1
|
AboTaleb HA, Alghamdi BS. Metformin and fibromyalgia pathophysiology: current insights and promising future therapeutic strategies. Mol Biol Rep 2024; 52:60. [PMID: 39692938 DOI: 10.1007/s11033-024-10159-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024]
Abstract
Fibromyalgia (FM) is a complex, chronic pain syndrome characterized by widespread musculoskeletal pain, fatigue, and cognitive disturbances. Despite its prevalence, the pathophysiology of FM remains poorly understood, with current treatments often providing limited relief. Recent studies have suggested that metformin, a widely used antidiabetic drug, may have potential therapeutic benefits for chronic pain conditions, including FM. This review aims to provide current insights into the role of metformin in FM pathophysiology, focusing on its neurotransmitter-modulating and anti-inflammatory effects. Metformin has been shown to mitigate neuroinflammation, protect neural tissues, and modulate key neurotransmitters involved in pain and mood regulation. These effects are particularly evident in animal models, where metformin has been observed to reduce pain sensitivity, improve mood-related behaviors, and decrease levels of pro-inflammatory cytokines like interleukin 1-beta (IL-1β). Additionally, the ability of metformin to influence serotonin, norepinephrine, and glutamate levels suggests a potential mechanism for its analgesic and mood-stabilizing effects. However, the current evidence is largely preclinical, and further research is needed to confirm these findings in human studies. This review aims to encourage researchers to explore the association between metformin and FM more deeply, with the hope of uncovering new therapeutic strategies that could offer relief to FM patients.
Collapse
Affiliation(s)
- Hanin Abdulbaset AboTaleb
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, 21589, Jeddah, Saudi Arabia.
- Neuroscience and Geroscience Research Unit, King Fahd Medical Research Center, King Abdulaziz University, 21589, Jeddah, Saudi Arabia.
| | - Badrah S Alghamdi
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
- Neuroscience and Geroscience Research Unit, King Fahd Medical Research Center, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| |
Collapse
|
2
|
AboTaleb HA, Alturkistani HA, Abd El-Aziz GS, Hindi EA, Halawani MM, Al-Thepyani MA, Alghamdi BS. The Antinociceptive Effects and Sex-Specific Neurotransmitter Modulation of Metformin in a Mouse Model of Fibromyalgia. Cells 2024; 13:1986. [PMID: 39682734 DOI: 10.3390/cells13231986] [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: 10/22/2024] [Revised: 11/06/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Fibromyalgia (FM) is a chronic and debilitating condition characterized by diffuse pain, often associated with symptoms such as fatigue, cognitive disturbances, and mood disorders. Metformin, an oral hypoglycemic agent, has recently gained attention for its potential benefits beyond glucose regulation. It has shown promise in alleviating neuropathic and inflammatory pain, suggesting that it could offer a novel approach to managing chronic pain conditions like FM. This study aimed to further explore metformin's analgesic potential by evaluating its effects in an experimental FM model induced by reserpine in both male and female mice. After the administration of 200 mg/kg metformin to male and female mice, the FM-related symptoms were assessed, including mechanical allodynia, thermal hyperalgesia, and depressive-like behaviors. A histological examination of the thalamus, hippocampus, and spinal cord was conducted using haematoxylin and eosin staining. The neurotransmitter and proinflammatory cytokines levels were measured in the brains and spinal cords. Our results have shown that metformin treatment for seven days significantly reversed these FM-like symptoms, reducing pain sensitivity and improving mood-related behaviors in both the male and female mice. Additionally, metformin exhibited neuroprotective effects, mitigating reserpine-induced damage in the hippocampus, thalamus, and spinal cord. It also significantly lowered the levels of the proinflammatory cytokine interleukin 1-beta (IL-1β) in the brain and spinal cord. Notably, metformin modulated the neurotransmitter levels differently between the sexes, decreasing glutamate and increasing serotonin and norepinephrine in the male mice, but not in the females. These findings underscore metformin's potential as an alternative therapy for FM, with sex-specific differences suggesting distinct mechanisms of action.
Collapse
Affiliation(s)
- Hanin Abdulbaset AboTaleb
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Neuroscience and Geroscience Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hani A Alturkistani
- Department of Clinical Anatomy, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Gamal S Abd El-Aziz
- Department of Clinical Anatomy, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Emad A Hindi
- Neuroscience and Geroscience Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Clinical Anatomy, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Mervat M Halawani
- Neuroscience and Geroscience Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Clinical Anatomy, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Mona Ali Al-Thepyani
- Neuroscience and Geroscience Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Chemistry, College of Sciences & Arts, King Abdulaziz University, Rabigh 21911, Saudi Arabia
| | - Badrah S Alghamdi
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Neuroscience and Geroscience Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
3
|
Zhang X, Jin T, Wang H, Han S, Liang Y. Microglia in morphine tolerance: cellular and molecular mechanisms and therapeutic potential. Front Pharmacol 2024; 15:1499799. [PMID: 39669194 PMCID: PMC11635611 DOI: 10.3389/fphar.2024.1499799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024] Open
Abstract
Morphine has a crucial role in treating both moderate to severe pain and chronic pain. However, prolonged administration of morphine can lead to tolerance of analgesia, resulting in increased doses and poor treatment of pain. Many patients, such as those with terminal cancer, require high doses of morphine for long periods. Addressing morphine tolerance can help this group of patients to escape pain, and the mechanisms behind this need to be investigated. Microglia are the key cells involved in morphine tolerance and chronic morphine administration leads to microglia activation, which in turn leads to activation of internal microglia signalling pathways and protein transcription, ultimately leading to the release of inflammatory factors. Inhibiting the activation of microglia internal signalling pathways can reduce morphine tolerance. However, the exact mechanism of how morphine acts on microglia and ultimately leads to tolerance is unknown. This article discusses the mechanisms of morphine induced microglia activation, reviews the signalling pathways within microglia and the associated therapeutic targets and possible drugs, and provides possible directions for clinical prevention or retardation of morphine induced analgesic tolerance.
Collapse
Affiliation(s)
- Xiangning Zhang
- Department of Anesthesiology, Women and Children’s Hospital, Peking University People’s Hospital, Qingdao University, Qingdao, Shandong, China
- Clinical Medical College, Qingdao University, Qingdao, Shandong, China
| | - Tingting Jin
- Department of Anesthesiology, Women and Children’s Hospital, Peking University People’s Hospital, Qingdao University, Qingdao, Shandong, China
- Clinical Medical College, Qingdao University, Qingdao, Shandong, China
| | - Haixia Wang
- Department of Anesthesiology, Women and Children’s Hospital, Peking University People’s Hospital, Qingdao University, Qingdao, Shandong, China
- Clinical Medical College, Qingdao University, Qingdao, Shandong, China
| | - Shuai Han
- Department of Anesthesiology, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yongxin Liang
- Department of Anesthesiology, Women and Children’s Hospital, Peking University People’s Hospital, Qingdao University, Qingdao, Shandong, China
- Clinical Medical College, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
4
|
Kummer K, Sheets PL. Targeting Prefrontal Cortex Dysfunction in Pain. J Pharmacol Exp Ther 2024; 389:268-276. [PMID: 38702195 PMCID: PMC11125798 DOI: 10.1124/jpet.123.002046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/12/2024] [Accepted: 04/02/2024] [Indexed: 05/06/2024] Open
Abstract
The prefrontal cortex (PFC) has justifiably become a significant focus of chronic pain research. Collectively, decades of rodent and human research have provided strong rationale for studying the dysfunction of the PFC as a contributing factor in the development and persistence of chronic pain and as a key supraspinal mechanism for pain-induced comorbidities such as anxiety, depression, and cognitive decline. Chronic pain alters the structure, chemistry, and connectivity of PFC in both humans and rodents. In this review, we broadly summarize the complexities of reported changes within both rodent and human PFC caused by pain and offer insight into potential pharmacological and nonpharmacological approaches for targeting PFC to treat chronic pain and pain-associated comorbidities. SIGNIFICANCE STATEMENT: Chronic pain is a significant unresolved medical problem causing detrimental changes to physiological, psychological, and behavioral aspects of life. Drawbacks of currently approved pain therapeutics include incomplete efficacy and potential for abuse producing a critical need for novel approaches to treat pain and comorbid disorders. This review provides insight into how manipulation of prefrontal cortex circuits could address this unmet need of more efficacious and safer pain therapeutics.
Collapse
Affiliation(s)
- Kai Kummer
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria (K.K.); Department of Pharmacology and Toxicology (P.L.S.), Medical Neurosciences Graduate Program (P.L.S.), and Stark Neurosciences Research Institute (P.L.S.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Patrick L Sheets
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria (K.K.); Department of Pharmacology and Toxicology (P.L.S.), Medical Neurosciences Graduate Program (P.L.S.), and Stark Neurosciences Research Institute (P.L.S.), Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
5
|
Yang L, Li M, Liu Y, Bai Y, Yin T, Chen Y, Jiang J, Liu S. MOTS-c is an effective target for treating cancer-induced bone pain through the induction of AMPK-mediated mitochondrial biogenesis. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1323-1339. [PMID: 38716540 PMCID: PMC11532206 DOI: 10.3724/abbs.2024048] [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: 11/16/2023] [Accepted: 02/20/2024] [Indexed: 10/17/2024] Open
Abstract
Bone cancer pain (BCP), due to cancer bone metastasis and bone destruction, is a common symptom of tumors, including breast, prostate, and lung tumors. Patients often experience severe pain without effective treatment. Here, using a mouse model of bone cancer, we report that MOTS-c, a novel mitochondrial-derived peptide, confers remarkable protection against cancer pain and bone destruction. Briefly, we find that the plasma level of endogenous MOTS-c is significantly lower in the BCP group than in the sham group. Accordingly, intraperitoneal administration of MOTS-c robustly attenuates bone cancer-induced pain. These effects are blocked by compound C, an AMPK inhibitor. Furthermore, MOTS-c treatment significantly enhances AMPKα 1/2 phosphorylation. Interestingly, mechanical studies indicate that at the spinal cord level, MOTS-c relieves pain by restoring mitochondrial biogenesis, suppressing microglial activation, and decreasing the production of inflammatory factors, which directly contribute to neuronal modulation. However, in the periphery, MOTS-c protects against local bone destruction by modulating osteoclast and immune cell function in the tumor microenvironment, providing long-term relief from cancer pain. Additionally, we find that chronic administration of MOTS-c has little effect on liver, renal, lipid or cardiac function in mice. In conclusion, MOTS-c improves BCP through peripheral and central synergistic effects on nociceptors, immune cells, and osteoclasts, providing a pharmacological and biological rationale for the development of mitochondrial peptide-based therapeutic agents for cancer-induced pain.
Collapse
Affiliation(s)
- Long Yang
- Jiangsu Province Key Laboratory of AnesthesiologyJiangsu Province Key Laboratory of Anesthesia and Analgesia Application TechnologyNMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhou221004China
| | - Miaomiao Li
- Jiangsu Province Key Laboratory of AnesthesiologyJiangsu Province Key Laboratory of Anesthesia and Analgesia Application TechnologyNMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhou221004China
| | - Yucheng Liu
- Department of Anesthesiologythe Affiliated Hospital of Xuzhou Medical UniversityXuzhou221018China
| | - Yang Bai
- Jiangsu Province Key Laboratory of AnesthesiologyJiangsu Province Key Laboratory of Anesthesia and Analgesia Application TechnologyNMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhou221004China
| | - Tianyu Yin
- Department of Anesthesiologythe Affiliated Hospital of Xuzhou Medical UniversityXuzhou221018China
| | - Yangyang Chen
- Department of Anesthesiologythe Affiliated Hospital of Xuzhou Medical UniversityXuzhou221018China
| | - Jinhong Jiang
- Jiangsu Province Key Laboratory of AnesthesiologyJiangsu Province Key Laboratory of Anesthesia and Analgesia Application TechnologyNMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhou221004China
| | - Su Liu
- Jiangsu Province Key Laboratory of AnesthesiologyJiangsu Province Key Laboratory of Anesthesia and Analgesia Application TechnologyNMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhou221004China
- Department of Anesthesiologythe Affiliated Hospital of Xuzhou Medical UniversityXuzhou221018China
| |
Collapse
|
6
|
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
|
7
|
Wang Z, Yang L, Xu L, Liao J, Lu P, Jiang J. Central and peripheral mechanism of MOTS-c attenuates pain hypersensitivity in a mice model of inflammatory pain. Neurol Res 2024; 46:165-177. [PMID: 37899006 DOI: 10.1080/01616412.2023.2258584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 09/04/2023] [Indexed: 10/31/2023]
Abstract
BACKGROUND Inflammatory pain is caused by damaged tissue or noxious stimuli, accompanied by the release of inflammatory mediators that often leads to severe hyperalgesia and allodynia with limited therapy options. Recently, a novel mitochondrial-derived peptide (named MOTS-c) was reported to regulate obesity, metabolic homeostasis and inflammatory response. The aim of this study was to investigate the effects of MOTS-c and its related regulatory mechanisms involved in inflammatory pain. METHODS Male Kunming mice (8-10 weeks-old) were intraplantar injected with formalin, capsaicin, λ-Carrageenan and complete Freund adjuvant (CFA) to establish acute and chronic inflammatory pain. The effects of MOTS-c on the above inflammatory pain mice and its underlying mechanisms were examined by behavioral tests, quantitative polymerase chain reaction (qPCR), western blotting, enzyme linked immunosorbent assay (ELISA), immunohistochemistry (IHC) and immunofluorescence (IF). RESULTS Behavioral experiments investigated the potential beneficial effects of MOTS-c on multiple acute and chronic inflammatory pain in mice. The results showed that MOTS-c treatment produced potent anti-allodynic effects in formalin-induced acute inflammatory pain, capsaicin-induced nocifensive behaviors and λ-Carrageenan/CFA-induced chronic inflammatory pain model. Further mechanistic studies revealed that central MOTS-c treatment significantly ameliorated CFA-evoked the release of inflammatory factors and activation of glial cells and neurons in the spinal dorsal horn. Moreover, peripheral MOTS-c treatment reduced CFA-evoked inflammatory responses in the surface structure of hindpaw skin, accompanied by inhibiting excitation of peripheral calcitonin gene-related peptide (CGRP) and P2X3 nociceptive neurons. CONCLUSIONS The present study indicates that MOTS-c may serve as a promising therapeutic target for inflammatory pain.
