1
|
Ma X, Qi Q, Wang W, Huang M, Wang H, Luo L, Xu X, Yuan T, Shi H, Jiang W, Xu T. Astrocytic pyruvate dehydrogenase kinase-lactic acid axis involvement in glia-neuron crosstalk contributes to morphine-induced hyperalgesia in mice. FUNDAMENTAL RESEARCH 2024; 4:820-828. [PMID: 39161415 PMCID: PMC11331729 DOI: 10.1016/j.fmre.2023.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
The activation of spinal astrocytes accounts for opioid-induced hyperalgesia (OIH), but the underlying mechanisms remain elusive. The presence of astrocyte-neuron lactate shuttle (ANLS) makes astrocytes necessary for some neural function and communication. The aim of this study was to explore the role of ANLS in the occurrence and maintenance of OIH. After 7 days consecutive morphine injection, a mice OIH model was established and astrocytic pyruvate dehydrogenase kinase 4 (PDK4), phosphorylated pyruvate dehydrogenase (p-PDH) and accumulation of L-lactate was elevated in the spinal dorsal horn. Intrathecally administration of inhibitors of PDK, lactate dehydrogenase 5 and monocarboxylate transporters to decrease the supply of L-lactate on neurons was observed to attenuate hypersensitivity behaviors induced by repeated morphine administration and downregulate the expression of markers of central sensitization in the spinal dorsal horns. The astrocyte line and the neuronal line were co-cultured to investigate the mechanisms in vitro. In this study, we demonstrated that morphine-induced hyperalgesia was sustained by lactate overload consequent upon aberrant function of spinal ANLS. In this process, PDK-p-PDH-lactate axis serves a pivotal role, which might therefore be a new target to improve long-term opioid treatment strategy in clinical practice.
Collapse
Affiliation(s)
- Xiaqing Ma
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Qi Qi
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Wenying Wang
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Min Huang
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Haiyan Wang
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Limin Luo
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xiaotao Xu
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Tifei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Haibo Shi
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai 200233, China
| | - Wei Jiang
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Tao Xu
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Department of Anesthesiology, Suzhou Hospital of Anhui Medical University, Suzhou 234000, China
| |
Collapse
|
2
|
Duron DI, Tanguturi P, Campbell CS, Chou K, Bejarano P, Gabriel KA, Bowden JL, Mishra S, Brackett C, Barlow D, Houseknecht KL, Blagg BSJ, Streicher JM. Inhibiting spinal cord-specific hsp90 isoforms reveals a novel strategy to improve the therapeutic index of opioid treatment. Sci Rep 2024; 14:14715. [PMID: 38926482 PMCID: PMC11208559 DOI: 10.1038/s41598-024-65637-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 06/21/2024] [Indexed: 06/28/2024] Open
Abstract
Opioids are the gold standard for the treatment of chronic pain but are limited by adverse side effects. In our earlier work, we showed that Heat shock protein 90 (Hsp90) has a crucial role in regulating opioid signaling in spinal cord; Hsp90 inhibition in spinal cord enhances opioid anti-nociception. Building on these findings, we injected the non-selective Hsp90 inhibitor KU-32 by the intrathecal route into male and female CD-1 mice, showing that morphine anti-nociceptive potency was boosted by 1.9-3.5-fold in acute and chronic pain models. At the same time, tolerance was reduced from 21-fold to 2.9 fold and established tolerance was rescued, while the potency of constipation and reward was unchanged. These results demonstrate that spinal Hsp90 inhibition can improve the therapeutic index of morphine. However, we also found that systemic non-selective Hsp90 inhibition blocked opioid pain relief. To avoid this effect, we used selective small molecule inhibitors and CRISPR gene editing to identify 3 Hsp90 isoforms active in spinal cord (Hsp90α, Hsp90β, and Grp94) while only Hsp90α was active in brain. We thus hypothesized that a systemically delivered selective inhibitor to Hsp90β or Grp94 could selectively inhibit spinal cord Hsp90 activity, resulting in enhanced opioid therapy. We tested this hypothesis using intravenous delivery of KUNB106 (Hsp90β) and KUNG65 (Grp94), showing that both drugs enhanced morphine anti-nociceptive potency while rescuing tolerance. Together, these results suggest that selective inhibition of spinal cord Hsp90 isoforms is a novel, translationally feasible strategy to improve the therapeutic index of opioids.
Collapse
Affiliation(s)
- David I Duron
- Department of Pharmacology, College of Medicine, University of Arizona, Box 245050, LSN563, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Parthasaradhireddy Tanguturi
- Department of Pharmacology, College of Medicine, University of Arizona, Box 245050, LSN563, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Christopher S Campbell
- Department of Pharmacology, College of Medicine, University of Arizona, Box 245050, LSN563, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Kerry Chou
- Department of Pharmacology, College of Medicine, University of Arizona, Box 245050, LSN563, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Paul Bejarano
- Department of Pharmacology, College of Medicine, University of Arizona, Box 245050, LSN563, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Katherin A Gabriel
- Department of Pharmacology, College of Medicine, University of Arizona, Box 245050, LSN563, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Jessica L Bowden
- Department of Pharmacology, College of Medicine, University of Arizona, Box 245050, LSN563, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Sanket Mishra
- Department of Chemistry and Biochemistry, College of Science, University of Notre Dame, Notre Dame, IN, USA
| | - Christopher Brackett
- Department of Chemistry and Biochemistry, College of Science, University of Notre Dame, Notre Dame, IN, USA
| | - Deborah Barlow
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, ME, USA
| | - Karen L Houseknecht
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, ME, USA
| | - Brian S J Blagg
- Department of Chemistry and Biochemistry, College of Science, University of Notre Dame, Notre Dame, IN, USA
| | - John M Streicher
- Department of Pharmacology, College of Medicine, University of Arizona, Box 245050, LSN563, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA.
- Comprehensive Center for Pain and Addiction, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
3
|
Liang Z, Li L, Bai L, Gao Y, Qiao Y, Wang X, Yv L, Xu JT. Spinal nerve transection-induced upregulation of SAP97 via promoting membrane trafficking of GluA1-containing AMPA receptors in the dorsal horn contributes to the pathogenesis of neuropathic pain. Neurobiol Dis 2024; 194:106471. [PMID: 38461868 DOI: 10.1016/j.nbd.2024.106471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/07/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024] Open
Abstract
Emerging evidence has implicated an important role of synapse-associated protein-97 (SAP97)-regulated GluA1-containing AMPARs membrane trafficking in cocaine restate and in contextual episodic memory of schizophrenia. Herein, we investigated the role of SAP97 in neuropathic pain following lumbar 5 spinal nerve transection (SNT) in rats. Our results showed that SNT led to upregulation of SAP97, enhanced the interaction between SAP97 and GluA1, and increased GluA1-containing AMPARs membrane trafficking in the dorsal horn. Microinjection of AAV-EGFP-SAP97 shRNA in lumbar 5 spinal dorsal horn inhibited SAP97 production, decreased SAP97-GluA1 interaction, reduced the membrane trafficking of GluA1-containing AMPARs, and partially attenuated neuropathic pain following SNT. Intrathecal injections of SAP97 siRNA or NASPM, an antagonist of GluA1-containing AMPARs, also partially reversed neuropathic pain on day 7, but not on day 14, after SNT. Spinal overexpression of SAP97 by AAV-EGFP-SAP97 enhanced SAP97-GluA1 interaction, increased the membrane insertion of GluA1-containing AMPARs, and induced abnormal pain in naïve rats. In addition, treatment with SAP97 siRNA or NASPM i.t. injection alleviated SNT-induced allodynia and hyperalgesia and exhibited a longer effect in female rats. Together, our results indicate that the SNT-induced upregulation of SAP97 via promoting GluA1-containing AMPARs membrane trafficking in the dorsal horn contributes to the pathogenesis of neuropathic pain. Targeting spinal SAP97 might be a promising therapeutic strategy to treatment of chronic pain.
Collapse
Affiliation(s)
- Zongyi Liang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Liren Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Liying Bai
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Yan Gao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Yiming Qiao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Xueli Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Lili Yv
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Ji-Tian Xu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China; Neuroscience Research Institute, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China.
| |
Collapse
|
4
|
Wang W, Ma X, Du W, Lin R, Li Z, Jiang W, Wang LY, Worley PF, Xu T. Small G-Protein Rheb Gates Mammalian Target of Rapamycin Signaling to Regulate Morphine Tolerance in Mice. Anesthesiology 2024; 140:786-802. [PMID: 38147625 DOI: 10.1097/aln.0000000000004885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
BACKGROUND Analgesic tolerance due to long-term use of morphine remains a challenge for pain management. Morphine acts on μ-opioid receptors and downstream of the phosphatidylinositol 3-kinase signaling pathway to activate the mammalian target of rapamycin (mTOR) pathway. Rheb is an important regulator of growth and cell-cycle progression in the central nervous system owing to its critical role in the activation of mTOR. The hypothesis was that signaling via the GTP-binding protein Rheb in the dorsal horn of the spinal cord is involved in morphine-induced tolerance. METHODS Male and female wild-type C57BL/6J mice or transgenic mice (6 to 8 weeks old) were injected intrathecally with saline or morphine twice daily at 12-h intervals for 5 consecutive days to establish a tolerance model. Analgesia was assessed 60 min later using the tail-flick assay. After 5 days, the spine was harvested for Western blot or immunofluorescence analysis. RESULTS Chronic morphine administration resulted in the upregulation of spinal Rheb by 4.27 ± 0.195-fold (P = 0.0036, n = 6), in turn activating mTOR by targeting rapamycin complex 1 (mTORC1). Genetic overexpression of Rheb impaired morphine analgesia, resulting in a tail-flick latency of 4.65 ± 1.10 s (P < 0.0001, n = 7) in Rheb knock-in mice compared to 10 s in control mice (10 ± 0 s). Additionally, Rheb overexpression in spinal excitatory neurons led to mTORC1 signaling overactivation. Genetic knockout of Rheb or inhibition of mTORC1 signaling by rapamycin potentiated morphine-induced tolerance (maximum possible effect, 52.60 ± 9.56% in the morphine + rapamycin group vs. 16.60 ± 8.54% in the morphine group; P < 0.0001). Moreover, activation of endogenous adenosine 5'-monophosphate-activated protein kinase inhibited Rheb upregulation and retarded the development of morphine-dependent tolerance (maximum possible effect, 39.51 ± 7.40% in morphine + metformin group vs. 15.58 ± 5.79% in morphine group; P < 0.0001). CONCLUSIONS This study suggests spinal Rheb as a key molecular factor for regulating mammalian target of rapamycin signaling. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Wenying Wang
- Department of Anesthesiology, Sixth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaqing Ma
- Department of Anesthesiology, Sixth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjie Du
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Raozhou Lin
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zhongping Li
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Wei Jiang
- Department of Anesthesiology, Sixth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu-Yang Wang
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada; and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Paul F Worley
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Tao Xu
- Department of Anesthesiology, Sixth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Anesthesiology, Suzhou Hospital of Anhui Medical University, Suzhou, China; and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
5
|
Micheli L, Lucarini E, Nobili S, Bartolucci G, Pallecchi M, Toti A, Ferrara V, Ciampi C, Ghelardini C, Di Cesare Mannelli L. Ultramicronized N-palmitoylethanolamine Contributes to Morphine Efficacy Against Neuropathic Pain: Implication of Mast Cells and Glia. Curr Neuropharmacol 2024; 22:88-106. [PMID: 36443965 PMCID: PMC10716887 DOI: 10.2174/1570159x21666221128091453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/04/2022] [Accepted: 11/05/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND In the current management of neuropathic pain, in addition to antidepressants and anticonvulsants, the use of opioids is wide, despite their related and well-known issues. OBJECTIVE N-palmitoylethanolamine (PEA), a natural fatty-acid ethanolamide whose anti-inflammatory, neuroprotective, immune-modulating and anti-hyperalgesic activities are known, represents a promising candidate to modulate and/or potentiate the action of opioids. METHODS This study was designed to evaluate if the preemptive and morphine concomitant administration of ultramicronized PEA, according to fixed or increasing doses of both compounds, delays the onset of morphine tolerance and improves its analgesic efficacy in the chronic constriction injury (CCI) model of neuropathic pain in rats. RESULTS Behavioral experiments showed that the preemptive and co-administration of ultramicronized PEA significantly decreased the effective dose of morphine and delayed the onset of morphine tolerance. The activation of spinal microglia and astrocytes, commonly occurring both on opioid treatment and neuropathic pain, was investigated through GFAP and Iba-1 immunofluorescence. Both biomarkers were found to be increased in CCI untreated or morphine treated animals in a PEA-sensitive manner. The increased density of endoneural mast cells within the sciatic nerve of morphine-treated and untreated CCI rats was significantly reduced by ultramicronized PEA. The decrease of mast cell degranulation, evaluated in terms of reduced plasma levels of histamine and N-methyl-histamine metabolite, was mainly observed at intermediate-high doses of ultramicronized PEA, with or without morphine. CONCLUSION Overall, these results show that the administration of ultramicronized PEA in CCI rats according to the study design fully fulfilled the hypotheses of this study.
Collapse
Affiliation(s)
- Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Stefania Nobili
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Gianluca Bartolucci
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmaceutical and Nutraceutical Sciences Section, University of Florence, Florence, 50019, Italy
| | - Marco Pallecchi
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmaceutical and Nutraceutical Sciences Section, University of Florence, Florence, 50019, Italy
| | - Alessandra Toti
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Valentina Ferrara
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Clara Ciampi
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| |
Collapse
|
6
|
Qiao Y, Li L, Bai L, Gao Y, Yang Y, Wang L, Wang X, Liang Z, Xu J. Upregulation of lysine-specific demethylase 6B aggravates inflammatory pain through H3K27me3 demethylation-dependent production of TNF-α in the dorsal root ganglia and spinal dorsal horn in rats. CNS Neurosci Ther 2023; 29:3479-3492. [PMID: 37287407 PMCID: PMC10580362 DOI: 10.1111/cns.14281] [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: 02/02/2023] [Revised: 05/03/2023] [Accepted: 05/09/2023] [Indexed: 06/09/2023] Open
Abstract
AIMS Lysine-specific demethylase 6B (KDM6B) serves as a key mediator of gene transcription. It regulates expression of proinflammatory cytokines and chemokines in variety of diseases. Herein, the role and the underlying mechanisms of KDM6B in inflammatory pain were studied. METHODS The inflammatory pain was conducted by intraplantar injection of complete Freund's adjuvant (CFA) in rats. Immunofluorescence, Western blotting, qRT-PCR, and chromatin immunoprecipitation (ChIP)-PCR were performed to investigate the underlying mechanisms. RESULTS CFA injection led to upregulation of KDM6B and decrease in the level of H3K27me3 in the dorsal root ganglia (DRG) and spinal dorsal horn. The mechanical allodynia and thermal hyperalgesia following CFA were alleviated by the treatment of intrathecal injection of GSK-J4, and by microinjection of AAV-EGFP-KDM6B shRNA in the sciatic nerve or in lumbar 5 dorsal horn. The increased production of tumor necrosis factor-α (TNF-α) following CFA in the DRGs and dorsal horn was inhibited by these treatments. ChIP-PCR showed that CFA-induced increased binding of nuclear factor κB with TNF-α promoter was repressed by the treatment of microinjection of AAV-EGFP-KDM6B shRNA. CONCLUSIONS These results suggest that upregulated KDM6B via facilitating TNF-α expression in the DRG and spinal dorsal horn aggravates inflammatory pain.
