor antibody. The results suggest that the inhibition of IL-1β and MMP-9 expressions in DRG underlain the modulation of morphine tolerance by the acute activation of MrgC receptors. The chronic activation of MrgC receptors can facilitate morphine-induced increase of MMP-9 and IL-1β expressions in DRG.
Collapse
Affiliation(s)
- Yue Zhang
- Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian 350117, China
| | - Kai Wang
- Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian 350117, China
| | - Minyan Lin
- Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian 350117, China
| | - Qi Li
- Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian 350117, China
| | - Yanguo Hong
- Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian 350117, China.
| |
Collapse
|
19
|
Abstract
This paper is the thirty-eighth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2015 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia, stress and social status, tolerance and dependence, learning and memory, eating and drinking, drug abuse and alcohol, sexual activity and hormones, pregnancy, development and endocrinology, mental illness and mood, seizures and neurologic disorders, electrical-related activity and neurophysiology, general activity and locomotion, gastrointestinal, renal and hepatic functions, cardiovascular responses, respiration and thermoregulation, and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
20
|
Chandran R, Mehta SL, Vemuganti R. Non-coding RNAs and neuroprotection after acute CNS injuries. Neurochem Int 2017; 111:12-22. [PMID: 28131900 DOI: 10.1016/j.neuint.2017.01.015] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 01/17/2017] [Accepted: 01/24/2017] [Indexed: 02/07/2023]
Abstract
Accumulating evidence indicates that various classes of non-coding RNAs (ncRNAs) including microRNAs (miRNAs), PIWI-interacting RNAs (piRNAs) and long non-coding RNAs (lncRNAs) play important roles in normal state as well as the diseases of the CNS. Interestingly, ncRNAs have been shown to interact with messenger RNA, DNA and proteins, and these interactions could induce epigenetic modifications and control transcription and translation, thereby adding a new layer of genomic regulation. The ncRNA expression profiles are known to be altered after acute CNS injuries including stroke, traumatic brain injury and spinal cord injury that are major contributors of morbidity and mortality worldwide. Hence, a better understanding of the functional significance of ncRNAs following CNS injuries could help in developing potential therapeutic strategies to minimize the neuronal damage in those conditions. The potential of ncRNAs in blood and CSF as biomarkers for diagnosis and/or prognosis of acute CNS injuries has also gained importance in the recent years. This review highlighted the current progress in the understanding of the role of ncRNAs in initiation and progression of secondary neuronal damage and their application as biomarkers after acute CNS injuries.
Collapse
Affiliation(s)
- Raghavendar Chandran
- Department of Neurological Surgery, University of Wisconsin-Madison and William S. Middleton Veterans Hospital, Madison, WI, USA
| | - Suresh L Mehta
- Department of Neurological Surgery, University of Wisconsin-Madison and William S. Middleton Veterans Hospital, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin-Madison and William S. Middleton Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
21
|
Neuropeptides and Microglial Activation in Inflammation, Pain, and Neurodegenerative Diseases. Mediators Inflamm 2017; 2017:5048616. [PMID: 28154473 PMCID: PMC5244030 DOI: 10.1155/2017/5048616] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/26/2016] [Accepted: 12/05/2016] [Indexed: 12/15/2022] Open
Abstract
Microglial cells are responsible for immune surveillance within the CNS. They respond to noxious stimuli by releasing inflammatory mediators and mounting an effective inflammatory response. This is followed by release of anti-inflammatory mediators and resolution of the inflammatory response. Alterations to this delicate process may lead to tissue damage, neuroinflammation, and neurodegeneration. Chronic pain, such as inflammatory or neuropathic pain, is accompanied by neuroimmune activation, and the role of glial cells in the initiation and maintenance of chronic pain has been the subject of increasing research over the last two decades. Neuropeptides are small amino acidic molecules with the ability to regulate neuronal activity and thereby affect various functions such as thermoregulation, reproductive behavior, food and water intake, and circadian rhythms. Neuropeptides can also affect inflammatory responses and pain sensitivity by modulating the activity of glial cells. The last decade has witnessed growing interest in the study of microglial activation and its modulation by neuropeptides in the hope of developing new therapeutics for treating neurodegenerative diseases and chronic pain. This review summarizes the current literature on the way in which several neuropeptides modulate microglial activity and response to tissue damage and how this modulation may affect pain sensitivity.