Collapse
Affiliation(s)
- Zhe Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
- School of Food and Biological Engineering, Xuzhou University of Technology, Xuzhou, China
| | - Long Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lingfei Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jinglei Liao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ping Lu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jinhong Jiang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
8
|
Morais MÍ, Braga AV, Silva RRL, Barbosa BCM, Costa SOAM, Rodrigues FF, Melo ISF, Matos RC, Carobin NV, Sabino AP, Coelho MM, Machado RR. Metformin inhibits paclitaxel-induced mechanical allodynia by activating opioidergic pathways and reducing cytokines production in the dorsal root ganglia and thalamus. Cytokine 2024; 174:156468. [PMID: 38101167 DOI: 10.1016/j.cyto.2023.156468] [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: 09/25/2023] [Revised: 11/17/2023] [Accepted: 12/03/2023] [Indexed: 12/17/2023]
Abstract
It has been shown that AMP-activated protein kinase (AMPK) is involved in the nociceptive processing. This observation has prompted us to investigate the effects of the AMPK activator metformin on the paclitaxel-induced mechanical allodynia, a well-established model of neuropathic pain. Mechanical allodynia was induced by four intraperitoneal (i.p) injections of paclitaxel (2 mg/kg.day) in mice. Metformin was administered per os (p.o.). Naltrexoneandglibenclamide were used to investigate mechanisms mediating metformin activity. Concentrations of cytokines in the dorsal root ganglia (DRG) and thalamus were determined. After a single p.o. administration, the two highest doses of metformin (500 and 1000 mg/kg) attenuated the mechanical allodynia. This response was attenuated by all doses of metformin (250, 500 and 1000 mg/kg) when two administrations, 2 h apart, were carried out. Naltrexone (5 and 10 mg/kg, i.p.), but not glibenclamide (20 and 40 mg/kg, p.o.), attenuated metformin activity. Concentrations of tumor necrosis factor (TNF)-α, interleukin (IL)-1β and CXCL-1 in the DRG were increased after administration of paclitaxel. Metformin (1000 mg/kg) reduced concentrations of TNF-α, IL-1β and CXCL-1 in the DRG. Concentration of IL-6, but not TNF-α, in the thalamus was increased after administration of paclitaxel. Metformin (1000 mg/kg) reduced concentration of IL-6 in the thalamus. In summary, metformin exhibits activity in the model of neuropathic pain induced by paclitaxel. This activity may be mediated by activation of opioidergic pathways and reduced production of TNF-α, IL-1β and CXCL-1 in the DRG and IL-6 in the thalamus.
Collapse
Affiliation(s)
- Marcela Í Morais
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alysson V Braga
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Roger R L Silva
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bárbara C M Barbosa
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Sarah O A M Costa
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Felipe F Rodrigues
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ivo S F Melo
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rafael C Matos
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Natália V Carobin
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Adriano P Sabino
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Márcio M Coelho
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Renes R Machado
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
9
|
Xu L, Tang X, Yang L, Chang M, Xu Y, Chen Q, Lu C, Liu S, Jiang J. Mitochondria-derived peptide is an effective target for treating streptozotocin induced painful diabetic neuropathy through induction of activated protein kinase/peroxisome proliferator-activated receptor gamma coactivator 1alpha -mediated mitochondrial biogenesis. Mol Pain 2024; 20:17448069241252654. [PMID: 38658141 PMCID: PMC11113074 DOI: 10.1177/17448069241252654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/12/2024] [Accepted: 04/10/2024] [Indexed: 04/26/2024] Open
Abstract
Painful Diabetic Neuropathy (PDN) is a common diabetes complication that frequently causes severe hyperalgesia and allodynia and presents treatment challenges. Mitochondrial-derived peptide (MOTS-c), a novel mitochondrial-derived peptide, has been shown to regulate glucose metabolism, insulin sensitivity, and inflammatory responses. This study aimed to evaluate the effects of MOTS-c in streptozocin (STZ)-induced PDN model and investigate the putative underlying mechanisms. We found that endogenous MOTS-c levels in plasma and spinal dorsal horn were significantly lower in STZ-treated mice than in control animals. Accordingly, MOTS-c treatment significantly improves STZ-induced weight loss, elevation of blood glucose, mechanical allodynia, and thermal hyperalgesia; however, these effects were blocked by dorsomorphin, an adenosine monophosphate-activated protein kinase (AMPK) inhibitor. In addition, MOTS-c treatment significantly enhanced AMPKα1/2 phosphorylation and PGC-1α expression in the lumbar spinal cord of PDN mice. Mechanistic studies indicated that MOTS-c significantly restored mitochondrial biogenesis, inhibited microglia activation, and decreased the production of pro-inflammatory factors, which contributed to the alleviation of pain. Moreover, MOTS-c decreased STZ-induced pain hypersensitivity in PDN mice by activating AMPK/PGC-1α signaling pathway. This provides the pharmacological and biological evidence for developing mitochondrial peptide-based therapeutic agents for PDN.
Collapse
Affiliation(s)
- Lingfei Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Xihui Tang
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Long Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Min Chang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Yuqing Xu
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qingsong Chen
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chen Lu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Su Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jinhong Jiang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
10
|
Yousuf MS, Sahn JJ, Yang H, David ET, Shiers S, Mancilla Moreno M, Iketem J, Royer DM, Garcia CD, Zhang J, Hong VM, Mian SM, Ahmad A, Kolber BJ, Liebl DJ, Martin SF, Price TJ. Highly specific σ 2R/TMEM97 ligand FEM-1689 alleviates neuropathic pain and inhibits the integrated stress response. Proc Natl Acad Sci U S A 2023; 120:e2306090120. [PMID: 38117854 PMCID: PMC10756276 DOI: 10.1073/pnas.2306090120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 11/21/2023] [Indexed: 12/22/2023] Open
Abstract
The sigma 2 receptor (σ2R) was described pharmacologically more than three decades ago, but its molecular identity remained obscure until recently when it was identified as transmembrane protein 97 (TMEM97). We and others have shown that σ2R/TMEM97 ligands alleviate mechanical hypersensitivity in mouse neuropathic pain models with a time course wherein maximal antinociceptive effect is approximately 24 h following dosing. We sought to understand this unique antineuropathic pain effect by addressing two key questions: do these σ2R/TMEM97 compounds act selectively via the receptor, and what is their downstream mechanism on nociceptive neurons? Using male and female conventional knockout mice for Tmem97, we find that a σ2R/TMEM97 binding compound, FEM-1689, requires the presence of the gene to produce antinociception in the spared nerve injury model in mice. Using primary mouse dorsal root ganglion neurons, we demonstrate that FEM-1689 inhibits the integrated stress response (ISR) and promotes neurite outgrowth via a σ2R/TMEM97-specific action. We extend the clinical translational value of these findings by showing that FEM-1689 reduces ISR and p-eIF2α levels in human sensory neurons and that it alleviates the pathogenic engagement of ISR by methylglyoxal. We also demonstrate that σ2R/TMEM97 is expressed in human nociceptors and satellite glial cells. These results validate σ2R/TMEM97 as a promising target for further development for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Muhammad Saad Yousuf
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX75080
- NuvoNuro Inc., Austin, TX78712
| | - James J. Sahn
- NuvoNuro Inc., Austin, TX78712
- Department of Chemistry, University of Texas at Austin, Austin, TX78712
| | - Hongfen Yang
- Department of Chemistry, University of Texas at Austin, Austin, TX78712
| | - Eric T. David
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX75080
| | - Stephanie Shiers
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX75080
| | - Marisol Mancilla Moreno
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX75080
| | - Jonathan Iketem
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX75080
| | - Danielle M. Royer
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX75080
| | - Chelsea D. Garcia
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX75080
| | - Jennifer Zhang
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX75080
| | - Veronica M. Hong
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX75080
| | - Subhaan M. Mian
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX75080
| | - Ayesha Ahmad
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX75080
| | - Benedict J. Kolber
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX75080
| | - Daniel J. Liebl
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL33136
| | - Stephen F. Martin
- NuvoNuro Inc., Austin, TX78712
- Department of Chemistry, University of Texas at Austin, Austin, TX78712
| | - Theodore J. Price
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX75080
- NuvoNuro Inc., Austin, TX78712
| |
Collapse
|
11
|
Wong C, Tavares-Ferreira D, Thörn Perez C, Sharif B, Uttam S, Amiri M, Lister KC, Hooshmandi M, Nguyen V, Séguéla P, Sonenberg N, Price TJ, Gkogkas CG, Khoutorsky A. 4E-BP1-dependent translation in nociceptors controls mechanical hypersensitivity via TRIM32/type I interferon signaling. SCIENCE ADVANCES 2023; 9:eadh9603. [PMID: 37922363 PMCID: PMC10624345 DOI: 10.1126/sciadv.adh9603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 10/03/2023] [Indexed: 11/05/2023]
Abstract
Activation of the mechanistic target of rapamycin complex 1 (mTORC1) contributes to the development of chronic pain. However, the specific mechanisms by which mTORC1 causes hypersensitivity remain elusive. The eukaryotic initiation factor 4E-binding protein 1 (4E-BP1) is a key mTORC1 downstream effector that represses translation initiation. Here, we show that nociceptor-specific deletion of 4E-BP1, mimicking activation of mTORC1-dependent translation, is sufficient to cause mechanical hypersensitivity. Using translating ribosome affinity purification in nociceptors lacking 4E-BP1, we identified a pronounced translational up-regulation of tripartite motif-containing protein 32 (TRIM32), an E3 ubiquitin ligase that promotes interferon signaling. Down-regulation of TRIM32 in nociceptors or blocking type I interferon signaling reversed the mechanical hypersensitivity in mice lacking 4E-BP1. Furthermore, nociceptor-specific ablation of TRIM32 alleviated mechanical hypersensitivity caused by tissue inflammation. These results show that mTORC1 in nociceptors promotes hypersensitivity via 4E-BP1-dependent up-regulation of TRIM32/interferon signaling and identify TRIM32 as a therapeutic target in inflammatory pain.