Collapse
Affiliation(s)
- Yiming Qiao
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Liren Li
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Liying Bai
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated HospitalZhengzhou UniversityZhengzhouChina
| | - Yan Gao
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Yin Yang
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Li Wang
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Xueli Wang
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Zongyi Liang
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Ji‐Tian Xu
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
- Neuroscience Research InstituteZhengzhou UniversityZhengzhouChina
| |
Collapse
|
7
|
Wang B, Ma L, Guo X, Du S, Feng X, Liang Y, Govindarajalu G, Wu S, Liu T, Li H, Patel S, Bekker A, Hu H, Tao YX. A sensory neuron-specific long non-coding RNA reduces neuropathic pain by rescuing KCNN1 expression. Brain 2023; 146:3866-3884. [PMID: 37012681 PMCID: PMC10473565 DOI: 10.1093/brain/awad110] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 02/21/2023] [Accepted: 03/10/2023] [Indexed: 04/05/2023] Open
Abstract
Nerve injury to peripheral somatosensory system causes refractory neuropathic pain. Maladaptive changes of gene expression in primary sensory neurons are considered molecular basis of this disorder. Long non-coding RNAs (lncRNAs) are key regulators of gene transcription; however, their significance in neuropathic pain remains largely elusive.Here, we reported a novel lncRNA, named sensory neuron-specific lncRNA (SS-lncRNA), for its expression exclusively in dorsal root ganglion (DRG) and trigeminal ganglion. SS-lncRNA was predominantly expressed in small DRG neurons and significantly downregulated due to a reduction of early B cell transcription factor 1 in injured DRG after nerve injury. Rescuing this downregulation reversed a decrease of the calcium-activated potassium channel subfamily N member 1 (KCNN1) in injured DRG and alleviated nerve injury-induced nociceptive hypersensitivity. Conversely, DRG downregulation of SS-lncRNA reduced the expression of KCNN1, decreased total potassium currents and afterhyperpolarization currents and increased excitability in DRG neurons and produced neuropathic pain symptoms.Mechanistically, downregulated SS-lncRNA resulted in the reductions of its binding to Kcnn1 promoter and heterogeneous nuclear ribonucleoprotein M (hnRNPM), consequent recruitment of less hnRNPM to the Kcnn1 promoter and silence of Kcnn1 gene transcription in injured DRG.These findings indicate that SS-lncRNA may relieve neuropathic pain through hnRNPM-mediated KCNN1 rescue in injured DRG and offer a novel therapeutic strategy specific for this disorder.
Collapse
Affiliation(s)
- Bing Wang
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Longfei Ma
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Xinying Guo
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Shibin Du
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Xiaozhou Feng
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Yingping Liang
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Gokulapriya Govindarajalu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Shaogen Wu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Tong Liu
- Center for Advanced Proteomics Research, Departments of Biochemistry, Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Hong Li
- Center for Advanced Proteomics Research, Departments of Biochemistry, Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Shivam Patel
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Alex Bekker
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Huijuan Hu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
- Department of Physiology, Pharmacology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
- Department of Physiology, Pharmacology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| |
Collapse
|
8
|
Adzic M, Lukic I, Mitic M, Glavonic E, Dragicevic N, Ivkovic S. Contribution of the opioid system to depression and to the therapeutic effects of classical antidepressants and ketamine. Life Sci 2023:121803. [PMID: 37245840 DOI: 10.1016/j.lfs.2023.121803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023]
Abstract
Major depressive disorder (MDD) afflicts approximately 5 % of the world population, and about 30-50 % of patients who receive classical antidepressant medications do not achieve complete remission (treatment resistant depressive patients). Emerging evidence suggests that targeting opioid receptors mu (MOP), kappa (KOP), delta (DOP), and the nociceptin/orphanin FQ receptor (NOP) may yield effective therapeutics for stress-related psychiatric disorders. As depression and pain exhibit significant overlap in their clinical manifestations and molecular mechanisms involved, it is not a surprise that opioids, historically used to alleviate pain, emerged as promising and effective therapeutic options in the treatment of depression. The opioid signaling is dysregulated in depression and numerous preclinical studies and clinical trials strongly suggest that opioid modulation can serve as either an adjuvant or even an alternative to classical monoaminergic antidepressants. Importantly, some classical antidepressants require the opioid receptor modulation to exert their antidepressant effects. Finally, ketamine, a well-known anesthetic whose extremely efficient antidepressant effects were recently discovered, was shown to mediate its antidepressant effects via the endogenous opioid system. Thus, although opioid system modulation is a promising therapeutical venue in the treatment of depression further research is warranted to fully understand the benefits and weaknesses of such approach.
Collapse
Affiliation(s)
- Miroslav Adzic
- Department of Molecular Biology and Endocrinology, Vinca - Institute for Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| | - Iva Lukic
- Department of Molecular Biology and Endocrinology, Vinca - Institute for Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milos Mitic
- Department of Molecular Biology and Endocrinology, Vinca - Institute for Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Emilija Glavonic
- Department of Molecular Biology and Endocrinology, Vinca - Institute for Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Nina Dragicevic
- Department of Pharmacy, Singidunum University, Belgrade, Serbia
| | - Sanja Ivkovic
- Department of Molecular Biology and Endocrinology, Vinca - Institute for Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
9
|
Cuitavi J, Torres-Pérez JV, Lorente JD, Campos-Jurado Y, Andrés-Herrera P, Polache A, Agustín-Pavón C, Hipólito L. Crosstalk between Mu-Opioid receptors and neuroinflammation: Consequences for drug addiction and pain. Neurosci Biobehav Rev 2023; 145:105011. [PMID: 36565942 DOI: 10.1016/j.neubiorev.2022.105011] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/29/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
Mu-Opioid Receptors (MORs) are well-known for participating in analgesia, sedation, drug addiction, and other physiological functions. Although MORs have been related to neuroinflammation their biological mechanism remains unclear. It is suggested that MORs work alongside Toll-Like Receptors to enhance the release of pro-inflammatory mediators and cytokines during pathological conditions. Some cytokines, including TNF-α, IL-1β and IL-6, have been postulated to regulate MORs levels by both avoiding MOR recycling and enhancing its production. In addition, Neurokinin-1 Receptor, also affected during neuroinflammation, could be regulating MOR trafficking. Therefore, inflammation in the central nervous system seems to be associated with altered/increased MORs expression, which might regulate harmful processes, such as drug addiction and pain. Here, we provide a critical evaluation on MORs' role during neuroinflammation and its implication for these conditions. Understanding MORs' functioning, their regulation and implications on drug addiction and pain may help elucidate their potential therapeutic use against these pathological conditions and associated disorders.
Collapse
Affiliation(s)
- Javier Cuitavi
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avda. Vicent Andrés Estellés s/n., 46100 Burjassot, Spain.
| | - Jose Vicente Torres-Pérez
- Department of Cellular Biology, Functional Biology and Physical Anthropology, University of Valencia, Avda. Vicent Andrés Estellés s/n., 46100 Burjassot, Spain
| | - Jesús David Lorente
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avda. Vicent Andrés Estellés s/n., 46100 Burjassot, Spain
| | - Yolanda Campos-Jurado
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avda. Vicent Andrés Estellés s/n., 46100 Burjassot, Spain
| | - Paula Andrés-Herrera
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avda. Vicent Andrés Estellés s/n., 46100 Burjassot, Spain
| | - Ana Polache
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avda. Vicent Andrés Estellés s/n., 46100 Burjassot, Spain
| | - Carmen Agustín-Pavón
- Department of Cellular Biology, Functional Biology and Physical Anthropology, University of Valencia, Avda. Vicent Andrés Estellés s/n., 46100 Burjassot, Spain
| | - Lucía Hipólito
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avda. Vicent Andrés Estellés s/n., 46100 Burjassot, Spain.
| |
Collapse
|
10
|
Liu M, Mai JW, Luo DX, Liu GX, Xu T, Xin WJ, Lin SY, Li ZY. NFATc2-dependent epigenetic downregulation of the TSC2/Beclin-1 pathway is involved in neuropathic pain induced by oxaliplatin. Mol Pain 2023; 19:17448069231158289. [PMID: 36733258 PMCID: PMC9941598 DOI: 10.1177/17448069231158289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 01/16/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Neuropathic pain is a common dose-limiting side effect of oxaliplatin, which hampers the effective treatment of tumors. Here, we found that upregulation of transcription factor NFATc2 decreased the expression of Beclin-1, a critical molecule in autophagy, in the spinal dorsal horn, and contributed to neuropathic pain following oxaliplatin treatment. Meanwhile, manipulating autophagy levels by intrathecal injection of rapamycin (RAPA) or 3-methyladenine (3-MA) differentially altered mechanical allodynia in oxaliplatin-treated or naïve rats. Utilizing chromatin immunoprecipitation-sequencing (ChIP-seq) assay combined with bioinformatics analysis, we found that NFATc2 negatively regulated the transcription of tuberous sclerosis complex protein 2 (TSC2), which contributed to the oxaliplatin-induced Beclin-1 downregulation. Further assays revealed that NFATc2 regulated histone H4 acetylation and methylation in the TSC2 promoter site 1 in rats' dorsal horns with oxaliplatin treatment. These results suggested that NFATc2 mediated the epigenetic downregulation of the TSC2/Beclin-1 autophagy pathway and contributed to oxaliplatin-induced mechanical allodynia, which provided a new therapeutic insight for chemotherapy-induced neuropathic pain.
Collapse
Affiliation(s)
- Meng Liu
- Department of Anesthesia and Pain Medicine, Guangzhou First People’s Hospital, Guangzhou, China
| | - Jing-Wen Mai
- Department of Anesthesiology, Huizhou Central People’s Hospital, Huizhou, China
| | - De-Xing Luo
- Department of Anesthesiology, Huizhou Central People’s Hospital, Huizhou, China
| | - Guan-Xi Liu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Huiai Hospital, Guangzhou, China
| | - Ting Xu
- Department of Emergency Medicine, The First Affiliated Hospital of Sun Yat-Sen University and Zhongshan Medical School, Sun Yat-Sen University, China
| | - Wen-Jun Xin
- Department of Emergency Medicine, The First Affiliated Hospital of Sun Yat-Sen University and Zhongshan Medical School, Sun Yat-Sen University, China
| | - Su-Yan Lin
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhen-Yu Li
- Department of Emergency Medicine, The First Affiliated Hospital of Sun Yat-Sen University and Zhongshan Medical School, Sun Yat-Sen University, China
| |
Collapse
|
11
|
Jin Y, Mao Y, Chen D, Tai Y, Hu R, Yang CL, Zhou J, Chen L, Liu X, Gu E, Jia C, Zhang Z, Tao W. Thalamocortical circuits drive remifentanil-induced postoperative hyperalgesia. J Clin Invest 2022; 132:158742. [PMID: 36519547 PMCID: PMC9754001 DOI: 10.1172/jci158742] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 10/18/2022] [Indexed: 12/15/2022] Open
Abstract
Remifentanil-induced hyperalgesia (RIH) is a severe but common postoperative clinical problem with elusive underlying neural mechanisms. Here, we discovered that glutamatergic neurons in the thalamic ventral posterolateral nucleus (VPLGlu) exhibited significantly elevated burst firing accompanied by upregulation of Cav3.1 T-type calcium channel expression and function in RIH model mice. In addition, we identified a glutamatergic neuronal thalamocortical circuit in the VPL projecting to hindlimb primary somatosensory cortex glutamatergic neurons (S1HLGlu) that mediated RIH. In vivo calcium imaging and multi-tetrode recordings revealed heightened S1HLGlu neuronal activity during RIH. Moreover, preoperative suppression of Cav3.1-dependent burst firing in VPLGlu neurons or chemogenetic inhibition of VPLGlu neuronal terminals in the S1HL abolished the increased S1HLGlu neuronal excitability while alleviating RIH. Our findings suggest that remifentanil induces postoperative hyperalgesia by upregulating T-type calcium channel-dependent burst firing in VPLGlu neurons to activate S1HLGlu neurons, thus revealing an ion channel-mediated neural circuit basis for RIH that can guide analgesic development.
Collapse
Affiliation(s)
- Yan Jin
- Stroke Center and Department of Neurology and,Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Yu Mao
- Stroke Center and Department of Neurology and,Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Danyang Chen
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Yingju Tai
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Rui Hu
- Department of Anesthesiology, The Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chen-Ling Yang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jing Zhou
- Department of head, neck, and breast Surgery, Western district of the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Lijian Chen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xuesheng Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Erwei Gu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chunhui Jia
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Zhi Zhang
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Wenjuan Tao
- Stroke Center and Department of Neurology and,Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
12
|
Chen L, Hu Y, Wang S, Cao K, Mai W, Sha W, Ma H, Zeng LH, Xu ZZ, Gao YJ, Duan S, Wang Y, Gao Z. mTOR-neuropeptide Y signaling sensitizes nociceptors to drive neuropathic pain. JCI Insight 2022; 7:159247. [PMID: 36194480 DOI: 10.1172/jci.insight.159247] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 09/29/2022] [Indexed: 12/15/2022] Open
Abstract
Neuropathic pain is a refractory condition that involves de novo protein synthesis in the nociceptive pathway. The mTOR is a master regulator of protein translation; however, mechanisms underlying its role in neuropathic pain remain elusive. Using the spared nerve injury-induced neuropathic pain model, we found that mTOR was preferentially activated in large-diameter dorsal root ganglion (DRG) neurons and spinal microglia. However, selective ablation of mTOR in DRG neurons, rather than microglia, alleviated acute neuropathic pain in mice. We show that injury-induced mTOR activation promoted the transcriptional induction of neuropeptide Y (Npy), likely via signal transducer and activator of transcription 3 phosphorylation. NPY further acted primarily on Y2 receptors (Y2R) to enhance neuronal excitability. Peripheral replenishment of NPY reversed pain alleviation upon mTOR removal, whereas Y2R antagonists prevented pain restoration. Our findings reveal an unexpected link between mTOR and NPY/Y2R in promoting nociceptor sensitization and neuropathic pain.