Collapse
|
22
|
Reversal of neurochemical alterations in the spinal dorsal horn and dorsal root ganglia by Mas-related gene (Mrg) receptors in a rat model of spinal nerve injury. Neurobiol Dis 2016; 91:274-83. [DOI: 10.1016/j.nbd.2016.03.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 03/12/2016] [Accepted: 03/21/2016] [Indexed: 12/17/2022] Open
|
23
|
Wang D, Xue Y, Chen Y, Ruan L, Hong Y. Mas-related gene (Mrg) C receptors inhibit mechanical allodynia and spinal microglia activation in the early phase of neuropathic pain in rats. Neurosci Lett 2016; 618:115-121. [PMID: 26952974 DOI: 10.1016/j.neulet.2016.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 03/01/2016] [Accepted: 03/02/2016] [Indexed: 02/06/2023]
Abstract
Mas-related gene (Mrg) C receptors are exclusively expressed in the trigeminal and dorsal root ganglia (DRG). However, their functional roles are poorly understood. This study was aimed to determine the effect of MrgC receptors on pain hypersensitivity in the early phase of neuropathic pain and its underlying mechanisms. Intrathecal (i.t.) administration of the selective MrgC receptor agonist bovine adrenal medulla 8-22 (BAM8-22) at 1 or 10nmol attenuated mechanical allodynia one day after L5 spinal nerve ligation (SNL) surgery. I.t. BAM8-22 (10 nmol) inhibited SNL-induced microglia activation in the spinal dorsal horn on day 2 post-SNL. The BAM8-22 treatment also abolished SNL-induced upregulation of neuronal nitric oxide synthesis (nNOS) in the dorsal root ganglia (DRG). On the other hand, SNL, but not sham, surgery reduced the expression of MrgC receptor mRNA in the injured L5 DRG without changing thier levels in the adjacent uninjured L4 or L6 DRG on day 2 following the surgery. These results suggest that the activation of MrgC receptors can relieve pain hypersensitivity by the inhibition of nNOS increase in DRG neurons and microglia activation in the spinal dorsal horn in the early time following peripheral nerve injury. This study provides evidence that MrgC receptors could be targeted as a novel therapy for neuropathic pain with limited unwanted effects.
Collapse
Affiliation(s)
- Dongmei Wang
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University Fuzhou, Fujian 350108, People's Republic of China
| | - Yaping Xue
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University Fuzhou, Fujian 350108, People's Republic of China
| | - Yajuan Chen
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University Fuzhou, Fujian 350108, People's Republic of China
| | - Liqin Ruan
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University Fuzhou, Fujian 350108, People's Republic of China
| | - Yanguo Hong
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University Fuzhou, Fujian 350108, People's Republic of China.
| |
Collapse
|
24
|
Wang D, Zeng J, Li Q, Huang J, Couture R, Hong Y. Contribution of adrenomedullin to the switch of G protein-coupled μ-opioid receptors from Gi to Gs in the spinal dorsal horn following chronic morphine exposure in rats. Br J Pharmacol 2016; 173:1196-207. [PMID: 26750148 DOI: 10.1111/bph.13419] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 12/30/2015] [Accepted: 01/07/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE Chronic exposure to morphine increases spinal adrenomedullin (AM) bioactivity resulting in the development and maintenance of morphine tolerance. This study investigated the possible involvement of AM in morphine-evoked alteration in μ-opioid receptor-coupled G proteins. EXPERIMENTAL APPROACH Agents were administered intrathecally (i.t.) in rats. Nociceptive behaviours and cumulative dose-response of morphine analgesia were assessed. Neurochemicals in the spinal dorsal horn were assayed by immunoprecipitation, Western blot analysis and ELISA. KEY RESULTS Intrathecal injection of AM (8 μg) for 9 days decreased and increased the levels of μ receptor-coupled Gi and Gs proteins respectively. Morphine stimulation (5 μg) after chronic treatment with AM also induced an increase in cAMP production in the spinal dorsal horn. Co-administration of the selective AM receptor antagonist AM22-52 inhibited chronic morphine-evoked switch of G protein-coupled μ receptor from Gi to Gs. Chronic exposure to AM increased the phosphorylation of cAMP-responsive element-binding protein (CREB) and ERK. Co-administration of the PKA inhibitor H-89 (5 μg) or MEK1 inhibitor PD98059 (1 μg) reversed the AM-induced thermal/mechanical hypersensitivity, decline in morphine analgesic potency, switch of G protein-coupled μ receptor and increase in cAMP. CONCLUSIONS AND IMPLICATIONS The present study supports the hypothesis that an increase in AM activity in the spinal dorsal horn contributes to the switch of the μ receptor-coupled G protein from Gi to Gs protein via the activation of cAMP/PKA/CREB and ERK signalling pathways in chronic morphine use.
Collapse
Affiliation(s)
- Dongmei Wang
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian, China
| | - Juan Zeng
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian, China
| | - Qi Li
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian, China
| | - Jianzhong Huang
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian, China
| | - Réjean Couture
- Department of Molecular and Integrative Physiology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Yanguo Hong
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian, China
| |
Collapse
|