Collapse
Affiliation(s)
- Calvin Wong
- Department of Anaesthesia, McGill University, Montreal, Canada
| | - Diana Tavares-Ferreira
- School of Behavioural and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, TX 75080, USA
| | - Carolina Thörn Perez
- Department of Anaesthesia, McGill University, Montreal, Canada
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Behrang Sharif
- Department of Physiology, McGill University, Montreal, Canada
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| | - Sonali Uttam
- Department of Anaesthesia, McGill University, Montreal, Canada
| | - Mehdi Amiri
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Canada
- Department of Biochemistry, McGill University, Montreal, Canada
| | - Kevin C. Lister
- Department of Anaesthesia, McGill University, Montreal, Canada
| | | | - Vivienne Nguyen
- Department of Anaesthesia, McGill University, Montreal, Canada
| | - Philippe Séguéla
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Nahum Sonenberg
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Canada
- Department of Biochemistry, McGill University, Montreal, Canada
| | - Theodore J. Price
- School of Behavioural and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, TX 75080, USA
| | - Christos G. Gkogkas
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, 45110 Ioannina, Greece
| | - Arkady Khoutorsky
- Department of Anaesthesia, McGill University, Montreal, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| |
Collapse
|
12
|
曹 静, 刘 海, 安 琪, 韩 枫. [Metformin alleviates pathologic pain in mice with radiation dermatitis by inhibiting p38MAPK/NF-κB signaling pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:1815-1820. [PMID: 37933660 PMCID: PMC10630197 DOI: 10.12122/j.issn.1673-4254.2023.10.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Indexed: 11/08/2023]
Abstract
OBJECTIVE To observe the therapeutic effect of metformin on pathological pain in mice with radiation dermatitis and explore the underlying mechanism. METHODS Thirty-two male adult ICR mice were randomized into normal control group, radiation dermatitis model group, metformin treatment (200 mg/kg) group and gabapentin (100 mg/kg) group (n=8).In the latter two groups, metformin treatment was administered after modeling via intraperitoneal injection and gabapentin by gavage on a daily basis for 16 days; the mice in the control group and model group received intraperitoneal injection of normal saline.After the last administration, radiation dermatitis was graded in each group.Mechanical withdraw threshold (MWT) and thermal withdrawal latency (TWL) of the mice were tested one day before and at 1, 4, 8, 12 and 16 days after modeling.Western blotting was used to measure the protein expression levels of p38MAPK, p-p38MAPK, NF-κB p65 and p-NF-κB p65 in the L4-L6 spinal cord, and the concentrations of IL-1β, IL-6 and TNF-α in the spinal cord tissue were determined with ELISA. RESULTS Compared with those in the control group, the mice in the other 3 groups showed obvious symptoms of radiation dermatitis after modeling (P<0.05), which were significantly alleviated by treatment with metformin (P<0.05).The mice in the model group exhibited significant decreases in MWT and TWL (P<0.05), which were improved by treatment with metformin and gabapentin (P<0.05).Compared with those in the model group, the levels of p-p38MAPK, p-NF-κB p65, IL-1β, IL-6 and TNF-α in the spinal cord were significantly decreased in the mice after metformin treatment (P<0.05). CONCLUSION Metformin can significantly ameliorate pathological pain symptoms in mice with radiation dermatitis possibly by inhibiting the activation of p38MAPK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- 静 曹
- />连云港市东方医院皮肤科,江苏 连云港 222042Department of Dermatology, Lianyungang Oriental Hospital, Lianyungang 222042, China
| | - 海波 刘
- />连云港市东方医院皮肤科,江苏 连云港 222042Department of Dermatology, Lianyungang Oriental Hospital, Lianyungang 222042, China
| | - 琪 安
- />连云港市东方医院皮肤科,江苏 连云港 222042Department of Dermatology, Lianyungang Oriental Hospital, Lianyungang 222042, China
| | - 枫 韩
- />连云港市东方医院皮肤科,江苏 连云港 222042Department of Dermatology, Lianyungang Oriental Hospital, Lianyungang 222042, China
| |
Collapse
|
13
|
Yousuf MS, Sahn JJ, Yang H, David ET, Shiers S, Moreno MM, Iketem J, Royer DM, Garcia CD, Zhang J, Hong VM, Mian SM, Ahmad A, Kolber BJ, Liebl DJ, Martin SF, Price TJ. Highly specific σ 2R/TMEM97 ligand alleviates neuropathic pain and inhibits the integrated stress response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.11.536439. [PMID: 37090527 PMCID: PMC10120691 DOI: 10.1101/2023.04.11.536439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
The Sigma 2 receptor (σ2R) was described pharmacologically more than three decades ago, but its molecular identity remained obscure until recently when it was identified as transmembrane protein 97 (TMEM97). We and others have shown that σ2R/TMEM97 ligands alleviate mechanical hypersensitivity in mouse neuropathic pain models with a time course wherein maximal anti-nociceptive effect is approximately 24 hours following dosing. We sought to understand this unique anti-neuropathic pain effect by addressing two key questions: do these σ2R/TMEM97 compounds act selectively via the receptor, and what is their downstream mechanism on nociceptive neurons? Using male and female conventional knockout (KO) mice for Tmem97, we find that a new σ2R/TMEM97 binding compound, FEM-1689, requires the presence of the gene to produce anti-nociception in the spared nerve injury model in mice. Using primary mouse dorsal root ganglion (DRG) neurons, we demonstrate that FEM-1689 inhibits the integrated stress response (ISR) and promotes neurite outgrowth via a σ2R/TMEM97-specific action. We extend the clinical translational value of these findings by showing that FEM-1689 reduces ISR and p-eIF2α levels in human sensory neurons and that it alleviates the pathogenic engagement of ISR by methylglyoxal. We also demonstrate that σ2R/TMEM97 is expressed in human nociceptors and satellite glial cells. These results validate σ2R/TMEM97 as a promising target for further development for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Muhammad Saad Yousuf
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
- NuvoNuro, Austin, TX 78712
| | - James J. Sahn
- Department of Chemistry, University of Texas at Austin, Austin, TX 78712
- NuvoNuro, Austin, TX 78712
| | - Hongfen Yang
- Department of Chemistry, University of Texas at Austin, Austin, TX 78712
| | - Eric T. David
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Stephanie Shiers
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Marisol Mancilla Moreno
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Jonathan Iketem
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Danielle M. Royer
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Chelsea D. Garcia
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Jennifer Zhang
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Veronica M. Hong
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Subhaan M. Mian
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Ayesha Ahmad
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Benedict J. Kolber
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | | | - Stephen F. Martin
- Department of Chemistry, University of Texas at Austin, Austin, TX 78712
- NuvoNuro, Austin, TX 78712
| | - Theodore J. Price
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
- NuvoNuro, Austin, TX 78712
| |
Collapse
|
14
|
Bakry HM, Mansour NO, ElKhodary TR, Soliman MM. Efficacy of metformin in prevention of paclitaxel-induced peripheral neuropathy in breast cancer patients: a randomized controlled trial. Front Pharmacol 2023; 14:1181312. [PMID: 37583905 PMCID: PMC10424931 DOI: 10.3389/fphar.2023.1181312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/17/2023] [Indexed: 08/17/2023] Open
Abstract
Background: Paclitaxel-induced peripheral neuropathy (PN) is a serious clinical problem with no approved drug for prevention. This study aimed to examine the neuroprotective effect of metformin against paclitaxel-induced PN in breast cancer patients. Methods: Patients with confirmed breast cancer diagnosis who were planned to receive paclitaxel were randomized to receive either metformin or placebo. Both groups received the standard chemotherapy protocol for breast cancer. Patients started metformin/placebo 1 week before paclitaxel initiation and continued study interventions thereafter for nine consecutive weeks. The primary outcome was the incidence of development of grade two or more paclitaxel-induced sensory PN. The PN was graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE). Patients' quality of life (QoL) was assessed by the Functional Assessment of Cancer Therapy/Gynecologic Oncology Group-Neurotoxicity (FACTGOG-Ntx) subscale. Pain severity was measured by the Brief Pain Inventory Short Form (BPI-SF). Serum levels of nerve growth factor (NGF) and neurotensin (NT) were measured at baseline and at the end paclitaxel treatment. Results: A total of 73 patients (36 in the metformin arm and 37 in the control arm) were evaluated. The cumulative incidence of development of grade two or more PN was significantly lower in the metformin arm (14 (38.9%) than the control arm (28 (75.7%); p = 0.001). At the end of paclitaxel treatment, patients' QoL was significantly better in the metformin arm [median (IQR) FACTGOG-Ntx subscale of (24.0 (20.5-26.5)] compared to the control arm (21.0 (18.0-24.0); p = 0.003). The metformin arm showed lower "average" and "worst" pain scores than those detected in the control arm. At the end of the paclitaxel treatment, there was a significant difference in the median serum NGF levels between the two arms, favoring metformin (p < 0.05), while NT serum levels were deemed comparable between the two study arms (p = 0.09). Conclusion: The use of metformin in breast cancer patients offered a marked protection against paclitaxel-induced PN, which translated to better patient QoL. Clinical Trial Registration: https://classic.clinicaltrials.gov/ct2/show/NCT05351021, identifier NCT05351021.
Collapse
Affiliation(s)
- Hala M. Bakry
- Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Noha O. Mansour
- Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Tawfik R. ElKhodary
- Oncology Center, Medical Oncology Unit, Mansoura University, Mansoura, Egypt
| | - Moetaza M. Soliman
- Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
15
|
Pușcașu C, Ungurianu A, Șeremet OC, Andrei C, Mihai DP, Negreș S. The Influence of Sildenafil-Metformin Combination on Hyperalgesia and Biochemical Markers in Diabetic Neuropathy in Mice. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1375. [PMID: 37629665 PMCID: PMC10456948 DOI: 10.3390/medicina59081375] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023]
Abstract
Background and objectives: Worldwide, approximately 500 million people suffer from diabetes and at least 50% of these people develop neuropathy. Currently, therapeutic strategies for reducing diabetic neuropathy (DN)-associated pain are limited and have several side effects. The purpose of the study was to evaluate the antihyperalgesic action of different sildenafil (phosphodiesterase-5 inhibitor) and metformin (antihyperglycemic agent) combinations in alloxan-induced DN. Methods: The study included 100 diabetic mice and 20 non-diabetic mice that were subjected to hot and cold stimulus tests. Furthermore, we determined the influence of this combination on TNF-α, IL-6 and nitrites levels in brain and liver tissues. Results: In both the hot-plate and tail withdrawal test, all sildenafil-metformin combinations administered in our study showed a significant increase in pain reaction latencies when compared to the diabetic control group. Furthermore, all combinations decreased blood glucose levels due to the hypoglycemic effect of metformin. Additionally, changes in nitrite levels and pro-inflammatory cytokines (TNF-α and IL-6) were observed after 14 days of treatment with different sildenafil-metformin combinations. Conclusions: The combination of these two substances increased the pain reaction latency of diabetic animals in a dose-dependent manner. Moreover, all sildenafil-metformin combinations significantly reduced the concentration of nitrites in the brain and liver, which are final products formed under the action of iNOS.
Collapse
Affiliation(s)
| | | | - Oana Cristina Șeremet
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (C.P.); (S.N.)
| | | | | | | |
Collapse
|
16
|
Liu TT, Qiu CY, Hu WP. Metformin inhibits spontaneous excitatory postsynaptic currents in spinal dorsal cord neurons from paclitaxel-treated rats. Front Synaptic Neurosci 2023; 15:1191383. [PMID: 37216004 PMCID: PMC10195993 DOI: 10.3389/fnsyn.2023.1191383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
Introduction Cancer patients treated with paclitaxel often develop chemotherapy-induced peripheral neuropathy, which has not been effectively treated with drugs. The anti-diabetic drug metformin is effective in the treatment of neuropathic pain. The aim of this study was to elucidate effect of metformin on paclitaxel-induced neuropathic pain and spinal synaptic transmission. Methods Electrophysiological experiments on rat spinal slices were performed in vitro and mechanical allodynia quantified in vitro. Results The present data demonstrated that intraperitoneal injection of paclitaxel produced mechanical allodynia and potentiated spinal synaptic transmission. Intrathecal injection of metformin significantly reversed the established mechanical allodynia induced by paclitaxel in rats. Either spinal or systemic administration of metformin significantly inhibited the increased frequency of spontaneous excitatory postsynaptic currents (sEPSCs) in spinal dorsal horn neurons from paclitaxel-treated rats. We found that 1 h incubation of metformin also reduced the frequency rather than the amplitude of sEPSCs in the spinal slices from paclitaxel-treated rats. Discussion These results suggested that metformin was able to depress the potentiated spinal synaptic transmission, which may contribute to alleviating the paclitaxel-induced neuropathic pain.
Collapse
Affiliation(s)
- Ting-Ting Liu
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Chun-Yu Qiu
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Wang-Ping Hu
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
- Department of Physiology, Hubei College of Chinese Medicine, Jingzhou, Hubei, China
| |
Collapse
|
17
|
Ray PR, Shiers S, Caruso JP, Tavares-Ferreira D, Sankaranarayanan I, Uhelski ML, Li Y, North RY, Tatsui C, Dussor G, Burton MD, Dougherty PM, Price TJ. RNA profiling of human dorsal root ganglia reveals sex differences in mechanisms promoting neuropathic pain. Brain 2023; 146:749-766. [PMID: 35867896 PMCID: PMC10169414 DOI: 10.1093/brain/awac266] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 05/16/2022] [Accepted: 06/22/2022] [Indexed: 11/12/2022] Open
Abstract
Neuropathic pain is a leading cause of high-impact pain, is often disabling and is poorly managed by current therapeutics. Here we focused on a unique group of neuropathic pain patients undergoing thoracic vertebrectomy where the dorsal root ganglia is removed as part of the surgery allowing for molecular characterization and identification of mechanistic drivers of neuropathic pain independently of preclinical models. Our goal was to quantify whole transcriptome RNA abundances using RNA-seq in pain-associated human dorsal root ganglia from these patients, allowing comprehensive identification of molecular changes in these samples by contrasting them with non-pain-associated dorsal root ganglia. We sequenced 70 human dorsal root ganglia, and among these 50 met inclusion criteria for sufficient neuronal mRNA signal for downstream analysis. Our expression analysis revealed profound sex differences in differentially expressed genes including increase of IL1B, TNF, CXCL14 and OSM in male and CCL1, CCL21, PENK and TLR3 in female dorsal root ganglia associated with neuropathic pain. Coexpression modules revealed enrichment in members of JUN-FOS signalling in males and centromere protein coding genes in females. Neuro-immune signalling pathways revealed distinct cytokine signalling pathways associated with neuropathic pain in males (OSM, LIF, SOCS1) and females (CCL1, CCL19, CCL21). We validated cellular expression profiles of a subset of these findings using RNAscope in situ hybridization. Our findings give direct support for sex differences in underlying mechanisms of neuropathic pain in patient populations.