Collapse
Affiliation(s)
- Lunhao Chen
- Spine Lab, Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yaling Hu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Siyuan Wang
- Spine Lab, Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kelei Cao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Weihao Mai
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
| | - Weilin Sha
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, China
| | - Huan Ma
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Ling-Hui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Zhen-Zhong Xu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yong-Jing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, China
| | - Shumin Duan
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yue Wang
- Spine Lab, Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihua Gao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| |
Collapse
|
13
|
Li DY, Gao SJ, Sun J, Zhang LQ, Wu JY, Song FH, Liu DQ, Zhou YQ, Mei W. Targeting the nitric oxide/cGMP signaling pathway to treat chronic pain. Neural Regen Res 2022; 18:996-1003. [PMID: 36254980 PMCID: PMC9827765 DOI: 10.4103/1673-5374.355748] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Nitric oxide (NO)/cyclic guanosine 3',5'-monophosphate (cGMP) signaling has been shown to act as a mediator involved in pain transmission and processing. In this review, we summarize and discuss the mechanisms of the NO/cGMP signaling pathway involved in chronic pain, including neuropathic pain, bone cancer pain, inflammatory pain, and morphine tolerance. The main process in the NO/cGMP signaling pathway in cells involves NO activating soluble guanylate cyclase, which leads to subsequent production of cGMP. cGMP then activates cGMP-dependent protein kinase (PKG), resulting in the activation of multiple targets such as the opening of ATP-sensitive K+ channels. The activation of NO/cGMP signaling in the spinal cord evidently induces upregulation of downstream molecules, as well as reactive astrogliosis and microglial polarization which participate in the process of chronic pain. In dorsal root ganglion neurons, natriuretic peptide binds to particulate guanylyl cyclase, generating and further activating the cGMP/PKG pathway, and it also contributes to the development of chronic pain. Upregulation of multiple receptors is involved in activation of the NO/cGMP signaling pathway in various pain models. Notably the NO/cGMP signaling pathway induces expression of downstream effectors, exerting both algesic and analgesic effects in neuropathic pain and inflammatory pain. These findings suggest that activation of NO/cGMP signaling plays a constituent role in the development of chronic pain, and this signaling pathway with dual effects is an interesting and promising target for chronic pain therapy.
Collapse
Affiliation(s)
- Dan-Yang Li
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Shao-Jie Gao
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jia Sun
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Long-Qing Zhang
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jia-Yi Wu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Fan-He Song
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Dai-Qiang Liu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Ya-Qun Zhou
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China,Correspondence to: Wei Mei, ; Ya-Qun Zhou, .
| | - Wei Mei
- Correspondence to: Wei Mei, ; Ya-Qun Zhou, .
| |
Collapse
|
14
|
Avci O, Ozdemir E, Taskiran AS, Inan ZDS, Gursoy S. Metformin prevents morphine-induced apoptosis in rats with diabetic neuropathy: a possible mechanism for attenuating morphine tolerance. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:1449-1462. [PMID: 36050544 DOI: 10.1007/s00210-022-02283-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022]
Abstract
Morphine is a drug of choice for the treatment of severe and chronic pain, but tolerance to the antinociceptive effect limits its use. The development of tolerance to morphine has recently been associated with neuronal apoptosis. In this study, our aim was to investigate the effects of metformin on morphine-induced neuronal apoptosis and antinociceptive tolerance in diabetic rats. Three days of cumulative dosing were administered to establish morphine tolerance in rats. The antinociceptive effects of metformin (50 mg/kg) and test dose of morphine (5 mg/kg) were considered at 30-min intervals by thermal antinociceptive tests. To induce diabetic neuropathy, streptozotocin (STZ, 65 mg/kg) was injected intraperitoneally. ELISA kits were used to measure caspase-3, bax, and bcl-2 levels from dorsal root ganglion (DRG) tissue. Semi-quantitative scoring system was used to evaluate apoptotic cells with the the terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) method. The findings suggest that co-administration of metformin with morphine to diabetic rats showed a significant increase in antinociceptive effect compared to morphine alone. The antinociceptive tests indicated that metformin significantly attenuated morphine antinociceptive tolerance in diabetic rats. In addition, metformin decreased the levels of apoptotic proteins caspase 3 and Bax in DRG neurons, while significantly increased the levels of antiapoptotic Bcl-2. Semi-quantitative scoring showed that metformin provided a significant reduction in apoptotic cell counts in diabetic rats. These data revealed that metformin demonstrated antiapoptotic activity in diabetic rat DRG neurons and attenuated morphine tolerance. The antiapoptotic activity of metformin probably plays a significant role in reducing morphine tolerance.
Collapse
Affiliation(s)
- Onur Avci
- Department of Anesthesiology and Reanimation, Sivas Cumhuriyet University School of Medicine, 58140, Sivas, Turkey
| | - Ercan Ozdemir
- Department of Physiology, Sivas Cumhuriyet University School of Medicine, Sivas, Turkey.
| | - Ahmet Sevki Taskiran
- Department of Physiology, Sivas Cumhuriyet University School of Medicine, Sivas, Turkey
| | - Zeynep Deniz Sahin Inan
- Department of Histology and Embryology, Sivas Cumhuriyet University School of Medicine, Sivas, Turkey
| | - Sinan Gursoy
- Department of Anesthesiology and Reanimation, Sivas Cumhuriyet University School of Medicine, 58140, Sivas, Turkey
| |
Collapse
|
15
|
Zhang J, Zhang X, Li L, Bai L, Gao Y, Yang Y, Wang L, Qiao Y, Wang X, Xu JT. Activation of Double-Stranded RNA-Activated Protein Kinase in the Dorsal Root Ganglia and Spinal Dorsal Horn Regulates Neuropathic Pain Following Peripheral Nerve Injury in Rats. Neurotherapeutics 2022; 19:1381-1400. [PMID: 35655111 PMCID: PMC9587175 DOI: 10.1007/s13311-022-01255-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2022] [Indexed: 10/18/2022] Open
Abstract
Double-stranded RNA (dsRNA)-activated kinase (PKR) is an important component in inflammation and immune dysfunction. However, the role of PKR in neuropathic pain remains unclear. Here, we showed that lumbar 5 spinal nerve ligation (SNL) led to a significant increase in the level of phosphorylated PKR (p-PKR) in both the dorsal root ganglia (DRG) and spinal dorsal horn. Images of double immunofluorescence staining revealed that p-PKR was expressed in myelinated A-fibers, unmyelinated C-fibers, and satellite glial cells in the DRG. In the dorsal horn, p-PKR was located in neuronal cells, astrocytes, and microglia. Data from behavioral tests showed that intrathecal (i.t.) injection of 2-aminopurine (2-AP), a specific inhibitor of PKR activation, and PKR siRNA prevented the reductions in PWT and PWL following SNL. Established neuropathic pain was also attenuated by i.t. injection of 2-AP and PKR siRNA, which started on day 7 after SNL. Prior repeated i.t. injections of PKR siRNA prevented the SNL-induced degradation of IκBα and IκBβ in the cytosol and the nuclear translocation of nuclear factor κB (NF-κB) p65 in both the DRG and dorsal horn. Moreover, the SNL-induced increase in interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α) production was diminished by this treatment. Collectively, these results suggest that peripheral nerve injury-induced PKR activation via NF-κB signaling-regulated expression of proinflammatory cytokines in the DRG and dorsal horn contributes to the pathogenesis of neuropathic pain. Our findings suggest that pharmacologically targeting PKR might be an effective therapeutic strategy for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Xuan Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Liren Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Liying Bai
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou, 450052, China
| | - Yan Gao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Yin Yang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Li Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Yiming Qiao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Xueli Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Ji-Tian Xu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China.
- Neuroscience Research Institute, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China.
| |
Collapse
|
16
|
Opioids and Vitamin C: Known Interactions and Potential for Redox-Signaling Crosstalk. Antioxidants (Basel) 2022; 11:antiox11071267. [PMID: 35883757 PMCID: PMC9312198 DOI: 10.3390/antiox11071267] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 12/10/2022] Open
Abstract
Opioids are among the most widely used classes of pharmacologically active compounds both clinically and recreationally. Beyond their analgesic efficacy via μ opioid receptor (MOR) agonism, a prominent side effect is central respiratory depression, leading to systemic hypoxia and free radical generation. Vitamin C (ascorbic acid; AA) is an essential antioxidant vitamin and is involved in the recycling of redox cofactors associated with inflammation. While AA has been shown to reduce some of the negative side effects of opioids, the underlying mechanisms have not been explored. The present review seeks to provide a signaling framework under which MOR activation and AA may interact. AA can directly quench reactive oxygen and nitrogen species induced by opioids, yet this activity alone does not sufficiently describe observations. Downstream of MOR activation, confounding effects from AA with STAT3, HIF1α, and NF-κB have the potential to block production of antioxidant proteins such as nitric oxide synthase and superoxide dismutase. Further mechanistic research is necessary to understand the underlying signaling crosstalk of MOR activation and AA in the amelioration of the negative, potentially fatal side effects of opioids.
Collapse
|
17
|
Fang Y, Peng X, Bu H, Jia X, Gao F, Liu C. Chemokine CXCL10 regulates pain behaviors via PI3K-AKT signaling pathway in mice. Neuropeptides 2022; 93:102243. [PMID: 35344847 DOI: 10.1016/j.npep.2022.102243] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/16/2021] [Accepted: 01/11/2022] [Indexed: 11/20/2022]
Abstract
The analgesic efficacy of morphine can be affected by a variety of factors. Our previous studies demonstrated that chemokine (CXC motif) ligand 10 (CXCL10) could induce algesia directly and attenuate the analgesic effect produced by a single dose of morphine. However, the underlying mechanism remains unclear. In the present study, we aimed to further investigate the mechanism of CXCL10-mediated inhibition on morphine analgesic effect. According to our findings, recombinant CXCL10 protein (rmCXCL10) could increase the phosphorylation of serine-threonine kinase AKT reduced by morphine in spinal cord. Blocking AKT activation by phosphoinositide 3-kinase (PI3K) inhibitor could effectively attenuate CXCL10-induced algesia, and reverse the decrease of paw withdrawal thresholds caused by the co-administration of morphine and rmCXCL10. Furthermore, rmCXCL10 could enhance the spinal expression of pro-inflammatory cytokines, including TNF-α, IL-6, and IL-1β, which could be blocked by PI3K inhibitor. In summary, these findings suggest that PI3K-AKT signaling pathway mediates the effect of CXCL10 on the regulation of morphine analgesia and the release of cytokines in spinal cord. Our study provides a new insight into the mechanism of chemokine-relative pain regulation.
Collapse
Affiliation(s)
- Yan Fang
- Department of Anesthesiology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiaoling Peng
- Department of Anesthesiology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Huilian Bu
- Department of Pain management, First affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xiaoqian Jia
- Department of Anesthesiology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Feng Gao
- Department of Anesthesiology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Cheng Liu
- Department of Anesthesiology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
18
|
He L, Xu W, Zhang C, Ding Z, Guo Q, Zou W, Wang J. Dysregulation of Vesicular Glutamate Transporter VGluT2 via BDNF/TrkB Pathway Contributes to Morphine Tolerance in Mice. Front Pharmacol 2022; 13:861786. [PMID: 35559256 PMCID: PMC9086316 DOI: 10.3389/fphar.2022.861786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
Morphine is widely used in the treatment of moderate to severe pain. Long-term use of morphine leads to various adverse effects, such as tolerance and hyperalgesia. Vesicular glutamate transporter 2 (VGluT2) accumulates glutamate into synaptic vesicles and plays multiple roles in the central nervous system. However, the specific role of VGluT2 in morphine tolerance has not been fully elucidated. Here, we investigated the regulatory role of VGluT2 in morphine tolerance and assessed the potential role of the brain-derived neurotrophic factor (BDNF)/tyrosine kinase B (TrkB) pathway in VGluT2 mediated morphine antinociceptive tolerance in mice. In the present study, we found that VGluT2 is upregulated in the spinal cord after the development of morphine tolerance. Furthermore, inhibition of VGluT2 with its antagonist (Chicago sky blue 6 B, CSB6B) or knockdown of VGluT2 by lentivirus restored the analgesic effect of morphine, suppressed the activation of astrocytes and microglia, and decreased glial-derived pro-inflammatory cytokines. Overexpression of VGluT2 by lentivirus facilitated morphine tolerance and mechanical hyperalgesia. In addition, we found the expression of BDNF is correlated with VGluT2 expression in the spinal cord after chronic morphine administration. Intrathecal injection of the BDNF/TrkB pathway antagonist K252a attenuated the development of morphine tolerance and decreased the expression of VGluT2 in the spinal cord, which suggested the BDNF/TrkB pathway participates in the regulation of VGluT2 in morphine tolerance. This study elucidates the functional capability of VGluT2 in modulating morphine tolerance and identifies a novel mechanism and promising therapeutic target for morphine tolerance.
Collapse
Affiliation(s)
- Liqiong He
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xu
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Chengliang Zhang
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhuofeng Ding
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wangyuan Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
19
|
Huang T, Xiao Y, Zhang Y, Wang C, Chen X, Li Y, Ge Y, Gao J. miR‑223 ameliorates thalamus hemorrhage‑induced central poststroke pain via targeting NLRP3 in a mouse model. Exp Ther Med 2022; 23:353. [PMID: 35493427 PMCID: PMC9019782 DOI: 10.3892/etm.2022.11280] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 01/28/2022] [Indexed: 11/24/2022] Open
Abstract
Central poststroke pain (CPSP) is a central neuropathic pain syndrome that occurs following a stroke and mainly manifests as pain and paresthesia in the body region corresponding to the brain injury area. At present, due to the lack of clinical attention given to CPSP, patients suffer from long-term pain that seriously affects their quality of life. Current literature indicates that microRNA (miR)-223 can impede inflammation and prevent collateral damage. The NLR family pyrin domain containing 3 (NLRP3) inflammasome induces IL-18 and IL-1β secretion and maturation and participates in the inflammatory response. Previous evidence has confirmed that miR-223 can negatively regulate NLRP3 in the development of inflammatory responses. However, whether the miR-223 targeting of NLRP3 is involved in CPSP remains unclear. In the present study, the expression of miR-223 was detected by reverse transcription-quantitative PCR analysis. The expression levels of NLRP3, caspase-1, ASC, IL-18, IL-1β, ERK1/2, p-ERK1/2 and GFAP were detected by western blot analysis. The results demonstrated that thalamic hemorrhagic stroke triggered by microinjection of collagenase Ⅳ (Coll IV) into the ventral posterior lateral (VPL) nucleus results in pain hypersensitivity. miR-223 expression level were significantly reduced in the CPSP model. The expression levels of NLRP3, caspase-1, ASC, IL-18 and IL-1β were significantly increased in the CPSP model. The expression level of GFAP was detected to determine astrocyte activation. The results demonstrated that astrocyte activation induced by Coll IV produced a CPSP model. The p-ERK1/2 expression level was demonstrated to be significantly increased in the CPSP model. The introduction of an miR-223 agomir significantly attenuated thalamic pain and significantly decreased the levels of NLRP3, caspase-1, ASC and proinflammatory cytokines (IL-18 and IL-1β). Furthermore, introducing a miR-223 antagomir into the VPL nucleus of naïve mice mimicked thalamic pain and significantly increased the levels of NLRP3, caspase-1, ASC and proinflammatory cytokine levels (IL-18 and IL-1β). These results indicated that miR-223 inhibited NLRP3 inflammasome activity (caspase-1, NLRP3 and ASC), which ameliorated thalamus hemorrhage-induced CPSP in mice via NLRP3 downregulation. In conclusion, these results may determine the mechanisms underlying CPSP and facilitate development of targeted therapy for CPSP.
Collapse
Affiliation(s)
- Tianfeng Huang
- Department of Anesthesiology, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225001, P.R. China
| | - Yinggang Xiao
- Department of Anesthesiology, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225001, P.R. China
| | - Yang Zhang
- Department of Anesthesiology, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225001, P.R. China
| | - Cunjin Wang
- Department of Anesthesiology, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225001, P.R. China
| | - Xiaoping Chen
- Department of Anesthesiology, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225001, P.R. China
| | - Yong Li
- Department of Anesthesiology, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225001, P.R. China
| | - Yali Ge
- Department of Anesthesiology, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225001, P.R. China
| | - Ju Gao
- Department of Anesthesiology, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225001, P.R. China
| |
Collapse
|
20
|
Deng M, Zhang Z, Xing M, Liang X, Li Z, Wu J, Jiang S, Weng Y, Guo Q, Zou W. LncRNA MRAK159688 facilitates morphine tolerance by promoting REST-mediated inhibition of mu opioid receptor in rats. Neuropharmacology 2022; 206:108938. [PMID: 34982972 DOI: 10.1016/j.neuropharm.2021.108938] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/17/2021] [Accepted: 12/28/2021] [Indexed: 12/11/2022]
Abstract
Morphine tolerance (MT) caused by the long-term use of morphine is a major medical problem. The molecular mechanism of morphine tolerance remains elusive. Here, we established a morphine tolerance model in rats and verified whether the long noncoding RNA (lncRNA) MRAK159688 is involved in morphine tolerance and its specific molecular mechanism. We show the significant upregulation of MRAK159688 expression in the spinal cord of morphine-tolerant rats. Overexpression of MRAK159688 by a lentivirus reduces the analgesic efficacy of morphine and induces pain behavior. Downregulation of MRAK159688 using a small interfering RNA (siRNA) attenuates the formation of morphine tolerance, partially reverses the development of morphine tolerance and alleviates morphine-induced hyperalgesia. MRAK159688 is located in the nucleus and cytoplasm of neurons, and it colocalizes with repressor element-1 silencing transcription factor (REST) in the nucleus. MRAK159688 potentiates the expression and function of REST, thereby inhibiting the expression of mu opioid receptor (MOR) and subsequently inducing morphine tolerance. Moreover, REST overexpression blocks the effects of MRAK159688 siRNA on relieving morphine tolerance. In general, chronic morphine administration-mediated upregulation of MRAK159688 in the spinal cord contributes to morphine tolerance and hyperalgesia by promoting REST-mediated inhibition of MOR. MRAK159688 downregulation may represent a novel RNA-based therapy for morphine tolerance.
Collapse
Affiliation(s)
- Meiling Deng
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zengli Zhang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; Department of Anesthesiology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, 300000, China
| | - Manyu Xing
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xia Liang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Zhengyiqi Li
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jing Wu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Shasha Jiang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yingqi Weng
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Wangyuan Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
21
|
Wan L, Jia RM, Ji LL, Qin XM, Hu L, Hu F, Han Y, Pan YB, Jiang CY, Liu WT. AMPK-autophagy-mediated inhibition of microRNA-30a-5p alleviates morphine tolerance via SOCS3-dependent neuroinflammation suppression. J Neuroinflammation 2022; 19:25. [PMID: 35093117 PMCID: PMC8800317 DOI: 10.1186/s12974-022-02384-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/10/2022] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The development of morphine tolerance is a clinical challenge for managing severe pain. Studies have shown that neuroinflammation is a critical aspect for the development of analgesic tolerance. We found that AMPK-autophagy activation could suppress neuroinflammation and improve morphine tolerance via the upregulation of suppressor of cytokine signaling 3 (SOCS3) by inhibiting the processing and maturation of microRNA-30a-5p. METHODS CD-1 mice were utilized for the tail-flick test to evaluate morphine tolerance. The microglial cell line BV-2 was utilized to investigate the mechanism of AMPK-autophagy-mediated posttranscriptional regulation of SOCS3. Proinflammatory cytokines were measured by western blotting and real-time PCR. The levels of SOCS3 and miRNA-processing enzymes were evaluated by western blotting, real-time PCR and immunofluorescence staining. RESULTS Based on experimental verification, miRNA-30a-5p could negatively regulate SOCS3. The AMPK activators AICAR, resveratrol and metformin downregulated miRNA-30a-5p. We found that AMPK activators specifically inhibited the processing and maturation of miRNA-30a-5p in microglia by degrading DICER and AGO2 via autophagy. Furthermore, a miRNA-30a-5p inhibitor significantly improved morphine tolerance via upregulation of SCOS3 in mice. It markedly increased the level of SOCS3 in the spinal cord of mice and subsequently inhibited morphine-induced phosphorylation of NF-κB p65. In addition, a miRNA-30a-5p inhibitor decreased the levels of IL-1β and TNF-α caused by morphine in microglia. CONCLUSION AMPK-autophagy activation suppresses neuroinflammation and improves morphine tolerance via the upregulation of SOCS3 by inhibiting miRNA-30a-5p.
Collapse
Affiliation(s)
- Li Wan
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Ru-Meng Jia
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Lu-Lu Ji
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Xin-Miao Qin
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Liang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Fan Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yuan Han
- Department of Anesthesiology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Yin-Bing Pan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Chun-Yi Jiang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| | - Wen-Tao Liu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
22
|
Kouraki A, Doherty M, Fernandes GS, Zhang W, Walsh DA, Kelly A, Valdes AM. Different genes may be involved in distal and local sensitisation: a genome-wide gene-based association study and meta-analysis. Eur J Pain 2021; 26:740-753. [PMID: 34958702 PMCID: PMC9303629 DOI: 10.1002/ejp.1902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/11/2021] [Accepted: 12/25/2021] [Indexed: 11/22/2022]
Abstract
Background Neuropathic pain symptoms and signs of increased pain sensitization in osteoarthritis (OA) patients may explain persistent pain after total joint replacement (TJR). Therefore, identifying genetic markers associated with pain sensitization and neuropathic‐like pain phenotypes could be clinically important in identifying targets for early intervention. Methods We performed a genome‐wide gene‐based association study (GWGAS) using pressure pain detection thresholds (PPTs) from distal pain‐free sites (anterior tibia), a measure of distal sensitization, and from proximal pain‐affected sites (lateral joint line), a measure of local sensitization, in 320 knee OA participants from the Knee Pain and related health in the Community (KPIC) cohort. We next performed gene‐based fixed‐effects meta‐analysis of PPTs and a neuropathic‐like pain phenotype using genome‐wide association study (GWAS) data from KPIC and from an independent cohort of 613 post‐TJR participants, respectively. Results The most significant genes associated with distal and local sensitization were OR5B3 and BRDT, respectively. We also found previously identified neuropathic pain‐associated genes—KCNA1, MTOR, ADORA1 and SCN3B—associated with PPT at the anterior tibia and an inflammatory pain gene—PTAFR—associated with PPT at the lateral joint line. Meta‐analysis results of anterior tibia and neuropathic‐like pain phenotypes revealed genes associated with bone morphogenesis, neuro‐inflammation, obesity, type 2 diabetes, cardiovascular disease and cognitive function. Conclusions Overall, our results suggest that different biological processes might be involved in distal and local sensitization, and common genetic mechanisms might be implicated in distal sensitization and neuropathic‐like pain. Future studies are needed to replicate these findings. Significance To the best of our knowledge, this is the first GWAS for pain sensitization and the first gene‐based meta‐analysis of pain sensitization and neuropathic‐like pain. Higher pain sensitization and neuropathic pain symptoms are associated with persistent pain after surgery hence, identifying genetic biomarkers and molecular pathways associated with these traits is clinically relevant.
Collapse
Affiliation(s)
- A Kouraki
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| | - M Doherty
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,Pain Centre Versus Arthritis, University of Nottingham, Nottingham, NG5 1PB, United Kingdom.,Versus Arthritis Centre for Sports, Exercise and Osteoarthritis, University of Nottingham, Nottingham, NG7 2UH, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| | - G S Fernandes
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 6EH, United Kingdom
| | - W Zhang
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,Pain Centre Versus Arthritis, University of Nottingham, Nottingham, NG5 1PB, United Kingdom.,Versus Arthritis Centre for Sports, Exercise and Osteoarthritis, University of Nottingham, Nottingham, NG7 2UH, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| | - D A Walsh
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,Pain Centre Versus Arthritis, University of Nottingham, Nottingham, NG5 1PB, United Kingdom.,Versus Arthritis Centre for Sports, Exercise and Osteoarthritis, University of Nottingham, Nottingham, NG7 2UH, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| | - A Kelly
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| | - A M Valdes
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,Pain Centre Versus Arthritis, University of Nottingham, Nottingham, NG5 1PB, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| |
Collapse
|
23
|
Ma L, Yu L, Jiang BC, Wang J, Guo X, Huang Y, Ren J, Sun N, Gao DS, Ding H, Lu J, Zhou H, Zou L, Gao Y, Wang L, Sun K, Ming Y, Meng Z, Tao YX, Yan M. ZNF382 controls mouse neuropathic pain via silencer-based epigenetic inhibition of Cxcl13 in DRG neurons. J Exp Med 2021; 218:e20210920. [PMID: 34762123 PMCID: PMC8590274 DOI: 10.1084/jem.20210920] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/06/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023] Open
Abstract
Nerve injury-induced changes of gene expression in dorsal root ganglion (DRG) are critical for neuropathic pain genesis. However, how these changes occur remains elusive. Here we report the down-regulation of zinc finger protein 382 (ZNF382) in injured DRG neurons after nerve injury. Rescuing this down-regulation attenuates nociceptive hypersensitivity. Conversely, mimicking this down-regulation produces neuropathic pain symptoms, which are alleviated by C-X-C motif chemokine 13 (CXCL13) knockdown or its receptor CXCR5 knockout. Mechanistically, an identified cis-acting silencer at distal upstream of the Cxcl13 promoter suppresses Cxcl13 transcription via binding to ZNF382. Blocking this binding or genetically deleting this silencer abolishes the ZNF382 suppression on Cxcl13 transcription and impairs ZNF382-induced antinociception. Moreover, ZNF382 down-regulation disrupts the repressive epigenetic complex containing histone deacetylase 1 and SET domain bifurcated 1 at the silencer-promoter loop, resulting in Cxcl13 transcriptional activation. Thus, ZNF382 down-regulation is required for neuropathic pain likely through silencer-based epigenetic disinhibition of CXCL13, a key neuropathic pain player, in DRG neurons.
Collapse
Affiliation(s)
- Longfei Ma
- Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lina Yu
- Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Bao-Chun Jiang
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Jiangsu, China
| | - Jingkai Wang
- Department of Orthopedics, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xinying Guo
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Yangyuxin Huang
- Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jinxuan Ren
- Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Na Sun
- Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Dave Schwinn Gao
- Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Ding
- Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jianan Lu
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hang Zhou
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lijing Zou
- Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yibo Gao
- Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lieju Wang
- Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Sun
- Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yue Ming
- Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zhipeng Meng
- Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ
| | - Min Yan
- Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
24
|
Li L, Bai L, Yang K, Zhang J, Gao Y, Jiang M, Yang Y, Zhang X, Wang L, Wang X, Qiao Y, Xu JT. KDM6B epigenetically regulated-interleukin-6 expression in the dorsal root ganglia and spinal dorsal horn contributes to the development and maintenance of neuropathic pain following peripheral nerve injury in male rats. Brain Behav Immun 2021; 98:265-282. [PMID: 34464689 DOI: 10.1016/j.bbi.2021.08.231] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 08/17/2021] [Accepted: 08/21/2021] [Indexed: 12/20/2022] Open
Abstract
The lysine specific demethylase 6B (KDM6B) has been implicated as a coregulator in the expression of proinflammatory mediators, and in the pathogenesis of inflammatory and arthritic pain. However, the role of KDM6B in neuropathic pain has yet to be studied. In the current study, the neuropathic pain was determined by assessing the paw withdrawal threshold (PWT) and paw withdrawal latency (PWL) following lumbar 5 spinal nerve ligation (SNL) in male rats. Immunohistochemistry, Western blotting, qRT-PCR, and chromatin immunoprecipitation (ChIP)-PCR assays were performed to investigate the underlying mechanisms. Our results showed that SNL led to a significant increase in KDM6B mRNA and protein in the ipsilateral L4/5 dorsal root ganglia (DRG) and spinal dorsal horn; and this increase correlated a markedly reduction in the level of H3K27me3 methylation in the same tissue. Double immunofluorescence staining revealed that the KDM6B expressed in myelinated A- and unmyelinated C-fibers in the DRG; and located in neuronal cells, astrocytes, and microglia in the dorsal horn. Behavioral data showed that SNL-induced mechanical allodynia and thermal hyperalgesia were impaired by the treatment of prior to i.t. injection of GSK-J4, a specific inhibitor of KDM6B, or KDM6B siRNA. Both microinjection of AAV2-EGFP-KDM6B shRNA in the lumbar 5 dorsal horn and sciatic nerve, separately, alleviated the neuropathic pain following SNL. The established neuropathic pain was also partially attenuated by repeat i.t. injections of GSK-J4 or KDM6B siRNA, started on day 7 after SNL. SNL also resulted in a remarkable increased expression of interleukin-6 (IL-6) in the DRG and dorsal horn. But this increase was dramatically inhibited by i.t. injection of GSK-J4 and KDM6B siRNA; and suppressed by prior to microinjection of AAV2-EGFP-KDM6B shRNA in the dorsal horn and sciatic nerve. Results of ChIP-PCR assay showed that SNL-induced enhanced binding of STAT3 with IL-6 promoter was inhibited by prior to i.t. injection of GSK-J4. Meanwhile, the level of H3K27me3 methylation was also decreased by the treatment. Together, our results indicate that SNL-induced upregulation of KDM6B via demethylating H3K27me3 facilitates the binding of STAT3 with IL-6 promoter, and subsequently mediated-increase in the expression of IL-6 in the DRG and dorsal horn contributes to the development and maintenance of neuropathic pain. Targeting KDM6B might a promising therapeutic strategy to treatment of chronic pain.