Collapse
Affiliation(s)
- Pradipta R Ray
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - James P Caruso
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA.,Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Megan L Uhelski
- Department of Pain Medicine, Division of Anesthesiology, MD Anderson Cancer Center, Houston, TX, USA
| | - Yan Li
- Department of Pain Medicine, Division of Anesthesiology, MD Anderson Cancer Center, Houston, TX, USA
| | - Robert Y North
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Claudio Tatsui
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Gregory Dussor
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Michael D Burton
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Patrick M Dougherty
- Department of Pain Medicine, Division of Anesthesiology, MD Anderson Cancer Center, Houston, TX, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
18
|
Downregulation of miR-137 Facilitates CD4+ T Cell Pyroptosis in Systemic Lupus Erythematosus via Stimulating AMPK Pathway. J Immunol Res 2023; 2023:1241774. [PMID: 36815949 PMCID: PMC9936506 DOI: 10.1155/2023/1241774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/03/2022] [Accepted: 12/17/2022] [Indexed: 02/11/2023] Open
Abstract
Objective From the pathogenic mechanism point of view, systemic lupus erythematosus (SLE) features prominently in T lymphocyte apoptosis. Yet the regulatory mechanism underlying SLE cell apoptosis remains to be explored. This research intends to clarify the role played by miR-137 in SLE and the underlying mechanisms. Methods Twenty SLE patients (SLE group) and twenty healthy controls (control group) were selected, from whom peripheral blood CD4+ T cells were isolated via magnetic-activated cell sorting. Reverse transcription-polymerase chain reaction (RT-PCR) quantified miR-137 and AMP-activated protein kinase (AMPK) in CD4+ T cells. Further, transfection of miR-137 mimics and inhibitors into CD4+ T cells was carried out to alter miR levels. Levels of pyroptosis, apoptosis, and inflammatory- and pyroptosis-related proteins were determined through PI staining, flow cytometry, and Western blotting, respectively. A luciferase reporter gene assay identified the targeting relation between miR-137 and AMPK. Results SLE patients showed downregulated miR-137 and upregulated AMPK in CD4+ T cells than controls. miR-137 upregulation by miR-137 mimic transfection inhibited Jurkat cell pyroptosis and apoptosis at both mRNA and protein levels and suppressed NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasome activity and pyroptosis-related protein gasdermin D (GSDMD), while miR-137 inhibitor transfection contributed to completely opposite effects. miR-137 directly targeted AMPK, as indicated by the luciferase reporter gene assay. Furthermore, miR-137 inhibitor intervention induced healthy CD4+ T cell pyroptosis and apoptosis via mediating AMPK, whereas miR-137 mimic transfection into CD4+ T cells of SLE patients leads to opposite results. Conclusion Upregulating miR-137 inhibits CD4+ T cell pyroptosis in SLE patients by modulating the AMPK pathway, suggesting the potential diagnostic and therapeutic role of miR-137 in SLE.
Collapse
|
19
|
König C, Ebersberger A, Eitner A, Wetzker R, Schaible HG. Prostaglandin EP3 receptor activation is antinociceptive in sensory neurons via PI3Kγ, AMPK and GRK2. Br J Pharmacol 2023; 180:441-458. [PMID: 36245399 DOI: 10.1111/bph.15971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 07/22/2022] [Accepted: 09/29/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Prostaglandin E2 is considered a major mediator of inflammatory pain, by acting on neuronal Gs protein-coupled EP2 and EP4 receptors. However, the neuronal EP3 receptor, colocalized with EP2 and EP4 receptor, is Gi protein-coupled and antagonizes the pronociceptive prostaglandin E2 effect. Here, we investigated the cellular signalling mechanisms by which the EP3 receptor reduces EP2 and EP4 receptor-evoked pronociceptive effects in sensory neurons. EXPERIMENTAL APPROACH Experiments were performed on isolated and cultured dorsal root ganglion (DRG) neurons from wild type, phosphoinositide 3-kinase γ (PI3Kγ)-/- , and PI3Kγkinase dead (KD)/KD mice. For subtype-specific stimulations, we used specific EP2, EP3, and EP4 receptor agonists from ONO Pharmaceuticals. As a functional readout, we recorded TTX-resistant sodium currents in patch-clamp experiments. Western blots were used to investigate the activation of intracellular signalling pathways. EP4 receptor internalization was measured using immunocytochemistry. KEY RESULTS Different pathways mediate the inhibition of EP2 and EP4 receptor-dependent pronociceptive effects by EP3 receptor stimulation. Inhibition of EP2 receptor-evoked pronociceptive effect critically depends on the kinase-independent function of the signalling protein PI3Kγ, and adenosine monophosphate activated protein kinase (AMPK) is involved. By contrast, inhibition of EP4 receptor-evoked pronociceptive effect is independent on PI3Kγ and mediated through activation of G protein-coupled receptor kinase 2 (GRK2), which enhances the internalization of the EP4 receptor after ligand binding. CONCLUSION AND IMPLICATIONS Activation of neuronal PI3Kγ, AMPK, and GRK2 by EP3 receptor activation limits cAMP-dependent pain generation by prostaglandin E2 . These new insights hold the potential for a novel approach in pain therapy.
Collapse
Affiliation(s)
- Christian König
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| | - Andrea Ebersberger
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| | - Annett Eitner
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany.,Department of Trauma, Hand and Reconstructive Surgery, Experimental Trauma Surgery, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| | - Reinhard Wetzker
- Clinic for Anesthesiology and Intensive Care, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| | - Hans-Georg Schaible
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| |
Collapse
|
20
|
Li X, Jin DS, Eadara S, Caterina MJ, Meffert MK. Regulation by noncoding RNAs of local translation, injury responses, and pain in the peripheral nervous system. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2023; 13:100119. [PMID: 36798094 PMCID: PMC9926024 DOI: 10.1016/j.ynpai.2023.100119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/17/2023] [Accepted: 01/21/2023] [Indexed: 06/18/2023]
Abstract
Neuropathic pain is a chronic condition arising from damage to somatosensory pathways that results in pathological hypersensitivity. Persistent pain can be viewed as a consequence of maladaptive plasticity which, like most enduring forms of cellular plasticity, requires altered expression of specific gene programs. Control of gene expression at the level of protein synthesis is broadly utilized to directly modulate changes in activity and responsiveness in nociceptive pathways and provides an effective mechanism for compartmentalized regulation of the proteome in peripheral nerves through local translation. Levels of noncoding RNAs (ncRNAs) are commonly impacted by peripheral nerve injury leading to persistent pain. NcRNAs exert spatiotemporal regulation of local proteomes and affect signaling cascades supporting altered sensory responses that contribute to hyperalgesia. This review discusses ncRNAs found in the peripheral nervous system (PNS) that are dysregulated following nerve injury and the current understanding of their roles in pathophysiological pain-related responses including neuroimmune interactions, neuronal survival and axon regeneration, Schwann cell dedifferentiation and proliferation, intercellular communication, and the generation of ectopic action potentials in primary afferents. We review progress in the field beyond cataloging, with a focus on the relevant target transcripts and mechanisms underlying pain modulation by ncRNAs.
Collapse
Affiliation(s)
- Xinbei Li
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, United States
| | - Daniel S. Jin
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, United States
| | - Sreenivas Eadara
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, United States
| | - Michael J. Caterina
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, United States
- Department of Neurosurgery and Neurosurgery Pain Research Institute, Johns Hopkins University School of Medicine, United States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, United States
| | - Mollie K. Meffert
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, United States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, United States
| |
Collapse
|
21
|
Drug repurposing – A search for novel therapy for the treatment of diabetic neuropathy. Biomed Pharmacother 2022; 156:113846. [DOI: 10.1016/j.biopha.2022.113846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/27/2022] [Accepted: 10/06/2022] [Indexed: 11/23/2022] Open
|
22
|
Cui Y, Hu C, Niu C, He M, Qiu X, Yao Q, Tian W, Xu Q. Electroacupuncture attenuates spared nerve injury-induced neuropathic pain possibly by promoting the progression of AMPK/mTOR-mediated autophagy in spinal microglia. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1278. [PMID: 36618785 PMCID: PMC9816825 DOI: 10.21037/atm-22-5273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 11/24/2022] [Indexed: 12/15/2022]
Abstract
Background Neuropathic pain (NP) is a syndrome that arises from central or peripheral nerve injury, which manifests primarily as hyperalgesia, spontaneous pain, and allodynia. The recent trend has exhibited a shift towards the development of therapies for managing NP. Activation of autophagy is involved in the function of the glial cells, which may be implicated further to attenuate pain. Methods In this study, the analgesic effects of electroacupuncture (EA) were evaluated among NP rats developed using spared nerve injury (SNI). Acupuncture treatment or EA was carried out after 7 days of SNI at two acupoints, i.e., the Zusanli (ST36) and Huantiao (GB30). Results The application of EA was found to attenuate mechanical hyperalgesia. The marker protein for microglial cells (CD11b) alone, without either the astrocyte marker or neuronal marker, was co-expressed with the autophagy indicator p62, as illustrated with immunofluorescence staining. Western blotting demonstrated that the expression levels of p62, Beclin-1, and LC3-II/LC3-I were elevated in the spinal cords of rats in the SNI group compared to the control levels. EA treatment resulted in reduced expression of p62, while the expressions of Beclin-1 and LC3-II/LC3-I were increased. The electron microscopy results indicated that EA could induce autophagy progression in the microglia of the spinal dorsal horn in SNI rats. Furthermore, we explored the causal relationship between EA-induced inhibition of NP and increased autophagic levels in microglia using the AMPK inhibitor compound C, and found that the mechanism of EA-induced analgesia may contribute to the promotion of AMPK/mTOR-mediated autophagy in spinal microglia. Conclusions Our work showed that the analgesic impact of EA is partly related to AMPK/mTOR pathway activation and autophagy induction in microglial cells, providing a potential therapeutic target for NP.
Collapse
Affiliation(s)
- Yaomei Cui
- Department of Anesthesiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Cheng Hu
- Department of Pain Management, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Cong Niu
- Department of Anesthesiology, The Second affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Menglin He
- The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xizi Qiu
- The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Qiang Yao
- The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Weiqian Tian
- Department of Anesthesiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Qian Xu
- Department of Anesthesiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| |
Collapse
|
23
|
Analgesic Effectiveness and Dorsal Root Ganglia Protein Modulation of a Peripheral Adenosine Monophosphate Kinase Alpha Activator (O304) Following Lumbar Disk Puncture in the Mouse. Anesth Analg 2022; 135:1293-1303. [PMID: 36201356 DOI: 10.1213/ane.0000000000006228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Disk herniation is a primary cause of radicular back pain. The purpose of this study was to evaluate the antiallodynic effective dose in 50% of the sample (ED 50 ) and dorsal root ganglion (DRG) protein modulation of a peripheral direct adenosine monophosphate kinase alpha (AMPKα) activator (O304) in a murine model of lumbar disk puncture. METHODS Male (n = 28) and female (n = 28) mice (C57BL6/J) were assessed for hind paw withdrawal threshold (PWT) and burrowing. Abdominal surgery was performed on all mice, and 48 received a lumbar disk puncture (27-G needle), with 8 serving as nondisk puncture controls. Assessments were repeated at day 7, and mice were then randomized into 5 groups of equal numbers of males and females: O304 at 100 mg/kg (n = 10), 150 mg/kg (n = 10), 200 mg/kg (n = 10), and 250 mg/kg (n = 10) or drug vehicle (n = 8). Starting on day 7, mice received daily gavages of O304 or vehicle for 7 days. On days 14 and 21 PWT and on day 14 burrowing were assessed. The area under the PWT by time curve (AUC) from day 7 to 21 was determined by trapezoidal integration. DRG protein modulation was evaluated in male (n = 10) and female (n = 10) mice (C57BL6/J). Following disk puncture, mice were randomized to receive O304 200 mg/kg or vehicle for 7 days starting on day 7. On day 14, mice were euthanized; the DRG harvested and immunoblot performed for mammalian target of rapamycin (mTOR), transient receptor potential ankyrin 1 (TRPA1), phosphorylated adenosine monophosphate kinase (p-AMPK), phosphorylated extracellular signal-regulated kinase (p-ERK), phosphorylated eukaryotic translation initiation factor 2 subunit 1 (p-EIF2S1), phosphorylated eukaryotic translation initiation factor 4e (p-EIF4E), and glyceraldehyde 3-phosphate dehydrogenase (GADPH). RESULTS Disk puncture decreased PWT greater in female mice compared with male mice and decreased burrowing at 7 days. PWTs were increased with increasing doses of O304 from 150 to 250 mg/g on day 14 and sustained through day 21. The ED 50 (95% confidence interval [CI]) for reducing mechanical allodynia was 140 (118-164) mg/kg. Burrowing was not increased at day 14 compared to day 7 by O304 administration. Compared to vehicle-treated animals, O304 increased (95% CI) the p-AMPK/GADPH ratio, difference 0.27 (0.08-0.45; P = . 004) and decreased (95% CI) the ratios of p-TRPA1, p-ERK1/2, pEIF4E, and p-EIF2S1 to GADPH by -0.49 (-0.61 to -0.37; P < . 001), -0.53 (-0.76 to -0.29; P < . 001), -0.27 (-0.42 to 0.11; P = . 001), and -0.21 (-0.32 to -0.08; P = . 003) in the DRG, respectively. CONCLUSIONS The direct peripheral AMPK activator O304 reduced allodynia in a dose-dependent manner, and immunoblot studies of the DRG showed that O304 increased p-AMPK and decreased TRPA1, p-ERK1/2, as well as translation factors involved in neuroplasticity. Our findings confirm the role of peripheral AMPKα activation in modulating nociceptive pain.