Collapse
Affiliation(s)
- Liren Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Liying Bai
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Kangli Yang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Jian Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Yan Gao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China; Neuroscience Research Institute, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Mingjun Jiang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Yin Yang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Xuan Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Li Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Xueli Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Yiming Qiao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Ji-Tian Xu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China; Neuroscience Research Institute, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China.
| |
Collapse
|
25
|
Huang T, Li Y, Hu W, Yu D, Gao J, Yang F, Xu Y, Wang Z, Zong L. Dexmedetomidine attenuates haemorrhage-induced thalamic pain by inhibiting the TLR4/NF-κB/ERK1/2 pathway in mice. Inflammopharmacology 2021; 29:1751-1760. [PMID: 34643849 PMCID: PMC8643300 DOI: 10.1007/s10787-021-00877-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/18/2021] [Indexed: 10/31/2022]
Abstract
BACKGROUND Thalamic pain, a neuropathic pain syndrome, frequently occurs after stroke. This research aimed to investigate the effect of dexmedetomidine (DEX) on thalamic pain. METHODS The cellular localization of the TLR4 protein was determined by immunostaining. The expression of Iba1, GFAP and protein associated with the TLR4/NF-κB/ERK1/2 pathway was measured by Western blotting. Continuous pain hypersensitivity was evaluated by behavioural tests. The results were analysed by one-way ANOVA, two-way ANOVA and Tukey's post hoc test. RESULTS The results demonstrated that DEX obviously alleviated thalamic pain induced by haemorrhage on the ipsilateral side and delayed the development of pain hypersensitivity. Furthermore, the expression levels of Iba1, GFAP and proteins associated with the TLR4/NF-κB/ERK1/2 signalling pathway were greatly increased in mice with thalamic pain, but these effects were reversed by DEX. CONCLUSION Our findings suggest that DEX alleviates the inflammatory response during thalamic pain through the TLR4/NF-κB/ERK1/2 signalling pathway and might be a potential therapeutic agent for thalamic pain.
Collapse
Affiliation(s)
- Tianfeng Huang
- Department of Anesthesiology, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital Affiliated with Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Yong Li
- Department of Anesthesiology, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital Affiliated with Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Wenqing Hu
- Department of Gastrointestinal Surgery, Changzhi People's Hospital, The Affiliated Hospital of Changzhi Medical College, Shanxi, No. 502 Changxing Middle Road, Luzhou District, Changzhi, 046000, People's Republic of China
| | - Dapeng Yu
- Department of Anesthesiology, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital Affiliated with Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Ju Gao
- Department of Anesthesiology, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital Affiliated with Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Fan Yang
- Department of Central Laboratory, Changzhi People's Hospital, Shanxi, The Affiliated Hospital of Changzhi Medical College, Changzhi, People's Republic of China
| | - Yingying Xu
- Department of General Surgery, Yizheng People's Hospital, Clinical Medical College, Yangzhou University, No. 61 Dongyuan South Road, Yangzhou, 211400, Jiangsu, People's Republic of China.
| | - Zehua Wang
- Department of Anesthesiology, Heji Hospital Affiliated To Changzhi Medical College, No. 271 Taihang East Road, Changzhi, 046000, Shanxi, People's Republic of China.
| | - Liang Zong
- Department of Gastrointestinal Surgery, Changzhi People's Hospital, The Affiliated Hospital of Changzhi Medical College, Shanxi, No. 502 Changxing Middle Road, Luzhou District, Changzhi, 046000, People's Republic of China. .,Department of Central Laboratory, Changzhi People's Hospital, Shanxi, The Affiliated Hospital of Changzhi Medical College, Changzhi, People's Republic of China.
| |
Collapse
|
26
|
Hu XM, Yang W, Zhang MT, Du LX, Tian JH, Zhu JY, Chen Y, Hai F, Liu SB, Mao-Ying QL, Chu YX, Zhou H, Wang YQ, Mi WL. Glial IL-33 signaling through an ST2-to-CXCL12 pathway in the spinal cord contributes to morphine-induced hyperalgesia and tolerance. Sci Signal 2021; 14:eabe3773. [PMID: 34516755 DOI: 10.1126/scisignal.abe3773] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Xue-Ming Hu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wei Yang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Meng-Ting Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Department of Encephalopathy, Jinhua Hospital of Traditional Chinese Medicine, Jinhua, Zhejiang 321017, China
| | - Li-Xia Du
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jia-He Tian
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jian-Yu Zhu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yu Chen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Feng Hai
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shen-Bin Liu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Qi-Liang Mao-Ying
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai 200032, China
| | - Yu-Xia Chu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai 200032, China
| | - Hong Zhou
- Department of Immunology, Anhui Medical University, Hefei, Anhui 230032, China
| | - Yan-Qing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai 200032, China
| | - Wen-Li Mi
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai 200032, China
| |
Collapse
|
27
|
Wang S, Dai Y. Roles of AMPK and Its Downstream Signals in Pain Regulation. Life (Basel) 2021; 11:life11080836. [PMID: 34440581 PMCID: PMC8401922 DOI: 10.3390/life11080836] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/06/2021] [Accepted: 08/12/2021] [Indexed: 12/20/2022] Open
Abstract
Pain is an unpleasant sensory and emotional state that decreases quality of life. A metabolic sensor, adenosine monophosphate-activated protein kinase (AMPK), which is ubiquitously expressed in mammalian cells, has recently attracted interest as a new target of pain research. Abnormal AMPK expression and function in the peripheral and central nervous systems are associated with various types of pain. AMPK and its downstream kinases participate in the regulation of neuron excitability, neuroinflammation and axonal and myelin regeneration. Numerous AMPK activators have reduced pain behavior in animal models. The current understanding of pain has been deepened by AMPK research, but certain issues, such as the interactions of AMPK at each step of pain regulation, await further investigation. This review examines the roles of AMPK and its downstream kinases in neurons and non-neuronal cells, as well as their contribution to pain regulation.
Collapse
Affiliation(s)
- Shenglan Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe 650-8530, Japan
- Correspondence: (S.W.); (Y.D.); Tel.: +86-10-53912197 (S.W.); +81-78-304-3147 (Y.D.)
| | - Yi Dai
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe 650-8530, Japan
- Traditional Medicine Research Center, Chinese Medicine Confucius Institute, Hyogo College of Medicine, Kobe 663-8501, Japan
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Nishinomiya 663-8501, Japan
- Correspondence: (S.W.); (Y.D.); Tel.: +86-10-53912197 (S.W.); +81-78-304-3147 (Y.D.)
| |
Collapse
|
28
|
Pan Z, Du S, Wang K, Guo X, Mao Q, Feng X, Huang L, Wu S, Hou B, Chang Y, Liu T, Chen T, Li H, Bachmann T, Bekker A, Hu H, Tao Y. Downregulation of a Dorsal Root Ganglion-Specifically Enriched Long Noncoding RNA is Required for Neuropathic Pain by Negatively Regulating RALY-Triggered Ehmt2 Expression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004515. [PMID: 34383386 PMCID: PMC8356248 DOI: 10.1002/advs.202004515] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/19/2021] [Indexed: 05/07/2023]
Abstract
Nerve injury-induced maladaptive changes of gene expression in dorsal root ganglion (DRG) neurons contribute to neuropathic pain. Long non-coding RNAs (lncRNAs) are emerging as key regulators of gene expression. Here, a conserved lncRNA is reported, named DRG-specifically enriched lncRNA (DS-lncRNA) for its high expression in DRG neurons. Peripheral nerve injury downregulates DS-lncRNA in injured DRG due, in part, to silencing of POU domain, class 4, transcription factor 3, a transcription factor that interacts with the DS-lncRNA gene promoter. Rescuing DS-lncRNA downregulation blocks nerve injury-induced increases in the transcriptional cofactor RALY-triggered DRG Ehmt2 mRNA and its encoding G9a protein, reverses the G9a-controlled downregulation of opioid receptors and Kcna2 in injured DRG, and attenuates nerve injury-induced pain hypersensitivities in male mice. Conversely, DS-lncRNA downregulation increases RALY-triggered Ehmt2/G9a expression and correspondingly decreases opioid receptor and Kcna2 expression in DRG, leading to neuropathic pain symptoms in male mice in the absence of nerve injury. Mechanistically, downregulated DS-lncRNA promotes more binding of increased RALY to RNA polymerase II and the Ehmt2 gene promoter and enhances Ehmt2 transcription in injured DRG. Thus, downregulation of DS-lncRNA likely contributes to neuropathic pain by negatively regulating the expression of RALY-triggered Ehmt2/G9a, a key neuropathic pain player, in DRG neurons.
Collapse
Affiliation(s)
- Zhiqiang Pan
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New JerseyNewarkNJ07103USA
| | - Shibin Du
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New JerseyNewarkNJ07103USA
| | - Kun Wang
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New JerseyNewarkNJ07103USA
| | - Xinying Guo
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New JerseyNewarkNJ07103USA
| | - Qingxiang Mao
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New JerseyNewarkNJ07103USA
| | - Xiaozhou Feng
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New JerseyNewarkNJ07103USA
| | - Lina Huang
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New JerseyNewarkNJ07103USA
| | - Shaogen Wu
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New JerseyNewarkNJ07103USA
| | - Bailing Hou
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New JerseyNewarkNJ07103USA
| | - Yun‐Juan Chang
- The Office of Advanced Research ComputingRutgersThe State University of New JerseyNewarkNJ07103USA
| | - Tong Liu
- Center for Advanced Proteomics ResearchDepartments of Biochemistry, Microbiology & Molecular GeneticsNew Jersey Medical School, RutgersThe State University of New JerseyNewarkNJ07103USA
| | - Tong Chen
- Center for Advanced Proteomics ResearchDepartments of Biochemistry, Microbiology & Molecular GeneticsNew Jersey Medical School, RutgersThe State University of New JerseyNewarkNJ07103USA
| | - Hong Li
- Center for Advanced Proteomics ResearchDepartments of Biochemistry, Microbiology & Molecular GeneticsNew Jersey Medical School, RutgersThe State University of New JerseyNewarkNJ07103USA
| | - Thomas Bachmann
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New JerseyNewarkNJ07103USA
| | - Alex Bekker
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New JerseyNewarkNJ07103USA
| | - Huijuan Hu
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New JerseyNewarkNJ07103USA
- Department of Physiology, Pharmacology & NeuroscienceNew Jersey Medical SchoolRutgersThe State University of New JerseyNewarkNJ07103USA
| | - Yuan‐Xiang Tao
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New JerseyNewarkNJ07103USA
- Department of Physiology, Pharmacology & NeuroscienceNew Jersey Medical SchoolRutgersThe State University of New JerseyNewarkNJ07103USA
- Department of Cell Biology & Molecular MedicineNew Jersey Medical SchoolRutgersThe State University of New JerseyNewarkNJ07103USA
| |
Collapse
|
29
|
Gledhill LJ, Babey AM. Synthesis of the Mechanisms of Opioid Tolerance: Do We Still Say NO? Cell Mol Neurobiol 2021; 41:927-948. [PMID: 33704603 DOI: 10.1007/s10571-021-01065-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 02/12/2021] [Indexed: 10/21/2022]
Abstract
The use of morphine as a first-line agent for moderate-to-severe pain is limited by the development of analgesic tolerance. Initially opioid receptor desensitization in response to repeated stimulation, thought to underpin the establishment of tolerance, was linked to a compensatory increase in adenylate cyclase responsiveness. The subsequent demonstration of cross-talk between N-methyl-D-aspartate (NMDA) glutamate receptors and opioid receptors led to the recognition of a role for nitric oxide (NO), wherein blockade of NO synthesis could prevent tolerance developing. Investigations of the link between NO levels and opioid receptor desensitization implicated a number of events including kinase recruitment and peroxynitrite-mediated protein regulation. Recent experimental advances and the identification of new cellular constituents have expanded the potential signaling candidates to include unexpected, intermediary compounds not previously linked to this process such as zinc, histidine triad nucleotide-binding protein 1 (HINT1), micro-ribonucleic acid (mi-RNA) and regulator of G protein signaling Z (RGSZ). A further complication is a lack of consistency in the protocols used to create tolerance, with some using acute methods measured in minutes to hours and others using days. There is also an emphasis on the cellular changes that are extant only after tolerance has been established. Although a review of the literature demonstrates a lack of spatio-temporal detail, there still appears to be a pivotal role for nitric oxide, as well as both intracellular and intercellular cross-talk. The use of more consistent approaches to verify these underlying mechanism(s) could provide an avenue for targeted drug development to rescue opioid efficacy.
Collapse
Affiliation(s)
- Laura J Gledhill
- CURA Pharmacy, St. John of God Hospital, Bendigo, VIC, 3550, Australia
| | - Anna-Marie Babey
- Faculty of Medicine and Health, University of New England, Armidale, NSW, 2351, Australia.
| |
Collapse
|
30
|
Jiang M, Zhang X, Wang X, Xu F, Zhang J, Li L, Xie X, Wang L, Yang Y, Xu JT. MicroRNA-124-3p attenuates the development of nerve injury-induced neuropathic pain by targeting early growth response 1 in the dorsal root ganglia and spinal dorsal horn. J Neurochem 2021; 158:928-942. [PMID: 34008206 DOI: 10.1111/jnc.15433] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 12/11/2022]
Abstract
Emerging evidence indicates the early growth response 1 (Egr1) plays an important role in the pathogenesis of chronic pain. However, the regulation of Egr1 expression in the DRG and spinal cord in neuropathic pain remains unclear. In the current study, the neuropathic pain was conducted by lumber 5 spinal nerve ligation (SNL) in rats. The role of miR-124-3p in Egr1 expression was examined. Our results showed that the SNL led to a significant increase in the expression of Egr1 mRNA and protein in the DRG and dorsal horn. This increased expression of Egr1 correlated with a reduction of miR-124-3p in the same region. Prior i.t. injection of Egr1 decoy AYX1 inhibited the expression of Egr1 and attenuated the neuropathic pain-like hypersensitivity following SNL. The dual-luciferase reporter assay revealed the luciferase activity of the Egr1 3'-UTR plasmid was inhibited by the miR-124-3p agomir. But this inhibition was completely reversed in the mutant 3'-UTR Egr1 group. In vivo, the SNL-induced behavioral signs of neuropathic pain and the increases in Egr1 mRNA and protein in the DRG and dorsal horn were prevented by prior to i.t. injection of miR-124-3p agomir. While, i.t. injection of miR-124-3p antagomir in naïve rats resulted in mechanical allodynia and thermal hyperalgesia and an overexpression of Egr1 in the DRG and dorsal horn. Together, our results suggest that the miR-124-3p-regulated Egr1 expression in the DRG and dorsal horn contributes to the development of neuropathic pain. Targeting miR-124-3p might be a promising therapeutic strategy in the treatment of chronic pain.
Collapse
Affiliation(s)
- Mingjun Jiang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xuan Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xueli Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Feng Xu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.,Department of Renal Transplantation, The Seventh People's Hospital of Zhengzhou, Zhengzhou, China
| | - Jian Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Liren Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiaohang Xie
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | | | - Yin Yang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ji-Tian Xu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.,Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
31
|
BDNF Participates in Chronic Constriction Injury-Induced Neuropathic Pain via Transcriptionally Activating P2X 7 in Primary Sensory Neurons. Mol Neurobiol 2021; 58:4226-4236. [PMID: 33963520 DOI: 10.1007/s12035-021-02410-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/27/2021] [Indexed: 12/11/2022]
Abstract
Neuropathic pain, resulting from the pathological changes of the somatosensory nervous system, remains a severe public health problem worldwide. The effect of treatment targeting neuropathic pain is very limited, as the underlying mechanism of neuropathic pain is largely unknown. In this study, we demonstrated that the expression level of brain-derived neurotrophic factor (BDNF) was remarkably and time-dependently increased in dorsal root ganglion (DRG) neurons. DRG microinjection of BDNF siRNA in DRG ameliorated chronic constriction injury (CCI) induced mechanical, thermal, and cold nociceptive hypersensitivities. Overexpressing BDNF through microinjection of the AAV5-BDNF in DRG caused enhanced responses to basal mechanical, thermal, and cold stimuli in mice exposed to CCI. Mechanically, the P2X7 promoter activity was enhanced by CCI-induced increase of DRG BDNF protein and was involved in the CCI-induced upregulation of DRG P2X7 protein. The overexpression of BDNF also increased P2X7 expression in DRG neurons, which was validated in in vivo and in vitro experiments. BDNF may exert crucial effect via transcriptionally activating the P2X7 gene in primary sensory neurons, since P2X7 acts as a role of endogenous agitator in neuropathic pain and BDNF largely co-expresses with P2X7 in DRG neurons. Therefore, our data provide evidence that BDNF may be a promising therapeutic target for neuropathic pain.
Collapse
|
32
|
Jiang B, Zhong X, Fang J, Zhang A, WangD W, Liang Y, Fang J, Chen F, Du J. Electroacupuncture Attenuates Morphine Tolerance in Rats with Bone Cancer Pain by Inhibiting PI3K/Akt/JNK1/2 Signaling Pathway in the Spinal Dorsal Horn. Integr Cancer Ther 2021; 20:1534735421995237. [PMID: 33660537 PMCID: PMC8164555 DOI: 10.1177/1534735421995237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Purpose: Morphine is often used for the treatment of moderate and severe cancer pain,
but long-term use can lead to morphine tolerance. Methods for effectively
inhibiting morphine tolerance and the related mechanism of action are of
great significance for the treatment of cancer pain. Previous studies have
shown that electroacupuncture (EA) can inhibit the occurrence of morphine
tolerance, but the mechanism is not yet clear. The aim of the present study
was to explore the signaling pathway by which EA attenuates the development
of bone cancer pain (BCP)-morphine tolerance (MT). Materials and methods: Changes in the paw withdrawal threshold (PWT) of rats with bone cancer
pain-morphine tolerance were observed in a study of EA combined with
intrathecal injection of a PI3K inhibitor (LY294002) or agonist
(insulin-like growth factor-1 [IGF-1]). We also tested the protein
expression of phosphorylated phosphatidylinositol 3-kinase (p-PI3K),
phosphorylated protein kinase B (p-Akt), phosphorylated c-Jun
NH2-terminal kinase 1/2 (p-JNK1/2), and β-arrestin2 in the L4-6
spinal dorsal horn of rats. Results: The protein expression of p-PI3K, p-Akt, p-JNK1/2, and β-arrestin2 was
upregulated in the L4-6 spinal dorsal horn of rats with bone cancer pain and
bone cancer pain-morphine tolerance. EA delayed the occurrence of morphine
tolerance in rats with bone cancer pain and downregulated the protein
expression of p-PI3K, p-Akt, p-JNK1/2, and β-arrestin2 in the L4-6 spinal
dorsal horn of rats with bone cancer pain-morphine tolerance. Intrathecal
injection of LY294002 attenuated the development of morphine tolerance and
downregulated the protein expression of p-Akt, p-JNK1/2, and β-arrestin2 in
the spinal dorsal horn of rats with bone cancer pain-morphine tolerance. In
addition, the inhibitory effect of EA on morphine tolerance was reversed by
IGF-1. Conclusion: The mechanism underlying the ability of EA to attenuate morphine tolerance
may be associated with inhibition of the PI3K/Akt/JNK1/2 signaling
pathway.
Collapse
Affiliation(s)
- Bin Jiang
- Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, Zhejiang, China.,Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Xuemei Zhong
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China.,The Second Clinical College of Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Junfan Fang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Aijun Zhang
- Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, Zhejiang, China
| | - Wen WangD
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Yi Liang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Jianqiao Fang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Feng Chen
- Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, Zhejiang, China
| | - Junying Du
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| |
Collapse
|
33
|
Yousuf MS, Shiers SI, Sahn JJ, Price TJ. Pharmacological Manipulation of Translation as a Therapeutic Target for Chronic Pain. Pharmacol Rev 2021; 73:59-88. [PMID: 33203717 PMCID: PMC7736833 DOI: 10.1124/pharmrev.120.000030] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dysfunction in regulation of mRNA translation is an increasingly recognized characteristic of many diseases and disorders, including cancer, diabetes, autoimmunity, neurodegeneration, and chronic pain. Approximately 50 million adults in the United States experience chronic pain. This economic burden is greater than annual costs associated with heart disease, cancer, and diabetes combined. Treatment options for chronic pain are inadequately efficacious and riddled with adverse side effects. There is thus an urgent unmet need for novel approaches to treating chronic pain. Sensitization of neurons along the nociceptive pathway causes chronic pain states driving symptoms that include spontaneous pain and mechanical and thermal hypersensitivity. More than a decade of preclinical research demonstrates that translational mechanisms regulate the changes in gene expression that are required for ongoing sensitization of nociceptive sensory neurons. This review will describe how key translation regulation signaling pathways, including the integrated stress response, mammalian target of rapamycin, AMP-activated protein kinase (AMPK), and mitogen-activated protein kinase-interacting kinases, impact the translation of different subsets of mRNAs. We then place these mechanisms of translation regulation in the context of chronic pain states, evaluate currently available therapies, and examine the potential for developing novel drugs. Considering the large body of evidence now published in this area, we propose that pharmacologically manipulating specific aspects of the translational machinery may reverse key neuronal phenotypic changes causing different chronic pain conditions. Therapeutics targeting these pathways could eventually be first-line drugs used to treat chronic pain disorders. SIGNIFICANCE STATEMENT: Translational mechanisms regulating protein synthesis underlie phenotypic changes in the sensory nervous system that drive chronic pain states. This review highlights regulatory mechanisms that control translation initiation and how to exploit them in treating persistent pain conditions. We explore the role of mammalian/mechanistic target of rapamycin and mitogen-activated protein kinase-interacting kinase inhibitors and AMPK activators in alleviating pain hypersensitivity. Modulation of eukaryotic initiation factor 2α phosphorylation is also discussed as a potential therapy. Targeting specific translation regulation mechanisms may reverse changes in neuronal hyperexcitability associated with painful conditions.
Collapse
Affiliation(s)
- Muhammad Saad Yousuf
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas (M.S.Y., S.I.S., T.J.P.) and 4E Therapeutics Inc, Austin, Texas (J.J.S.)
| | - Stephanie I Shiers
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas (M.S.Y., S.I.S., T.J.P.) and 4E Therapeutics Inc, Austin, Texas (J.J.S.)
| | - James J Sahn
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas (M.S.Y., S.I.S., T.J.P.) and 4E Therapeutics Inc, Austin, Texas (J.J.S.)
| | - Theodore J Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas (M.S.Y., S.I.S., T.J.P.) and 4E Therapeutics Inc, Austin, Texas (J.J.S.)
| |
Collapse
|
34
|
Li J, Luan F, Song J, Dong J, Shang M. Clinical Efficacy of Controlled-Release Morphine Tablets Combined with Celecoxib in Pain Management and the Effects on WNK1 Expression. Clinics (Sao Paulo) 2021; 76:e1907. [PMID: 33503173 PMCID: PMC7798123 DOI: 10.6061/clinics/2021/e1907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/29/2020] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES This study was designed to evaluate the clinical efficacy of controlled-release morphine tablets combined with celecoxib in relieving osteocarcinoma-related pain and the effects of the combination on WNK1 expression. METHODS A total of 110 patients with osteocarcinoma-related pain were selected and divided into two groups based on the treatment administered, including the control group (treated with controlled-release morphine tablets alone) and the study group (treated with a combination of controlled-release morphine tablets and celecoxib). We compared the treatment efficacy, pain level (visual analog scale (VAS)), time of onset of breakthrough pain (BTP), dose of morphine, incidence of adverse events, quality of life (QOL) score, and With-no-lysine 1 (WNK1) expression in the peripheral blood (PB) as determined with qRT-PCR before and after treatment, of the two groups. RESULTS The total effective rate of the study group was higher than that of the control group, while the VAS score, time of onset of BTP, dose of morphine, incidence of adverse events, QOL score, and relative WNK1 expression in the PB were lower than those of the control group (p<0.05). CONCLUSION Combination treatment with controlled-release morphine tablets and celecoxib can be extensively used in the clinical setting because it effectively improves the symptoms, QOL score, and adverse effects in patients with osteocarcinoma-related pain.
Collapse
Affiliation(s)
- Jian Li
- Department of Joint Surgery, the Fourth People's Hospital of Jinan, Jinan, China
| | - Fanghai Luan
- Department of Orthopedic Surgery, the Fourth People's Hospital of Jinan, Jinan, China
| | - Jiangfeng Song
- Department of Orthopedic, Ju County People's Hospital, Rizhao, China
| | - Jianhua Dong
- Department of Orthopedic, Ju County People's Hospital, Rizhao, China
| | - Mingfu Shang
- Department of Spinal Cord Repairing, 960 Hospital of the Joint Logistics Support Force of PLA, Jinan, China
- *Corresponding author. E-mail:
| |
Collapse
|
35
|
Ling YJ, Ding TY, Dong FL, Gao YJ, Jiang BC. Intravenous Administration of Triptonide Attenuates CFA-Induced Pain Hypersensitivity by Inhibiting DRG AKT Signaling Pathway in Mice. J Pain Res 2020; 13:3195-3206. [PMID: 33293856 PMCID: PMC7718987 DOI: 10.2147/jpr.s275320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/19/2020] [Indexed: 01/11/2023] Open
Abstract
Background Currently, medical treatment of inflammatory pain is limited by a lack of safe and effective therapies. Triptonide (TPN), a major component of Tripterygium wilfordii Hook.f. with low toxicity, has been shown to have good anti-inflammatory and neuroprotective effects. The present study aims to investigate the effects of TPN on chronic inflammatory pain. Materials and Methods Inflammatory pain was induced by intraplantar injection of complete Freund’s adjuvant (CFA). TPN’s three different doses were intravenously administered to compare the analgesic efficacy: 0.1 mg/kg, 0.5 mg/kg, and 2.0 mg/kg. The foot swelling was quantitated by measuring paw volume. Mechanical allodynia and thermal hyperalgesia were assessed with von Frey filament testing and Hargreaves’ test, respectively. Western blots, qRT–PCR and immunofluorescence tests were used to analyze the expression of pAKT, tumor necrosis factor-α (TNF-α), interleukin 1 beta (IL-1β), and interleukin 6 (IL-6). Two AKT inhibitors, AKT inhibitor Ⅳ and MK-2206, were used to examine AKT’s effects on pain behavior and cytokines expression. Results Intravenous treatment with TPN attenuated CFA-induced paw edema, mechanical allodynia, and thermal hyperalgesia. Western blotting and immunofluorescence results showed that CFA induced AKT activation in the dorsal root ganglion (DRG) neurons. However, these effects were suppressed by treatment with TPN. Furthermore, TPN treatment inhibited CFA-induced increase of pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-6. Consistent with the in vivo data, TPN inhibited LPS-induced Akt phosphorylation and inflammatory mediator production in ND7/23 cells. Finally, intrathecal treatment with AKT inhibitor Ⅳ or MK-2206, attenuated CFA-induced mechanical allodynia and thermal hyperalgesia, and simultaneously decreased the mRNA expression of TNF-α, IL-1β, and IL-6 in DRG. Conclusion These data indicate that TPN attenuates CFA-induced pain potentially via inhibiting AKT-mediated pro-inflammatory cytokines production in DRG. TPN may be used for the treatment of chronic inflammatory pain.
Collapse
Affiliation(s)
- Yue-Juan Ling
- Institute of Pain Medicine, Nantong University, Nantong, Jiangsu 226019, People's Republic of China.,Institute of Special Environmental Medicine, Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Ting-Yu Ding
- Institute of Pain Medicine, Nantong University, Nantong, Jiangsu 226019, People's Republic of China.,Institute of Special Environmental Medicine, Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Fu-Lu Dong
- School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Yong-Jing Gao
- Institute of Pain Medicine, Nantong University, Nantong, Jiangsu 226019, People's Republic of China.,Institute of Special Environmental Medicine, Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Bao-Chun Jiang
- Institute of Pain Medicine, Nantong University, Nantong, Jiangsu 226019, People's Republic of China.,Institute of Special Environmental Medicine, Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| |
Collapse
|
36
|
Effect of heat shock protein 70 modulators on the development of morphine analgesic tolerance in rats. Behav Pharmacol 2020; 31:179-185. [PMID: 31770112 PMCID: PMC7077967 DOI: 10.1097/fbp.0000000000000528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The clinical use of opioid analgesics, such as morphine, is limited by analgesic tolerance, molecular mechanism of which is not well understood. Recently, molecular chaperone heat shock protein 70 (Hsp70) has been demonstrated to play important roles in morphine-induced neuroadaptation. Here, we focused on the involvement of Hsp70 in the development of analgesic tolerance to morphine. Rats were treated with morphine (5, 10, 20 mg/kg, subcutaneously) or saline once daily for 10 consecutive days. Hsp70 modulator N-formyl-3, 4-methylenedioxybenzylidine-γ-butyrolactam [KNK437, 100 mg/kg, intraperitoneally (i.p.)], geranylgeranylacetone (500 mg/kg, i.p.) or pifithrin-μ (20 mg/kg, i.p.) was administered before morphine (10 mg/kg, subcutaneously)/saline treatment. Analgesic effect of morphine was measured using the tail-flick latency test, and Hsp70 protein expression was examined by western blot. Analgesic effect of morphine decreased gradually with the increase in the number of days of morphine injection, indicating development of analgesic tolerance. A significant increase of Hsp70 expression in the periaqueductal gray was observed during the development of analgesic tolerance after repeated morphine injections. The development of morphine analgesic tolerance was suppressed by pre-treatment with Hsp70 transcriptional inhibitor KNK437 or functional antagonist pifithrin-μ, while promoted by pre-treatment with Hsp70 transcriptional inducer geranylgeranylacetone. Our results demonstrated that the development of morphine analgesic tolerance was dual regulated by Hsp70 modulators, suggesting Hsp70 as an interesting and new target for preventing the development of opioid analgesic tolerance.