Collapse
|
24
|
Xu Q, Niu C, Li J, Hu C, He M, Qiu X, Yao Q, Tian W, Zhang M. Electroacupuncture alleviates neuropathic pain caused by spared nerve injury by promoting AMPK/mTOR-mediated autophagy in dorsal root ganglion macrophage. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1341. [PMID: 36660615 PMCID: PMC9843338 DOI: 10.21037/atm-22-5920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/16/2022] [Indexed: 12/28/2022]
Abstract
Background Dorsal root ganglia (DRG) plays an important role in mediating the peripheral sensation transduction through the primary afferent neurons in pain research. Neuropathic pain (NP) is a syndrome of hyperalgesia, spontaneous pain and allodynia caused by central or peripheral nerve injury. Recent trends of study are turning towards the development of therapies for the management of NP. Activation of autophagy in glial cells in the spinal cord has been reported to be associated with attenuation of NP, but the autophagic process in DRG is rarely studied. Methods The analgesic effect of electroacupuncture (EA) was evaluated in NP-induced rats developed using spared nerve injury (SNI). Acupuncture or EA was performed after 7 days of SNI at Zusanli (ST36) and Huantiao (GB30) acupoints. Then, the activation status of autophagy process in DRGs of rats treated with SNI and EA were investigated, and the possible mechanism of the analgesic effect of EA were explored. Results Application of EA has been found to reduce mechanical hyperalgesia. Autophagy indicator p62 was colocalized with the marker proteins for macrophages (CD11b), but not with NeuN (marker protein for neurons) or GFAP (marker protein for satellite glial cells), as shown by immunofluorescence. Western blots results indicate that the expression levels of p62, Beclin-1 and LC3-II in the L4-L6 DRG of rats in the SNI group were increased, compared with that in the control group. EA treatment resulted in decreased expression of p62 and increased expression of Beclin-1 and LC3-II/LC3-I. Furthermore, we explored the causal relationship between EA-induced suppression of NP and increased levels of autophagy in DRG using electron microscopy and the AMPK (AMP-activated protein kinase) inhibitor compound C. Conclusions SNI achieved a significant upregulation of autophagy levels in DRG macrophages. Furthermore, EA attenuated NP, which may contribute to the promotion of AMPK/mTOR (mammalian target of rapamycin)-mediated autophagy in DRG macrophages. Therefore, this strategy provides a new target for therapeutic intervention of NP.
Collapse
Affiliation(s)
- Qian Xu
- Department of Anesthesiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Cong Niu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, The Second Hospital of Chinese Medicine in Jiangsu Province, Nanjing, China
| | - Jiajing Li
- Department of Anesthesiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Cheng Hu
- Department of Pain Management, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Menglin He
- The First Clinical Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xizi Qiu
- The First Clinical Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qiang Yao
- The First Clinical Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weiqian Tian
- Department of Anesthesiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Minhao Zhang
- Department of Anesthesiology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China;,Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
25
|
Sex Differences in Neuropathy: The Paradigmatic Case of MetFormin. Int J Mol Sci 2022; 23:ijms232314503. [PMID: 36498830 PMCID: PMC9738696 DOI: 10.3390/ijms232314503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
As a widely prescribed anti-diabetic drug, metformin has been receiving novel attention for its analgesic potential. In the study of the complex etiology of neuropathic pain (NeP), male and female individuals exhibit quite different responses characterized by higher pain sensitivity and greater NeP incidence in women. This "gender gap" in our knowledge of sex differences in pain processing strongly limits the sex-oriented treatment of patients suffering from NeP. Besides, the current investigation of the analgesic potential of metformin has not addressed the "gender gap" problem. Hence, this study focuses on metformin and sex-dependent analgesia in a murine model of NeP induced by chronic constriction injury of the sciatic nerve. We investigated sexual dimorphism in signaling pathways involved by 7 days of metformin administration, such as changes in AMP-activated protein kinase and the positive regulation of autophagy machinery, discovering that metformin affected in a sexually dimorphic manner the immunological and inflammatory response to nerve lesion. These effects were complemented by morphological and adaptive changes occurring after peripheral nerve injury. Altogether these data can contribute to explaining a number of potential mechanisms responsible for the complete recovery from NeP found in male mice, as opposed to the failure of long-lasting recovery in female animals.
Collapse
|
26
|
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: 1.7] [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
|
27
|
Silva Santos Ribeiro P, Willemen HLDM, Eijkelkamp N. Mitochondria and sensory processing in inflammatory and neuropathic pain. FRONTIERS IN PAIN RESEARCH 2022; 3:1013577. [PMID: 36324872 PMCID: PMC9619239 DOI: 10.3389/fpain.2022.1013577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/26/2022] [Indexed: 01/24/2023] Open
Abstract
Rheumatic diseases, such as osteoarthritis and rheumatoid arthritis, affect over 750 million people worldwide and contribute to approximately 40% of chronic pain cases. Inflammation and tissue damage contribute to pain in rheumatic diseases, but pain often persists even when inflammation/damage is resolved. Mechanisms that cause this persistent pain are still unclear. Mitochondria are essential for a myriad of cellular processes and regulate neuronal functions. Mitochondrial dysfunction has been implicated in multiple neurological disorders, but its role in sensory processing and pain in rheumatic diseases is relatively unexplored. This review provides a comprehensive understanding of how mitochondrial dysfunction connects inflammation and damage-associated pathways to neuronal sensitization and persistent pain. To provide an overall framework on how mitochondria control pain, we explored recent evidence in inflammatory and neuropathic pain conditions. Mitochondria have intrinsic quality control mechanisms to prevent functional deficits and cellular damage. We will discuss the link between neuronal activity, mitochondrial dysfunction and chronic pain. Lastly, pharmacological strategies aimed at reestablishing mitochondrial functions or boosting mitochondrial dynamics as therapeutic interventions for chronic pain are discussed. The evidence presented in this review shows that mitochondria dysfunction may play a role in rheumatic pain. The dysfunction is not restricted to neuronal cells in the peripheral and central nervous system, but also includes blood cells and cells at the joint level that may affect pain pathways indirectly. Pre-clinical and clinical data suggest that modulation of mitochondrial functions can be used to attenuate or eliminate pain, which could be beneficial for multiple rheumatic diseases.
Collapse
Affiliation(s)
| | | | - Niels Eijkelkamp
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
28
|
Ma J, Liu J, Chen Y, Yu H, Xiang L. Metformin Promotes Axonal Regeneration and Functional Recovery in Diabetic Rat Model of Sciatic Nerve Transection Injury. NEUROSCI 2022; 3:366-375. [PMID: 39483432 PMCID: PMC11523732 DOI: 10.3390/neurosci3030026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/16/2022] [Indexed: 11/03/2024] Open
Abstract
In our previous study, metformin was able to promote nerve regeneration after sciatic nerve crushing in rats under diabetic conditions. However, a crush injury also has a strong ability to spontaneously recover. Therefore, in our present study, a model of transection injury of the sciatic nerve in diabetic rats was utilized to detect whether metformin could still promote nerve regeneration. Diabetes was induced via an injection of 50 mg/kg of streptozotocin in rats. After transection injury of the sciatic nerve, the rats were randomly divided into a high-dose metformin group (500 mg/kg/d), mid-dose metformin group (200 mg/kg/d), low-dose metformin group (30 mg/kg/d) and control group (normal saline). The metformin or normal saline was intraperitoneally injected for 4 weeks. Then, behavioral, electrophysiological and morphometric analyses were performed. The results showed that metformin could significantly promote functional restoration and axonal regeneration of the sciatic nerve after transection injury under diabetic conditions. Furthermore, high doses and middle doses of metformin presented more of this ability than a low dose of metformin. In conclusion, metformin is able to accelerate sciatic nerve repair after transection injury under diabetic conditions, showing the therapeutic potential of metformin in the management of nerve injuries during diabetes mellitus.
Collapse
Affiliation(s)
- Junxiong Ma
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110000, China; (J.M.); (J.L.); (Y.C.)
| | - Jun Liu
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110000, China; (J.M.); (J.L.); (Y.C.)
| | - Yu Chen
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110000, China; (J.M.); (J.L.); (Y.C.)
| | - Hailong Yu
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110000, China; (J.M.); (J.L.); (Y.C.)
| | - Liangbi Xiang
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110000, China; (J.M.); (J.L.); (Y.C.)
| |
Collapse
|
29
|
Haddad M, Eid S, Harb F, Massry MEL, Azar S, Sauleau EA, Eid AA. Activation of 20-HETE Synthase Triggers Oxidative Injury and Peripheral Nerve Damage in Type 2 Diabetic Mice. THE JOURNAL OF PAIN 2022; 23:1371-1388. [PMID: 35339661 DOI: 10.1016/j.jpain.2022.02.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 01/26/2022] [Accepted: 02/24/2022] [Indexed: 12/13/2022]
Abstract
Diabetic Peripheral Neuropathy (DPN), highly prevalent among patients with diabetes, is characterized by peripheral nerve dysfunction. Reactive Oxygen Species (ROS) overproduction has been suggested to orchestrate diabetic complications including DPN. Untargeted antioxidant therapy has exhibited limited efficacy, highlighting a critical need to explore ROS sources altered in a cell-specific manner in DPN. Cytochromes P450 (CYP) enzymes are prominent sources of ROS. Particularly, the 20-HETE synthase, CYP4A, is reported to mediate diabetes-induced renal, retinal, and cardiovascular injuries. This work investigates the role of CYP4A/20-HETE in DPN and their mechanisms of action. Non-obese type 2 Diabetic mice (MKR) were used and treated with a CYP4A-inhibitor (HET0016) or AMPK-activator (Metformin). Peripheral nerves of MKR mice reflect increased CYP4A and 20-HETE levels, concurrent with altered myelin proteins and sensorimotor deficits. This was associated with increased ROS production and altered Beclin-1 and LC3 protein levels, indicative of disrupted autophagic responses in tandem with AMPK inactivation. AMPK activation via Metformin restored nerve integrity, reduced ROS production, and regulated autophagy. Interestingly, similar outcomes were revealed upon HET0016 treatment whereby ROS production, autophagic responses, and AMPK signaling were normalized in diabetic mice. Altogether, the results highlight hyperglycemia-mediated oxidative injury in DPN through a novel CYP4A/20-HETE/AMPK pathological axis. PERSPECTIVE: To our knowledge, this is the first study to highlight the role of CYPs/20-HETE-induced oxidative injury in the pathogenesis of diabetic peripheral neuropathy. Targeting the identified pathological axis CYP4A/20-HETE/AMPK may be of clinical potential in predicting and alleviating peripheral nerve injury in patients with Type 2 Diabetes Mellitus.
Collapse
Affiliation(s)
- Mary Haddad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon; Department of Biostatistics, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7357 ICube, University of Strasbourg, Strasbourg, France
| | - Stéphanie Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Frederic Harb
- Department of Life and Earth Sciences, Faculty of Sciences, Lebanese University, Fanar, Lebanon
| | - Mohamed E L Massry
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Sami Azar
- Department of Internal Medicine, Division of Diabetes and Endocrinology, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon; AUB Diabetes, American University of Beirut, Beirut, Lebanon
| | - Erik-Andre Sauleau
- Department of Biostatistics, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7357 ICube, University of Strasbourg, Strasbourg, France
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon; AUB Diabetes, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
30
|
Song FH, Liu DQ, Zhou YQ, Mei W. SIRT1: A promising therapeutic target for chronic pain. CNS Neurosci Ther 2022; 28:818-828. [PMID: 35396903 PMCID: PMC9062570 DOI: 10.1111/cns.13838] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/12/2022] [Accepted: 03/30/2022] [Indexed: 12/14/2022] Open
Abstract
Chronic pain remains an unresolved problem. Current treatments have limited efficacy. Thus, novel therapeutic targets are urgently required for the development of more effective analgesics. An increasing number of studies have proved that sirtuin 1 (SIRT1) agonists can relieve chronic pain. In this review, we summarize recent progress in understanding the roles and mechanisms of SIRT1 in mediating chronic pain associated with peripheral nerve injury, chemotherapy‐induced peripheral neuropathy, spinal cord injury, bone cancer, and complete Freund's adjuvant injection. Emerging studies have indicated that SIRT1 activation may exert positive effects on chronic pain relief by regulating inflammation, oxidative stress, and mitochondrial dysfunction. Therefore, SIRT1 agonists may serve as potential therapeutic drugs for chronic pain.
Collapse
Affiliation(s)
- Fan-He Song
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dai-Qiang Liu
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Qun Zhou
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Mei
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Vitamin B12 Deficiency in Patients with Diabetes on Metformin: Arab Countries. Nutrients 2022; 14:nu14102046. [PMID: 35631186 PMCID: PMC9144422 DOI: 10.3390/nu14102046] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 11/30/2022] Open
Abstract
Background: Diabetes is a global pandemic, especially in Arab countries. Aim: The goal of this study was to review the published studies that were conducted to determine the relationship between metformin treatment for type 2 diabetes mellitus (T2DM) and vitamin B12 deficiency and to identify possible complications in this relationship. Methods: I searched for all relevant studies published in English before 2020 on the PubMed and Web of Knowledge databases using the following terms: “metformin”, “vitamin B12”, “neuropathy”, “diabetes mellitus”, and Middle Eastern countries. Results: Eleven studies were included in this review which indicated an association between taking metformin and B12 deficiency in patients with T2DM in Arab countries. This B12 deficiency was found to be negatively associated with the dose and duration of metformin therapy. The physician’s knowledge of current ADA recommendations regarding supplementation with and screening of the B12 level for T2DM patients on metformin was also found to have an effect. Conclusion: Metformin therapy is associated with B12 deficiency among people with T2DM in Arabic countries. Thus, diabetes must be managed in compliance with current guidelines and recommendations, and B12 levels must be routinely monitored, particularly in those who have been long-term takers of metformin, to ensure the suitable management of diabetes and its complications.
Collapse
|
32
|
Garner KM, Burton MD. The sex-specific role of sensory neuron LKB1 on metabolic stress-induced mechanical hypersensitivity and mitochondrial respiration. Am J Physiol Regul Integr Comp Physiol 2022; 323:R227-R243. [PMID: 35503781 PMCID: PMC9306793 DOI: 10.1152/ajpregu.00279.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pain disorders induce metabolic stress in peripheral sensory neurons by reducing mitochondrial output, shifting cellular metabolism, and altering energy use. These processes implicate neuronal metabolism as an avenue for creating novel therapeutics. Liver kinase B1 (LKB1) mediates the cellular response to metabolic stress by inducing the AMPK pathway. The LKB1-AMPK pathway increases energy producing processes, including mitochondrial output. These processes inhibit pain by directly or indirectly restoring energetic balance within a cell. Although the LKB1-AMPK pathway has been linked to pain relief, it is not yet known which cell is responsible for this property, as well any direct ties to cellular metabolism. To elucidate this, we developed a genetic mouse model where LKB1 is selectively removed from Nav1.8-pain sensory neurons and metabolically stressed them by fasting for 24 hours. We found females, but not males, had neuron-specific, LKB1-dependent restoration of metabolic stress-induced mitochondrial metabolism. This was reflected in mechanical hypersensitivity, where the absence of LKB1 led to hypersensitivity in female, but not male, animals. This discrepancy suggests a sex- and cell-specific contribution to LKB1-depdendent fasting-induced mechanical hypersensitivity. While our data represent a potential role for LKB1 in anti-pain pathways in a metabolic-specific manner, more must be done to investigate these sex differences.
Collapse
Affiliation(s)
- Katherine Marie Garner
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States
| | - Michael D Burton
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States
| |
Collapse
|
33
|
The Antidiabetic Drug Metformin Regulates Voltage-Gated Sodium Channel Na V1.7 via the Ubiquitin-Ligase NEDD4-2. eNeuro 2022; 9:ENEURO.0409-21.2022. [PMID: 35131865 PMCID: PMC8906783 DOI: 10.1523/eneuro.0409-21.2022] [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: 07/20/2021] [Revised: 12/10/2021] [Accepted: 01/01/2022] [Indexed: 12/14/2022] Open
Abstract
The antidiabetic drug metformin has been shown to reduce pain hypersensitivity in preclinical models of chronic pain and in neuropathic pain in humans. Multiple intracellular pathways have been described as metformin targets. Among them, metformin is an activator of the adenosine 5′-monophosphate protein kinase that can in turn modulate the activity of the E3 ubiquitin ligase NEDD4-2 and thus post-translational expression of voltage-gated sodium channels (NaVs). In this study, we found that the bulk of the effect of metformin on Na1.7 is dependent on NEDD4-2. In HEK cells, the expression of NaV1.7 at the membrane fraction, obtained by a biotinylation approach, is only reduced by metformin when cotransfected with NEDD4-2. Similarly, in voltage-clamp recordings, metformin significantly reduced NaV1.7 current density when cotransfected with NEDD4-2. In mouse dorsal root ganglion (DRG) neurons, without changing the biophysical properties of NaV1.7, metformin significantly decreased NaV1.7 current densities, but not in Nedd4L knock-out mice (SNS-Nedd4L−/−). In addition, metformin induced a significant reduction in NEDD4-2 phosphorylation at the serine-328 residue in DRG neurons, an inhibitory phosphorylation site of NEDD4-2. In current-clamp recordings, metformin reduced the number of action potentials elicited by DRG neurons from Nedd4Lfl/fl, with a partial decrease also present in SNS-Nedd4L−/− mice, suggesting that metformin can also change neuronal excitability in an NEDD4-2-independent manner. We suggest that NEDD4-2 is a critical player for the effect of metformin on the excitability of nociceptive neurons; this action may contribute to the relief of neuropathic pain.
Collapse
|
34
|
Cao XJ, Wu R, Qian HY, Chen X, Zhu HY, Xu GY, Sun YZ, Zhang PA. Metformin attenuates diabetic neuropathic pain via AMPK/NF-κB signaling pathway in dorsal root ganglion of diabetic rats. Brain Res 2021; 1772:147663. [PMID: 34555415 DOI: 10.1016/j.brainres.2021.147663] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/26/2021] [Accepted: 09/15/2021] [Indexed: 10/20/2022]
Abstract
Neuropathic pain is a common complication of diabetes mellitus with poorly relieved by conventional analgesics. Metformin, a first-line drug for type 2 diabetes, reduces blood glucose by activating adenosine monophosphate protein kinase (AMPK) signalling system. However, the effect of Metformin on diabetic neuropathic pain is still unknown. In the present study, we showed that Metformin was capable of attenuating diabetes induced mechanical allodynia, and the analgesia effect could be blocked by Compound C (an AMPK inhibitor). Importantly, Metformin enhanced the phosphorylation level of AMPK in L4-6 DRGs of diabetic rats but not affect the expression of total AMPK. Intrathecal injection of AICAR (an AMPK agonist) could activate AMPK and alleviate the mechanical allodynia of diabetic rats. Additionally, phosphorylated AMPK and NF-κB was co-localized in small and medium neurons of L4-6 DRGs. Interestingly, the regulation of NF-κB in diabetic rats was obviously reduced when AMPK was activated by AICAR. Notably, Metformin could decrease NF-κB expression in L4-6 DRGs of diabetic rats, but the decrease was blocked by Compound C. In conclusion, Metformin alleviates diabetic mechanical allodynia via activation of AMPK signaling pathway in L4-6 DRGs of diabetic rats, which might be mediated by the downregulation of NF-κB, and this providing certain basis for Metformin to become a potential drug in the clinical treatment of diabetic neuropathic pain.
Collapse
Affiliation(s)
- Xiao-Jun Cao
- Department of Endocrine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215123, PR China
| | - Rui Wu
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215123, PR China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, PR China
| | - He-Ya Qian
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215123, PR China
| | - Xiang Chen
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215123, PR China
| | - Hong-Yan Zhu
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215123, PR China
| | - Guang-Yin Xu
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215123, PR China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, PR China
| | - Ye-Zi Sun
- Department of Endocrine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215123, PR China.
| | - Ping-An Zhang
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215123, PR China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
35
|
Gerum M, Simonin F. Behavioral characterization, potential clinical relevance and mechanisms of latent pain sensitization. Pharmacol Ther 2021; 233:108032. [PMID: 34763010 DOI: 10.1016/j.pharmthera.2021.108032] [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: 04/16/2021] [Revised: 10/22/2021] [Accepted: 11/03/2021] [Indexed: 10/19/2022]
Abstract
Chronic pain is a debilitating disorder that can occur as painful episodes that alternates with bouts of remission and occurs despite healing of the primary insult. Those episodes are often triggered by stressful events. In the last decades, a similar situation has been evidenced in a wide variety of rodent models (including inflammatory pain, neuropathy and opioid-induced hyperalgesia) where animals develop a chronic latent hyperalgesia that silently persists after behavioral signs of pain resolution. This state, referred as latent pain sensitization, is due to the compensatory activation of antinociceptive systems, such as the opioid system or NPY and its receptors. A transitory phase of hyperalgesia can then be reinstated by pharmacological or genetic blockade of these antinociceptive systems or by submitting animals to acute stress. Those observations reveal that there is a constant endogenous analgesia responsible for chronic pain inhibition that might paradoxically contribute to maintain this maladaptive state and could then participate to the transition from acute to chronic pain. Thus, demonstration of the existence of this phenomenon in humans and a better understanding of the mechanisms by which latent pain sensitization develops and maintains over long periods of time will be of particular interest to help identifying new therapeutic strategies and targets for chronic pain treatment. The present review aims to recapitulate behavioral expression, potential clinical relevance, cellular mechanisms and intracellular signaling pathways involved so far in latent pain sensitization.
Collapse
Affiliation(s)
- Manon Gerum
- Biotechnologie et Signalisation Cellulaire, UMR7242 CNRS, Université de Strasbourg, Institut du Médicament de Strasbourg, Illkirch-Graffenstaden, France
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR7242 CNRS, Université de Strasbourg, Institut du Médicament de Strasbourg, Illkirch-Graffenstaden, France.
| |
Collapse
|
36
|
Bae EH, Greenwald MK, Schwartz AG. Chemotherapy-Induced Peripheral Neuropathy: Mechanisms and Therapeutic Avenues. Neurotherapeutics 2021; 18:2384-2396. [PMID: 34676514 PMCID: PMC8804039 DOI: 10.1007/s13311-021-01142-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2021] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a serious and often persistent adverse consequence of certain chemotherapeutic agents. It is a major dose-limiting factor of many first-line chemotherapies, affecting 20-50% of patients at standard doses and nearly all patients at high doses. As cancer survivorship continues to increase with improvements in early diagnosis and treatment, more patients will experience CIPN despite completing cancer treatment, which interferes with recovery, leading to chronic pain and worsening quality of life. The National Cancer Institute has identified CIPN as a priority in translational research. To date, there are no FDA-approved drugs for preventing or treating CIPN, with emerging debate on mechanisms and promising new targets. This review highlights current literature and suggests novel approaches to CIPN based on proposed mechanisms of action that aim either to confer neuroprotection against chemotherapy-induced neurotoxicity or reverse the downstream effects of painful neuropathy.
Collapse
Affiliation(s)
- Esther H Bae
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, 3901 Chrysler Service Drive, Suite 2A, Detroit, MI, 48201, USA
| | - Mark K Greenwald
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, 3901 Chrysler Service Drive, Suite 2A, Detroit, MI, 48201, USA.
- Karmanos Cancer Institute, Detroit, MI, USA.
| | - Ann G Schwartz
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, 3901 Chrysler Service Drive, Suite 2A, Detroit, MI, 48201, USA
- Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
37
|
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: 2.0] [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
|
38
|
Wang SM, Goguadze N, Kimura Y, Yasui Y, Pan B, Wang TY, Nakamura Y, Lin YT, Hogan QH, Wilson KL, Su TP, Wu HE. Genomic Action of Sigma-1 Receptor Chaperone Relates to Neuropathic Pain. Mol Neurobiol 2021; 58:2523-2541. [PMID: 33459966 PMCID: PMC8128747 DOI: 10.1007/s12035-020-02276-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/28/2020] [Indexed: 11/16/2022]
Abstract
Sigma-1 receptors (Sig-1Rs) are endoplasmic reticulum (ER) chaperones implicated in neuropathic pain. Here we examine if the Sig-1R may relate to neuropathic pain at the level of dorsal root ganglia (DRG). We focus on the neuronal excitability of DRG in a "spare nerve injury" (SNI) model of neuropathic pain in rats and find that Sig-1Rs likely contribute to the genesis of DRG neuronal excitability by decreasing the protein level of voltage-gated Cav2.2 as a translational inhibitor of mRNA. Specifically, during SNI, Sig-1Rs translocate from ER to the nuclear envelope via a trafficking protein Sec61β. At the nucleus, the Sig-1R interacts with cFos and binds to the promoter of 4E-BP1, leading to an upregulation of 4E-BP1 that binds and prevents eIF4E from initiating the mRNA translation for Cav2.2. Interestingly, in Sig-1R knockout HEK cells, Cav2.2 is upregulated. In accordance with those findings, we find that intra-DRG injection of Sig-1R agonist (+)pentazocine increases frequency of action potentials via regulation of voltage-gated Ca2+ channels. Conversely, intra-DRG injection of Sig-1R antagonist BD1047 attenuates neuropathic pain. Hence, we discover that the Sig-1R chaperone causes neuropathic pain indirectly as a translational inhibitor.