Collapse
|
37
|
Ucha M, Roura-Martínez D, Ambrosio E, Higuera-Matas A. The role of the mTOR pathway in models of drug-induced reward and the behavioural constituents of addiction. J Psychopharmacol 2020; 34:1176-1199. [PMID: 32854585 DOI: 10.1177/0269881120944159] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Exposure to drugs of abuse induces neuroadaptations in critical nodes of the so-called reward systems that are thought to mediate the transition from controlled drug use to the compulsive drug-seeking that characterizes addictive disorders. These neural adaptations are likely to require protein synthesis, which is regulated, among others, by the mechanistic target of the rapamycin kinase (mTOR) signalling cascade. METHODS We have performed a narrative review of the literature available in PubMed about the involvement of the mTOR pathway in drug-reward and addiction-related phenomena. AIMS The aim of this study was to review the underlying architecture of this complex intracellular network and to discuss the alterations of its components that are evident after exposure to drugs of abuse. The aim was also to delineate the effects that manipulations of the mTOR network have on models of drug reward and on paradigms that recapitulate some of the psychological components of addiction. RESULTS There is evidence for the involvement of the mTOR pathway in the acute and rewarding effects of drugs of abuse, especially psychostimulants. However, the data regarding opiates are scarce. There is a need to use sophisticated animal models of addiction to ascertain the real role of the mTOR pathway in this pathology and not just in drug-mediated reward. The involvement of this pathway in behavioural addictions and impulsivity should also be studied in detail in the future. CONCLUSIONS Although there is a plethora of data about the modulation of mTOR by drugs of abuse, the involvement of this signalling pathway in addictive disorders requires further research.
Collapse
Affiliation(s)
- Marcos Ucha
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| | - David Roura-Martínez
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| | - Emilio Ambrosio
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| | - Alejandro Higuera-Matas
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| |
Collapse
|
38
|
Li Z, Peng X, Jia X, Su P, Liu D, Tu Y, Xu Q, Gao F. Spinal heat shock protein 27 participates in PDGFRβ-mediated morphine tolerance through PI3K/Akt and p38 MAPK signalling pathways. Br J Pharmacol 2020; 177:5046-5062. [PMID: 32559815 PMCID: PMC7589020 DOI: 10.1111/bph.15169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE The development of antinociceptive morphine tolerance is a clinically intractable problem. Earlier work has demonstrated the pivotal roles of PDGF and its receptor PDGFRβ in morphine tolerance. Here, we have investigated the role of spinal heat shock protein 27 (HSP27) in morphine tolerance and its relationship with PDGFRβ activation. EXPERIMENTAL APPROACH Rats were treated with morphine for 9 days, and its anti-nociceptive effect against thermal pain was evaluated by a tail-flick latency test. Western blot, real-time PCR, immunofluorescent staining, and various antagonists, agonists, and siRNA lentiviral vectors elucidated the roles of HSP27, PDGFRβ, and related signalling pathways in morphine tolerance. KEY RESULTS Chronic morphine administration increased expression and phosphorylation of HSP27 in the spinal cord. Down-regulating HSP27 attenuated the development of morphine tolerance. PDGFRβ antagonism inhibited HSP27 activation and attenuated and reversed morphine tolerance. PDGFRβ induction increased HSP27 expression and activation and partly decreased morphine analgesia. PDGFRβ inhibition reduced Akt and p38 MAPK activity in morphine tolerance. PI3K and p38 inhibitors reversed morphine tolerance and suppressed morphine-induced HSP27 phosphorylation. CONCLUSION AND IMPLICATIONS This study demonstrated for the first time that spinal HSP27 participates in PDGFRβ-mediated morphine tolerance via the PI3K/Akt and p38 MAPK signalling pathways. These findings suggest a potential clinical strategy for prolonging the antinociceptive effects of opioids during long-term pain control.
Collapse
Affiliation(s)
- Zheng Li
- Department of Anesthesiology, Tongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoling Peng
- Department of Anesthesiology, Tongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoqian Jia
- Department of Anesthesiology, Tongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Peng Su
- Department of Anesthesiology, Sichuan Academy of Medical SciencesSichuan Provincial People's HospitalChengduChina
| | - Daiqiang Liu
- Department of Anesthesiology, Tongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Ye Tu
- Department of Anesthesiology, Tongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Qiaoqiao Xu
- Department of Anesthesiology, Tongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Feng Gao
- Department of Anesthesiology, Tongji HospitalHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
39
|
Huang T, Fu G, Gao J, Zhang Y, Cai W, Wu S, Jia S, Xia S, Bachmann T, Bekker A, Tao YX. Fgr contributes to hemorrhage-induced thalamic pain by activating NF-κB/ERK1/2 pathways. JCI Insight 2020; 5:139987. [PMID: 33055425 PMCID: PMC7605540 DOI: 10.1172/jci.insight.139987] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/09/2020] [Indexed: 12/22/2022] Open
Abstract
Thalamic pain, a type of central poststroke pain, frequently occurs following ischemia/hemorrhage in the thalamus. Current treatment of this disorder is often ineffective, at least in part due to largely unknown mechanisms that underlie thalamic pain genesis. Here, we report that hemorrhage caused by microinjection of type IV collagenase or autologous whole blood into unilateral ventral posterior lateral nucleus and ventral posterior medial nucleus of the thalamus increased the expression of Fgr, a member of the Src family nonreceptor tyrosine kinases, at both mRNA and protein levels in thalamic microglia. Pharmacological inhibition or genetic knockdown of thalamic Fgr attenuated the hemorrhage-induced thalamic injury on the ipsilateral side and the development and maintenance of mechanical, heat, and cold pain hypersensitivities on the contralateral side. Mechanistically, the increased Fgr participated in hemorrhage-induced microglial activation and subsequent production of TNF-α likely through activation of both NF-κB and ERK1/2 pathways in thalamic microglia. Our findings suggest that Fgr is a key player in thalamic pain and a potential target for the therapeutic management of this disorder.
Collapse
Affiliation(s)
| | | | - Ju Gao
- Department of Anesthesiology
| | | | | | | | | | | | | | | | - Yuan-Xiang Tao
- Department of Anesthesiology
- Department of Pharmacology, Physiology & Neuroscience; and
- Department of Cell Biology & Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| |
Collapse
|
40
|
Dozio V, Daali Y, Desmeules J, Sanchez JC. Deep proteomics and phosphoproteomics reveal novel biological pathways perturbed by morphine, morphine-3-glucuronide and morphine-6-glucuronide in human astrocytes. J Neurosci Res 2020; 100:220-236. [PMID: 32954564 DOI: 10.1002/jnr.24731] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/06/2020] [Accepted: 08/31/2020] [Indexed: 01/08/2023]
Abstract
Tolerance and hyperalgesia associated with chronic exposure to morphine are major limitations in the clinical management of chronic pain. At a cellular level, neuronal signaling can in part account for these undesired side effects, but unknown mechanisms mediated by central nervous system glial cells are likely also involved. Here we applied data-independent acquisition mass spectrometry to perform a deep proteome and phosphoproteome analysis of how human astrocytes responds to opioid stimulation. We unveil time- and dose-dependent effects induced by morphine and its major active metabolites morphine-3-glucuronide (M3G) and morphine-6-glucuronide that converging on activation of mitogen-activated protein kinase and mammalian target of rapamycin signaling pathways. We also find that especially longer exposure to M3G leads to significant dysregulation of biological pathways linked to extracellular matrix organization, antigen presentation, cell adhesion, and glutamate homeostasis, which are crucial for neuron- and leukocyte-astrocyte interactions.
Collapse
Affiliation(s)
- Vito Dozio
- Department of Medicine, University of Geneva, Geneva, Switzerland.,Swiss Centre for Applied Human Toxicology, Basel, Switzerland
| | - Youssef Daali
- Swiss Centre for Applied Human Toxicology, Basel, Switzerland.,Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Emergency Medicine and Intensive Cares, Geneva University Hospitals, Geneva, Switzerland
| | - Jules Desmeules
- Swiss Centre for Applied Human Toxicology, Basel, Switzerland.,Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Emergency Medicine and Intensive Cares, Geneva University Hospitals, Geneva, Switzerland
| | - Jean-Charles Sanchez
- Department of Medicine, University of Geneva, Geneva, Switzerland.,Swiss Centre for Applied Human Toxicology, Basel, Switzerland
| |
Collapse
|
41
|
Persistent Rheb-induced mTORC1 activation in spinal cord neurons induces hypersensitivity in neuropathic pain. Cell Death Dis 2020; 11:747. [PMID: 32920594 PMCID: PMC7487067 DOI: 10.1038/s41419-020-02966-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/21/2020] [Accepted: 08/31/2020] [Indexed: 12/21/2022]
Abstract
The small GTPase Ras homolog enriched in the brain (Rheb) can activate mammalian target of rapamycin (mTOR) and regulate the growth and cell cycle progression. We investigated the role of Rheb-mediated mTORC1 signaling in neuropathic pain. A chronic constriction injury (CCI) model was dopted. CCI induced obvious spinal Rheb expression and phosphorylation of mTOR, S6, and 4-E-BP1. Blocking mTORC1 signal with rapamycin alleviated the neuropathic pain and restored morphine efficacy in CCI model. Immunofluoresence showed a neuronal co-localization of CCI-induced Rheb and pS6. Rheb knockin mouse showed a similar behavioral phenotype as CCI. In spinal slice recording, CCI increased the firing frequency of neurons expressing HCN channels; inhibition of mTORC1 with rapamycin could reverse the increased spinal neuronal activity in neuropathic pain. Spinal Rheb is induced in neuropathic pain, which in turn active the mTORC1 signaling in CCI. Spinal Rheb-mTOR signal plays an important role in regulation of spinal sensitization in neuropathic pain, and targeting mTOR may give a new strategy for pain management.
Collapse
|
42
|
Gao Y, Bai L, Zhou W, Yang Y, Zhang J, Li L, Jiang M, Mi Y, Li TT, Zhang X, Zhang W, Xu JT. PARP-1-regulated TNF-α expression in the dorsal root ganglia and spinal dorsal horn contributes to the pathogenesis of neuropathic pain in rats. Brain Behav Immun 2020; 88:482-496. [PMID: 32283287 DOI: 10.1016/j.bbi.2020.04.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 01/04/2023] Open
Abstract
Emerging evidence has implicated poly-(ADP-ribose) polymerase 1 (PARP-1), a transcriptional coregulator, in a variety of inflammatory diseases. In the current study, the role of PARP-1 in neuropathic pain and the underlying mechanisms were investigated. Neuropathic pain was determined by assessing the paw withdrawal threshold (PWT) and paw withdrawal latency (PWL) following lumbar 5 spinal nerve ligation (SNL) in male rates. Western blotting, qRT-PCR, immunohistochemistry, chromatin immunoprecipitation (ChIP), and Co-IP assays were performed to elucidate the mechanisms. The results showed that SNL resulted in a significant increase in the expression and activation of PARP-1 in the ipsilateral L4/5 dorsal root ganglia (DRG) and spinal dorsal horn, which occurred on day one, reached peak on day 7, and persisted more than 2 weeks after surgery. Double immunofluorescence staining revealed that PARP-1 was expressed exclusively in DRG A-type and C-type neurons. In the spinal cord, PARP-1 mainly colocalized with the neuronal marker NeuN and the astrocytic marker GFAP specifically in the superficial lamina. Prior intrathecal (i.t.) injection of PJ-34, a PARPs inhibitor, or Tiq-A, a specific PARP-1 inhibitor, dose-dependently prevented the reductions in PWT and PWL following SNL. Established neuropathic pain-like hypersensitivity was also attenuated with i.t. injection of PJ-34 and Tiq-A starting on day 7 following SNL, a timepoint at which neuropathic pain was fully established. SNL-induced mechanical allodynia and thermal hyperalgesia were also alleviated by i.t. injection of PARP-1 siRNA following a reduction in PARP-1 expression in the dorsal horn. Moreover, the SNL-induced increases in TNF-α protein and mRNA in the dorsal horn and DRG were dramatically suppressed by i.t. injection of Tiq-A or PARP-1 siRNA. The i.t. lipopolysaccharide (LPS)-induced increase in the production of TNF-α in the dorsal horn was also inhibited by prior to i.t. injection of PARP-1 siRNA. Results of ChIP assay showed that SNL-induced PARP-1 activation promoted the binding of NF-κB p65 with the TNF-α promoter in the dorsal horn and that PARP-1 inhibition reduced this binding and suppressed TNF-α expression. Co-IP assay revealed that SNL caused a significant increase in the level of histone H1 poly(ADP)-ribosylation. Together, these results indicate that PARP-1-regulated TNF-α expression in the DRG and spinal dorsal horn following SNL contributes to the development and maintenance of neuropathic pain. Targeting PARP-1 might be a promising therapeutic strategy for the treatment of the chronic pain.
Collapse
Affiliation(s)
- Yan Gao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Liying Bai
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Wenjuan Zhou
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Yin Yang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Jian Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Liren Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Mingjun Jiang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Yang Mi
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Tong-Tong Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Xuan Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Wei Zhang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Ji-Tian Xu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China; Neuroscience Research Institute, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China.
| |
Collapse
|
43
|
Stine C, Coleman DL, Flohrschutz AT, Thompson AL, Mishra S, Blagg BS, Largent-Milnes TM, Lei W, Streicher JM. Heat shock protein 90 inhibitors block the antinociceptive effects of opioids in mouse chemotherapy-induced neuropathy and cancer bone pain models. Pain 2020; 161:1798-1807. [PMID: 32701840 PMCID: PMC8607824 DOI: 10.1097/j.pain.0000000000001886] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Heat shock protein 90 (Hsp90) is a ubiquitous signal transduction regulator, and Hsp90 inhibitors are in clinical development as cancer therapeutics. However, there have been very few studies on the impact of Hsp90 inhibitors on pain or analgesia, a serious concern for cancer patients. We previously found that Hsp90 inhibitors injected into the brain block opioid-induced antinociception in tail flick, paw incision, and HIV neuropathy pain. This study extended from that initial work to test the cancer-related clinical impact of Hsp90 inhibitors on opioid antinociception in cancer-induced bone pain in female BALB/c mice and chemotherapy-induced peripheral neuropathy in male and female CD-1 mice. Mice were treated with Hsp90 inhibitors (17-AAG, KU-32) by the intracerebroventricular, intrathecal, or intraperitoneal routes, and after 24 hours, pain behaviors were evaluated after analgesic drug treatment. Heat shock protein 90 inhibition in the brain or systemically completely blocked morphine and oxymorphone antinociception in chemotherapy-induced peripheral neuropathy; this effect was partly mediated by decreased ERK and JNK MAPK activation and by increased protein translation, was not altered by chronic treatment, and Hsp90 inhibition had no effect on gabapentin antinociception. We also found that the Hsp90 isoform Hsp90α and the cochaperone Cdc37 were responsible for the observed changes in opioid antinociception. By contrast, Hsp90 inhibition in the spinal cord or systemically partially reduced opioid antinociception in cancer-induced bone pain. These results demonstrate that Hsp90 inhibitors block opioid antinociception in cancer-related pain, suggesting that Hsp90 inhibitors for cancer therapy could decrease opioid treatment efficacy.