Collapse
MESH Headings
- Animals
- Calcium Channels, N-Type/genetics
- Calcium Channels, N-Type/metabolism
- Endoplasmic Reticulum/metabolism
- Eukaryotic Initiation Factor-4E/metabolism
- Ganglia, Spinal/metabolism
- Gene Expression Regulation
- Genome
- HEK293 Cells
- Humans
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Nerve Tissue/injuries
- Nerve Tissue/pathology
- Neuralgia/genetics
- Nuclear Envelope/metabolism
- Promoter Regions, Genetic/genetics
- Protein Biosynthesis
- Proto-Oncogene Proteins c-fos/metabolism
- RNA Caps/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Receptors, sigma/agonists
- Receptors, sigma/genetics
- Receptors, sigma/metabolism
- SEC Translocation Channels/metabolism
- Transcription, Genetic
- Sigma-1 Receptor
- Rats
Collapse
Affiliation(s)
- Shao-Ming Wang
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Nino Goguadze
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Yuriko Kimura
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Yuko Yasui
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Bin Pan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Tzu-Yun Wang
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
- Department of Psychiatry, College of Medicine, National Cheng Kung University, Tainan City, 70101, Taiwan
| | - Yoki Nakamura
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
- Department of Pharmacology, Graduate School of Biomedical & Health Science, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Yu-Ting Lin
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Katherine L Wilson
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA.
| | - Hsiang-En Wu
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| |
Collapse
|
39
|
Inyang KE, Folger JK, Laumet G. Can FDA-Approved Immunomodulatory Drugs be Repurposed/Repositioned to Alleviate Chronic Pain? J Neuroimmune Pharmacol 2021; 16:531-547. [PMID: 34041656 DOI: 10.1007/s11481-021-10000-z] [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: 03/22/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022]
Abstract
Pain is among the most widespread chronic health condition confronting society today and our inability to manage chronic pain contributes to the opioid abuse epidemic in America. The immune system is known to contribute to acute and chronic pain, but only limited therapeutic treatments such as non-steroid anti-inflammatory drugs have resulted from this knowledge. The last decade has shed light on neuro-immune interactions mediating the development, maintenance, and resolution of chronic pain. Here, we do not aim to perform a comprehensive review of all immune mechanisms involved in chronic pain, but to briefly review the contribution of the main cytokines and immune cells (macrophages, microglia, mast cells and T cells) to chronic pain. Given the urgent need to address the Pain crisis, we provocatively propose to repurpose/reposition FDA-approved immunomodulatory drugs for their potential to alleviate chronic pain. Repositioning or repurposing offers an attractive way to accelerate the arrival of new analgesics.
Collapse
Affiliation(s)
| | - Joseph K Folger
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Geoffroy Laumet
- Department of Physiology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
40
|
Carvalho-E-Silva AP, Harmer AR, Ferreira ML, Ferreira PH. The effect of the anti-diabetic drug metformin on musculoskeletal pain: A cross-sectional study with 21,889 individuals from the UK biobank. Eur J Pain 2021; 25:1264-1273. [PMID: 33561890 DOI: 10.1002/ejp.1747] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Although there is growing evidence of metformin's pleiotropic effects, including possible effects on pain, there is a lack of studies investigating the association of metformin with the prevalence of musculoskeletal pain among a large cohort with type 2 diabetes cohort. METHODS Cross-sectional analyses were conducted with UK Biobank data from 21,889 participants with type 2 diabetes. Type 2 diabetes, metformin use and musculoskeletal (back, knee, hip and neck/shoulder) pain were self-reported. Participants reported musculoskeletal pain that had interfered with their usual activities in the last month (recent pain), and for more than 3 months (chronic pain). We performed logistic regression analyses for recent and chronic pain for each site and for multisite pain among participants with diabetes who did or did not take metformin. RESULTS Participants using metformin had lower odds of musculoskeletal pain for back [recent OR 0.91, 95%CI 0.85 to 0.97; chronic OR 0.87, 95%CI 0.81 to 0.93], knee [recent OR 0.91, 95%CI 0.85 to 0.97; chronic OR 0.87, 95%CI 0.81 to 0.94] and neck/shoulder regions [chronic OR 0.92, 95%CI 0.85 to 0.99] but not hip pain. Participants using metformin also had lower odds of reporting chronic multisite musculoskeletal pain. The associations were generally stronger among women. CONCLUSIONS People with diabetes taking metformin were less likely to report back, knee, neck/shoulder and multisite musculoskeletal pain than those not taking metformin. Therefore, when treating these patients, clinicians should be aware that metformin may contribute to fewer reports of musculoskeletal pain. These effects should be investigated in future studies. SIGNIFICANCE People with type 2 diabetes taking metformin are less likely to present with musculoskeletal pain than those not taking metformin. Metformin may have a protective effect for musculoskeletal pain, which appears to be stronger among women than men.
Collapse
Affiliation(s)
- Ana Paula Carvalho-E-Silva
- Musculoskeletal Health Research Group, Faculty of Medicine and Health, School of Health Sciences, The University of Sydney, Sydney, NSW, Australia.,Institute of Bone and Joint Research, The Kolling Institute, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Alison R Harmer
- Musculoskeletal Health Research Group, Faculty of Medicine and Health, School of Health Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Manuela L Ferreira
- Institute of Bone and Joint Research, The Kolling Institute, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Paulo H Ferreira
- Musculoskeletal Health Research Group, Faculty of Medicine and Health, School of Health Sciences, The University of Sydney, Sydney, NSW, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
41
|
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: 7.3] [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
|
42
|
Demaré S, Kothari A, Calcutt NA, Fernyhough P. Metformin as a potential therapeutic for neurological disease: mobilizing AMPK to repair the nervous system. Expert Rev Neurother 2020; 21:45-63. [PMID: 33161784 PMCID: PMC9482886 DOI: 10.1080/14737175.2021.1847645] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Metformin is currently first line therapy for type 2 diabetes (T2D). The mechanism of action of metformin involves activation of AMP-activated protein kinase (AMPK) to enhance mitochondrial function (for example, biogenesis, refurbishment and dynamics) and autophagy. Many neurodegenerative diseases of the central and peripheral nervous systems arise from metabolic failure and toxic protein aggregation where activated AMPK could prove protective. Areas covered: The authors review literature on metformin treatment in Parkinson’s disease, Huntington’s disease and other neurological diseases of the CNS along with neuroprotective effects of AMPK activation and suppression of the mammalian target of rapamycin (mTOR) pathway on peripheral neuropathy and neuropathic pain. The authors compare the efficacy of metformin with the actions of resveratrol. Expert opinion: Metformin, through activation of AMPK and autophagy, can enhance neuronal bioenergetics, promote nerve repair and reduce toxic protein aggregates in neurological diseases. A long history of safe use in humans should encourage development of metformin and other AMPK activators in preclinical and clinical research. Future studies in animal models of neurological disease should strive to further dissect in a mechanistic manner the pathways downstream from metformin-dependent AMPK activation, and to further investigate mTOR dependent and independent signaling pathways driving neuroprotection.
Collapse
Affiliation(s)
- Sarah Demaré
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre , Winnipeg, MB, Canada.,Department of Pharmacology and Therapeutics, University of Manitoba , Winnipeg, MB, Canada
| | - Asha Kothari
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre , Winnipeg, MB, Canada.,Department of Pharmacology and Therapeutics, University of Manitoba , Winnipeg, MB, Canada
| | - Nigel A Calcutt
- Department of Pathology, University of California San Diego , La Jolla, CA, USA
| | - Paul Fernyhough
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre , Winnipeg, MB, Canada.,Department of Pharmacology and Therapeutics, University of Manitoba , Winnipeg, MB, Canada
| |
Collapse
|
43
|
Kim SH, Park TS, Jin HY. Metformin Preserves Peripheral Nerve Damage with Comparable Effects to Alpha Lipoic Acid in Streptozotocin/High-Fat Diet Induced Diabetic Rats. Diabetes Metab J 2020; 44:842-853. [PMID: 32602278 PMCID: PMC7801759 DOI: 10.4093/dmj.2019.0190] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 11/28/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Metformin is widely marketed medication for the treatment of diabetes, but its pharmacological effect on diabetic peripheral neuropathy remains unclear. In this study, the effect of metformin on peripheral nerves in diabetic rats was investigated using diverse neuronal parameters of nerve fibers. METHODS Rats were assigned to one of four groups (n=7 to 10 per group): normal, diabetes mellitus (DM), DM+metformin (100 mg/kg), and DM+alpha lipoic acid (ALA, 100 mg/kg). DM was induced by streptozotocin/high-fat diet (STZ/HFD). After 12 weeks, the sensory thresholds to mechanical and heat stimuli were assessed. Repeated sensory tests, immunofluorescence microscopic comparison of peripheral nerves, and biochemical blood analysis were performed after 24 weeks. RESULTS Both DM+metformin and DM+ALA groups showed similar trends to diverse sensory tests at 24 weeks compared to DM group although the degree of change were different according to the stimulated senses. There was no significant difference in the comparison of the intraepidermal nerve fiber density (IENFD) of peripheral nerves between the DM+metformin and DM+ALA groups (11.83±0.07 fibers/mm vs. 12.37±1.82 fibers/mm, respectively). Both groups showed preserved IENFD significantly compared with DM group (8.46±1.98 fibers/mm, P<0.05). Sciatic nerve morphology of the experimental animals showed a similar trend to the IENFD, with respect to axonal diameter, myelin sheath thickness, and myelinated fiber diameter. CONCLUSION Metformin has beneficial pharmacological effects on the preservation of peripheral nerves in diabetic rats and its effects are comparable to those of ALA.
Collapse
Affiliation(s)
- Sun Hee Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Tae Sun Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Heung Yong Jin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| |
Collapse
|
44
|
Wang S, Qi S, Kogure Y, Kanda H, Tian L, Yamamoto S, Noguchi K, Dai Y. The ubiquitin E3 ligase Nedd4-2 relieves mechanical allodynia through the ubiquitination of TRPA1 channel in db/db mice. Eur J Neurosci 2020; 53:1691-1704. [PMID: 33236491 DOI: 10.1111/ejn.15062] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 11/09/2020] [Accepted: 11/17/2020] [Indexed: 01/25/2023]
Abstract
Neural precursor cell-expressed developmentally downregulated protein 4-2 (Nedd4-2) is a member of the E3 ubiquitin ligase family that is highly expressed in sensory neurons and involved in pain modulation via downregulation of ion channels in excitable membranes. Ubiquitination involving Nedd4-2 is regulated by adenosine monophosphate-activated protein kinase (AMPK), which is impaired in the dorsal root ganglion (DRG) neurons of db/db mice. AMPK negatively regulates the expression of transient receptor potential ankyrin 1 (TRPA1), a recognised pain sensor expressed on the membrane of DRG neurons, consequently relieving mechanical allodynia in db/db mice. Herein, we studied the involvement of Nedd4-2 in painful diabetic neuropathy and observed that Nedd4-2 negatively regulated diabetic mechanical allodynia. Nedd4-2 was co-expressed with TRPA1 in mouse DRG neurons. Nedd4-2 was involved in TRPA1 ubiquitination, this ubiquitination, as well as Nedd4-2-TRPA1 interaction, was decreased in db/db mice. Moreover, Nedd4-2 levels were decreased in db/db mice, while an abnormal intracellular distribution was observed in short-term high glucose-cultured DRG neurons. AMPK activators not only restored Nedd4-2 distribution but also increased Nedd4-2 expression. These findings demonstrate that Nedd4-2 is a potent regulator of TRPA1 and that the abnormal expression of Nedd4-2 in DRG neurons contributes to diabetic neuropathic pain.