Collapse
Affiliation(s)
- Carrie Stine
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724 USA
| | - Deziree L. Coleman
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724 USA
| | - Austin T. Flohrschutz
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724 USA
| | - Austen L. Thompson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724 USA
| | - Sanket Mishra
- Department of Chemistry and Biochemistry, College of Science, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Brian S. Blagg
- Department of Chemistry and Biochemistry, College of Science, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Tally M. Largent-Milnes
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724 USA
| | - Wei Lei
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724 USA
- Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Presbyterian College, Clinton, SC 29325 USA
| | - John M. Streicher
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724 USA
| |
Collapse
|
44
|
Injection of minocycline into the periaqueductal gray attenuates morphine withdrawal signs. Neurosci Lett 2020; 736:135283. [PMID: 32739271 DOI: 10.1016/j.neulet.2020.135283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/07/2020] [Accepted: 07/28/2020] [Indexed: 11/22/2022]
Abstract
This study investigated the effects of minocycline microinjections, into the midbrain periaqueductal gray (PAG), on morphine withdrawal and the expression of pannexin-1 (panx1), phosphorylated mammalian target of rapamycin (p-mTOR), protein kinase A (PKA), and cAMP response element-binding protein (CREB). Rats were injected with morphine, intraperitoneally, at increasing doses, twice per day, to establish animal models of morphine exposure. Minocycline was administered into the PAG before the first intraperitoneal (i.p.) injection of morphine each day, on days 1-4. On the last day of the experiment, all rats were injected with naloxone, and morphine withdrawal was observed, and then changes in the expression levels of ionized calcium-binding adaptor molecule 1 (Iba1) and its downstream factors, panx1, p-mTOR, PKA, and CREB were evaluated by western blot and immunohistochemistry analyses. Morphine withdrawal increased microglial activation, whereas minocycline could inhibit microglial activation and withdrawal and the downregulation of panx1, p-mTOR, PKA, and CREB expression, reducing the effects of morphine withdrawal.
Collapse
|
45
|
Xu B, Liu SS, Wei J, Jiao ZY, Mo C, Lv CM, Huang AL, Chen QB, Ma L, Guan XH. Role of Spinal Cord Akt-mTOR Signaling Pathways in Postoperative Hyperalgesia Induced by Plantar Incision in Mice. Front Neurosci 2020; 14:766. [PMID: 32848550 PMCID: PMC7396510 DOI: 10.3389/fnins.2020.00766] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/29/2020] [Indexed: 12/29/2022] Open
Abstract
Poor postoperative pain (POP) control increases perioperative morbidity, prolongs hospitalization days, and causes chronic pain. However, the specific mechanism(s) underlying POP is unclear and the identification of optimal perioperative treatment remains elusive. Akt and mammalian target of rapamycin (mTOR) are expressed in the spinal cord, dorsal root ganglion, and sensory axons. In this study, we explored the role of Akt and mTOR in pain-related behaviors induced by plantar incision in mice. Plantar incision activated spinal Akt and mTOR in a dose-dependent manner. Pre-treatment with Akt inhibitors intrathecally prevented the activation of mTOR dose-dependently. In addition, blocking the Akt-mTOR signaling cascade attenuated pain-related behaviors and spinal Fos protein expression induced by plantar incision. Our observations demonstrate that Akt-mTOR might be a potential therapeutic target for the treatment of POP.
Collapse
Affiliation(s)
- Bing Xu
- Department of Rehabilitation, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Su-Su Liu
- Department of Anesthesiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jin Wei
- Department of Anesthesiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zi-Yin Jiao
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Cheng Mo
- Department of Anesthesiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Cheng-Mei Lv
- Department of Anesthesiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ai-Lan Huang
- Department of Anesthesiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Qi-Bo Chen
- Department of Rehabilitation, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Li Ma
- Department of Anesthesiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xue-Hai Guan
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
46
|
MicroRNA-330 Directs Downregulation of the GABA BR2 in the Pathogenesis of Pancreatic Cancer Pain. J Mol Neurosci 2020; 70:1541-1551. [PMID: 32621101 DOI: 10.1007/s12031-020-01607-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/20/2020] [Indexed: 12/22/2022]
Abstract
Pancreatic cancer is one of the most aggressive and deadly malignancies with a very poor prognosis. Pancreatic cancer-induced visceral pain is very common and is generally presented among the initial symptoms in patients; such pain is strongly associated with poor quality of life, impaired functional activity, and decreased survival. However, the principal neurobiological mechanisms of pain caused by pancreatic cancer have not been fully elucidated. Accumulating studies have shown that miRNAs play a major role in chronic pain by suppressing key molecules involved in nociception. In the present study, we report that microRNA (miR)-330 is highly expressed in the spinal dorsal horn (SDH) of nude mice with pancreatic cancer pain. Mimicking pancreatic carcinoma-induced SDH miR-330 upregulation by microinjection of miR-330 mimic into the SDH significantly induced abdominal mechanical allodynia in normal nude mice. Additionally, we found that the expression of GABABR2 was significantly decreased in the SDH of nude mice with pancreatic cancer pain and was regulated directly by miR-330 both in vitro and in vivo. Furthermore, inhibition of miR-330 rescued the expression of GABABR2 and alleviated pancreatic carcinoma-induced abdominal pain hypersensitivity in nude mice with pancreatic carcinoma. These results show that miR-330 participates in the genesis of pancreatic carcinoma-induced pain hypersensitivity by inhibiting GABABR2 expression in the SDH and might be a potential therapeutic target for pancreatic cancer pain.
Collapse
|
47
|
Li Y, Guo X, Sun L, Xiao J, Su S, Du S, Li Z, Wu S, Liu W, Mo K, Xia S, Chang Y, Denis D, Tao Y. N 6-Methyladenosine Demethylase FTO Contributes to Neuropathic Pain by Stabilizing G9a Expression in Primary Sensory Neurons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902402. [PMID: 32670741 PMCID: PMC7341103 DOI: 10.1002/advs.201902402] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 02/27/2020] [Indexed: 05/23/2023]
Abstract
Nerve injury-induced change in gene expression in primary sensory neurons of dorsal root ganglion (DRG) is critical for neuropathic pain genesis. N6-methyladenosine (m6A) modification of RNA represents an additional layer of gene regulation. Here, it is reported that peripheral nerve injury increases the expression of the m6A demethylase fat-mass and obesity-associated proteins (FTO) in the injured DRG via the activation of Runx1, a transcription factor that binds to the Fto gene promoter. Mimicking this increase erases m6A in euchromatic histone lysine methyltransferase 2 (Ehmt2) mRNA (encoding the histone methyltransferase G9a) and elevates the level of G9a in DRG and leads to neuropathic pain symptoms. Conversely, blocking this increase reverses a loss of m6A sites in Ehmt2 mRNA and destabilizes the nerve injury-induced G9a upregulation in the injured DRG and alleviates nerve injury-associated pain hypersensitivities. FTO contributes to neuropathic pain likely through stabilizing nerve injury-induced upregulation of G9a, a neuropathic pain initiator, in primary sensory neurons.
Collapse
Affiliation(s)
- Yize Li
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Xinying Guo
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Linlin Sun
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Jifang Xiao
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Songxue Su
- Department of Physiology, Pharmacology & NeuroscienceNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E661NewarkNJ07103USA
| | - Shibin Du
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Zhen Li
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Shaogen Wu
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Weili Liu
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Kai Mo
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Shangzhou Xia
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Yun‐Juan Chang
- The Office of Advanced Research ComputingRutgers, The State University of New Jersey 185 S. Orange Ave., MSB C‐630NewarkNJ07103USA
| | - Daniel Denis
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Yuan‐Xiang Tao
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
- Department of Physiology, Pharmacology & NeuroscienceNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E661NewarkNJ07103USA
- Department of Cell Biology & Molecular MedicineNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E661NewarkNJ07103USA
| |
Collapse
|
48
|
Chu WG, Wang FD, Sun ZC, Ma SB, Wang X, Han WJ, Wang F, Bai ZT, Wu SX, Freichel M, Xie RG, Luo C. TRPC1/4/5 channels contribute to morphine-induced analgesic tolerance and hyperalgesia by enhancing spinal synaptic potentiation and structural plasticity. FASEB J 2020; 34:8526-8543. [PMID: 32359120 DOI: 10.1096/fj.202000154rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 02/10/2024]
Abstract
Opioid analgesics remain the mainstay for managing intractable chronic pain, but their use is limited by detrimental side effects such as analgesic tolerance and hyperalgesia. Calcium-dependent synaptic plasticity is a key determinant in opiates tolerance and hyperalgesia. However, the exact substrates for this calcium-dependent synaptic plasticity in mediating these maladaptive processes are largely unknown. Canonical transient receptor potential 1, 4, and 5 (TRPC1, 4, 5) proteins assemble into heteromultimeric nonselective cation channels with high Ca2+ permeability and influence various neuronal functions. However, whether and how TRPC1/4/5 channels contribute to the development of opiates tolerance and hyperalgesia remains elusive. Here, we show that TRPC1/4/5 channels contribute to the generation of morphine tolerance and hyperalgesia. Chronic morphine exposure leads to upregulation of TRPC1/4/5 channels in the spinal cord. Spinally expressed TRPC1, TPRC4, and TRPC5 are required for chronic morphine-induced synaptic long-term potentiation (LTP) as well as remodeling of synaptic spines in the dorsal horn, thereby orchestrating functional and structural plasticity during the course of morphine-induced hyperalgesia and tolerance. These effects are attributed to TRPC1/4/5-mediated Ca2+ elevation in the spinal dorsal horn induced by chronic morphine treatment. This study identifies TRPC1/4/5 channels as a promising novel target to prevent the unwanted morphine tolerance and hyperalgesia.
Collapse
Affiliation(s)
- Wen-Guang Chu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Fu-Dong Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- The Fourth Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Zhi-Chuan Sun
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Sui-Bin Ma
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Xu Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- Research Center for Resource Polypeptide Drugs & College of Life Sciences, Yanan University, Yanan, China
| | - Wen-Juan Han
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Fei Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Zhan-Tao Bai
- Research Center for Resource Polypeptide Drugs & College of Life Sciences, Yanan University, Yanan, China
| | - Sheng-Xi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Rou-Gang Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Ceng Luo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
49
|
Niu Q, Xing F, Gu HW, Bai L, Zhang J, Yuan JJ, Mao YY, Li ZS, Zhang W, Xu JT. Upregulation of Myeloid Zinc Finger 1 in Dorsal Root Ganglion via Regulating Matrix Metalloproteinase-2/9 and Voltage-gated Potassium 1.2 Expression Contributes to Complete Freund’s Adjuvant-induced Inflammatory Pain. Neuroscience 2020; 432:174-187. [DOI: 10.1016/j.neuroscience.2020.02.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 11/30/2022]
|
50
|
Qian J, Zhu Y, Bai L, Gao Y, Jiang M, Xing F, Zhang J, Zhao W, Gu H, Mi Y, Tao YX, Xu JT. Chronic morphine-mediated upregulation of high mobility group box 1 in the spinal cord contributes to analgesic tolerance and hyperalgesia in rats. Neurotherapeutics 2020; 17:722-742. [PMID: 31879851 PMCID: PMC7283437 DOI: 10.1007/s13311-019-00800-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Analgesic tolerance and hyperalgesia hinder the long-term utility of opioids. We examined whether spinal high mobility group box 1 (HMGB1) is involved in morphine tolerance and its underlying mechanisms by using a model of repeated intrathecal (i.t.) injections of morphine. The results showed that chronic i.t. morphine exposure led to increased expression of HMGB1, Toll-like receptor 4 (TLR4), and receptor for advanced glycation end products (RAGE) and their mRNAs in the dorsal horn. Morphine challenge also promoted HMGB1 expression and release in cultured spinal neurons, but these effects were inhibited by TAK-242, naloxone (antagonists of TLR4), and TLR4 siRNA. Intrathecal coadministration of morphine with TAK-242 or PDTC (inhibitor of NF-κB activation) also reduced HMGB1 expression in the spinal cord. Repeated i.t. coinjections of morphine with glycyrrhizin (GL, an HMGB1 inhibitor) or HMGB1 siRNA prevented reduction of the maximal possible analgesic effect (MPAE) of morphine and alleviated morphine withdrawal-induced hyperalgesia. The established morphine tolerance and hyperalgesia were partially reversed when i.t. injections of GL or HMGB1 antibody started at day 7 of morphine injection. Repeated i.t. injections of morphine with HMGB1 siRNA inhibited the activation of NF-κB, but not that of JNK and p38. A single i.t. injection of HMGB1 in naïve rats caused pain-related hypersensitivity and reduction in MPAE. Moreover, phosphorylated NF-κB p65, TNF-α, and IL-1β levels in the dorsal horn were upregulated following this treatment, but this upregulation was prevented by coinjection with TAK-242. Together, these results suggest that morphine-mediated upregulation of spinal HMGB1 contributes to analgesic tolerance and hyperalgesia via activation of TLR4/NF-κB signaling, and the HMGB1 inhibitor might be a promising adjuvant to morphine in the treatment of intractable pain in the clinic.
Collapse
Affiliation(s)
- Junliang Qian
- Department of Physiology and Neurobiology, Zhengzhou University School of Basic Medical Sciences, 100 Science Avenue, Zhengzhou, 450001, China
| | - Yanan Zhu
- Department of Physiology and Neurobiology, Zhengzhou University School of Basic Medical Sciences, 100 Science Avenue, Zhengzhou, 450001, China
| | - Liying Bai
- Department of Anesthesiology, The First Affiliated Hospital, Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
| | - Yan Gao
- Department of Physiology and Neurobiology, Zhengzhou University School of Basic Medical Sciences, 100 Science Avenue, Zhengzhou, 450001, China
- Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, 100 Science Avenue, Zhengzhou, 450001, China
| | - Mingjun Jiang
- Department of Physiology and Neurobiology, Zhengzhou University School of Basic Medical Sciences, 100 Science Avenue, Zhengzhou, 450001, China
| | - Fei Xing
- Department of Anesthesiology, The First Affiliated Hospital, Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
| | - Jian Zhang
- Department of Physiology and Neurobiology, Zhengzhou University School of Basic Medical Sciences, 100 Science Avenue, Zhengzhou, 450001, China
| | - Wenchao Zhao
- Department of Physiology and Neurobiology, Zhengzhou University School of Basic Medical Sciences, 100 Science Avenue, Zhengzhou, 450001, China
| | - Hanwen Gu
- Department of Anesthesiology, The First Affiliated Hospital, Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
| | - Yang Mi
- Department of Physiology and Neurobiology, Zhengzhou University School of Basic Medical Sciences, 100 Science Avenue, Zhengzhou, 450001, China
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, E-661, Newark, NJ, 07103, USA
| | - Ji-Tian Xu
- Department of Physiology and Neurobiology, Zhengzhou University School of Basic Medical Sciences, 100 Science Avenue, Zhengzhou, 450001, China.
- Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, 100 Science Avenue, Zhengzhou, 450001, China.
| |
Collapse
|