Collapse
Affiliation(s)
- Shenglan Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China.,Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Hyogo, Japan
| | - Simin Qi
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yoko Kogure
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Hyogo, Japan
| | - Hirosato Kanda
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Hyogo, Japan.,Traditional Medicine Research Center, Chinese Medicine Confucius Institute at Hyogo College of Medicine, Kobe, Hyogo, Japan.,Department of Anatomy and Neuroscience, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Lin Tian
- Department of Gerontology, Guang'anmen Hospital, China Academy of Chinese Medical Science, Beijing, China
| | - Satoshi Yamamoto
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Hyogo, Japan
| | - Koichi Noguchi
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Yi Dai
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Hyogo, Japan.,Traditional Medicine Research Center, Chinese Medicine Confucius Institute at Hyogo College of Medicine, Kobe, Hyogo, Japan.,Department of Anatomy and Neuroscience, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| |
Collapse
|
45
|
Baeza-Flores GDC, Guzmán-Priego CG, Parra-Flores LI, Murbartián J, Torres-López JE, Granados-Soto V. Metformin: A Prospective Alternative for the Treatment of Chronic Pain. Front Pharmacol 2020; 11:558474. [PMID: 33178015 PMCID: PMC7538784 DOI: 10.3389/fphar.2020.558474] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/02/2020] [Indexed: 12/15/2022] Open
Abstract
Metformin (biguanide) is a drug widely used for the treatment of type 2 diabetes. This drug has been used for 60 years as a highly effective antihyperglycemic agent. The search for the mechanism of action of metformin has produced an enormous amount of research to explain its effects on gluconeogenesis, protein metabolism, fatty acid oxidation, oxidative stress, glucose uptake, autophagy and pain, among others. It was only up the end of the 1990s and beginning of this century that some of its mechanisms were revealed. Metformin induces its beneficial effects in diabetes through the activation of a master switch kinase named AMP-activated protein kinase (AMPK). Two upstream kinases account for the physiological activation of AMPK: liver kinase B1 and calcium/calmodulin-dependent protein kinase kinase 2. Once activated, AMPK inhibits the mechanistic target of rapamycin complex 1 (mTORC1), which in turn avoids the phosphorylation of p70 ribosomal protein S6 kinase 1 and phosphatidylinositol 3-kinase/protein kinase B signaling pathways and reduces cap-dependent translation initiation. Since metformin is a disease-modifying drug in type 2 diabetes, which reduces the mTORC1 signaling to induce its effects on neuronal plasticity, it was proposed that these mechanisms could also explain the antinociceptive effect of this drug in several models of chronic pain. These studies have highlighted the efficacy of this drug in chronic pain, such as that from neuropathy, insulin resistance, diabetic neuropathy, and fibromyalgia-type pain. Mounting evidence indicates that chronic pain may induce anxiety, depression and cognitive impairment in rodents and humans. Interestingly, metformin is able to reverse some of these consequences of pathological pain in rodents. The purpose of this review was to analyze the current evidence about the effects of metformin in chronic pain and three of its comorbidities (anxiety, depression and cognitive impairment).
Collapse
Affiliation(s)
- Guadalupe Del Carmen Baeza-Flores
- Laboratorio de Mecanismos de Dolor, División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Mexico
| | - Crystell Guadalupe Guzmán-Priego
- Laboratorio de Mecanismos de Dolor, División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Mexico
| | - Leonor Ivonne Parra-Flores
- Laboratorio de Mecanismos de Dolor, División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Mexico
| | - Janet Murbartián
- Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Jorge Elías Torres-López
- Laboratorio de Mecanismos de Dolor, División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Mexico.,Departamento de Anestesiología, Hospital Regional de Alta Especialidad "Dr. Juan Graham Casasús", Villahermosa, Mexico
| | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| |
Collapse
|
46
|
Guo L, Du J, Yuan DF, Zhang Y, Zhang S, Zhang HC, Mi JW, Ning YL, Chen MJ, Wen DL, Sun JH, Liu D, Zeng L, Zhang A, Jiang J, Huang H. Optimal H 2O 2 preconditioning to improve bone marrow mesenchymal stem cells' engraftment in wound healing. Stem Cell Res Ther 2020; 11:434. [PMID: 33032649 PMCID: PMC7545926 DOI: 10.1186/s13287-020-01910-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/27/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The transplantation of bone marrow mesenchymal stem cells (BMSCs) is a promising therapeutic strategy for wound healing. However, the poor migration capacity and low survival rate of transplanted BMSCs in wounds weaken their potential application. OBJECTIVE To identify the optimal protocol for BMSCs preconditioned with H2O2 and improve the therapeutic efficacy using H2O2-preconditioned BMSCs in wound healing. METHODS Mouse BMSCs were exposed to various concentrations of H2O2, and the key cellular functional properties were assessed to determine the optimal precondition with H2O2. The H2O2-preconditioned BMSCs were transplanted into mice with full-thickness excisional wounds to evaluate their healing capacity and tissue engraftment. RESULTS Treatment BMSCs with 50 μM H2O2 for 12 h could significantly enhance their proliferation, migration, and survival by maximizing the upregulation of cyclin D1, SDF-1, and its receptors CXCR4/7 expressions, and activating the PI3K/Akt/mTOR pathway, but inhibiting the expression of p16 and GSK-3β. Meanwhile, oxidative stress-induced BMSC apoptosis was also significantly attenuated by the same protocol pretreatment with a decreased ratio of Bax/Bcl-2 and cleaved caspase-9/3 expression. Moreover, after the identification of the optimal protocol of H2O2 precondition in vitro, the migration and tissue engraftment of transfused BMSCs with H2O2 preconditioning were dramatically increased into the wound site as compared to the un-preconditioned BMSCs. The increased microvessel density and the speedy closure of the wounds were observed after the transfusion of H2O2-preconditioned BMSCs. CONCLUSIONS The findings suggested that 50 μM H2O2 pretreated for 12 h is the optimal precondition for the transplantation of BMSCs, which gives a considerable insight that this protocol may be served as a promising candidate for improving the therapeutic potential of BMSCs for wound healing.
Collapse
Affiliation(s)
- Ling Guo
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Surgical Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Juan Du
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Surgical Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Dan-Feng Yuan
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Surgical Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Ya Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Surgical Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Shu Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Surgical Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Hua-Cai Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Surgical Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jun-Wei Mi
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Surgical Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Ya-Lei Ning
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Surgical Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Min-Jia Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Surgical Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Da-Lin Wen
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Surgical Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jian-Hui Sun
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Surgical Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Di Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Surgical Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Ling Zeng
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Surgical Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Anqiang Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Surgical Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jianxin Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Surgical Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Hong Huang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Surgical Research, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
47
|
Is metformin a geroprotector? A peek into the current clinical and experimental data. Mech Ageing Dev 2020; 191:111350. [DOI: 10.1016/j.mad.2020.111350] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/25/2020] [Accepted: 09/01/2020] [Indexed: 02/08/2023]
|
48
|
A disintegrin and metalloproteinase domain 17-epidermal growth factor receptor signaling contributes to oral cancer pain. Pain 2020; 161:2330-2343. [PMID: 32453136 PMCID: PMC9244849 DOI: 10.1097/j.pain.0000000000001926] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer cells secrete pronociceptive mediators that sensitize adjacent sensory neurons and cause pain. Identification and characterization of these mediators could pinpoint novel targets for cancer pain treatment. In this study, we identified candidate genes in cancer cell lines that encode for secreted or cell surface proteins that may drive nociception. To undertake this work, we used an acute cancer pain mouse model, transcriptomic analysis of publicly available human tumor-derived cell line data, and a literature review. Cancer cell line supernatants were assigned a phenotype based on evoked nociceptive behavior in an acute cancer pain mouse model. We compared gene expression data from nociceptive and nonnociceptive cell lines. Our analyses revealed differentially expressed genes and pathways; many of the identified genes were not previously associated with cancer pain signaling. Epidermal growth factor receptor (EGFR) and disintegrin metalloprotease domain 17 (ADAM17) were identified as potential targets among the differentially expressed genes. We found that the nociceptive cell lines contained significantly more ADAM17 protein in the cell culture supernatant compared to nonnociceptive cell lines. Cytoplasmic EGFR was present in almost all (>90%) tongue primary afferent neurons in mice. Monoclonal antibody against EGFR, cetuximab, inhibited cell line supernatant-induced nociceptive behavior in an acute oral cancer pain mouse model. We infer from these data that ADAM17-EGFR signaling is involved in cancer mediator-induced nociception. The differentially expressed genes and their secreted protein products may serve as candidate therapeutic targets for oral cancer pain and warrant further evaluation.
Collapse
|
49
|
Martinez N, Sánchez A, Diaz P, Broekhuizen R, Godoy J, Mondaca S, Catenaccio A, Macanas P, Nervi B, Calvo M, Court F. Metformin protects from oxaliplatin induced peripheral neuropathy in rats. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2020; 8:100048. [PMID: 32490289 PMCID: PMC7260677 DOI: 10.1016/j.ynpai.2020.100048] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/20/2020] [Accepted: 05/12/2020] [Indexed: 12/14/2022]
Abstract
Oxaliplatin is a commonly used drug to treat cancer, extending the rate of disease-free survival by 20% in colorectal cancer. However, oxaliplatin induces a disabling form of neuropathy resulting in more than 60% of patients having to reduce or discontinue oxaliplatin, negatively impacting their chance of survival. Oxaliplatin-induced neuropathies are accompanied by degeneration of sensory fibers in the epidermis and hyperexcitability of sensory neurons. These morphological and functional changes have been associated with sensory symptoms such as dysesthesia, paresthesia and mechanical and cold allodynia. Various strategies have been proposed to prevent or treat oxaliplatin-induced neuropathies without success. The anti-diabetic drug metformin has been recently shown to exert neuroprotection in other chemotherapy-induced neuropathies, so here we aimed to test if metformin can prevent the development of oxaliplatin-induced neuropathy in a rat model of this condition. Animals treated with oxaliplatin developed significant intraepidermal fiber degeneration, a mild gliosis in the spinal cord, and mechanical and cold hyperalgesia. The concomitant use of metformin prevented degeneration of intraepidermal fibers, gliosis, and the altered sensitivity. Our evidence further supports metformin as a new approach to prevent oxaliplatin-induced neuropathy with a potential important clinical impact.
Collapse
Affiliation(s)
- N.W. Martinez
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor de Chile, Santiago 8580745, Chile
- Department of Physiology, Faculty of Biology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A. Sánchez
- Department of Physiology, Faculty of Biology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - P. Diaz
- Department of Physiology, Faculty of Biology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - R. Broekhuizen
- Department of Hematology and Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - J. Godoy
- Department of Neurology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - S. Mondaca
- Department of Hematology and Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A. Catenaccio
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor de Chile, Santiago 8580745, Chile
| | - P. Macanas
- Department of Hematology and Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - B. Nervi
- Department of Hematology and Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - M. Calvo
- Department of Physiology, Faculty of Biology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - F.A. Court
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor de Chile, Santiago 8580745, Chile
- FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago 8580745, Chile
- Buck Institute for Research on Ageing, Novato, San Francisco, CA 94945, USA
| |
Collapse
|
50
|
He WY, Zhang B, Zhao WC, He J, Wang Y, Zhang L, Xiong QM, Wang HB. mTOR activation due to APPL1 deficiency exacerbates hyperalgesia via Rab5/Akt and AMPK signaling pathway in streptozocin-induced diabetic rats. Mol Pain 2020; 15:1744806919880643. [PMID: 31530215 PMCID: PMC6878613 DOI: 10.1177/1744806919880643] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Painful diabetic neuropathy is a common complication of diabetes mellitus with obscure underlying mechanisms. The adaptor protein APPL1 is critical in mediating the insulin sensitizing and insulin signaling. In neurons, APPL1 reportedly affects synaptic plasticity, while its role in the pathogenesis of painful diabetic neuropathy is masked. Our Western blotting revealed significantly decreased APPL1 expression in the dorsal horn in streptozocin-induced rats versus the control rats, coupled with concomitant mechanical and thermal hyperalgesia. Afterward, the determination of exact localization of APPL1 in spinal cord by immunofluorescent staining assay revealed highly expressed APPL1 in the lamina of spinal dorsal horn in control rats, with the overexpression in neurons, microglia, and underexpression in astrocytes. The APPL1 expression in laminae I and II was significantly downregulated in painful diabetic neuropathy rats. In addition, APPL1 deficiency or overexpression contributed to the increase or decrease of Map and Bassoon, respectively. The localization and immunoactivity of APPL1 and mammalian target of rapamycin (mTOR) were determined in spinal dorsal horn in painful diabetic neuropathy rats and control rats by immunohistochemistry, suggesting pronounced decrease in APPL1 expression in the superficial layer of the spinal cord in painful diabetic neuropathy rats, with p-mTOR expression markedly augmented. APPL1 knockdown by infection with lentiviral vector facilitated the activation of mTOR and abrogated mechanical withdrawal threshold values in painful diabetic neuropathy rats. Genetically overexpressed APPL1 significantly eliminated the activation of mTOR and resulted in the augmented mechanical withdrawal threshold values and thermal withdrawal latency values. Furthermore, the APPL1 levels affect phosphorylation of adenosine monophosphate-activated protein kinase (AMPK), and Akt, as well as the small GTPase, Rab5 expression in painful diabetic neuropathy rats. Our results uncovered a novel mechanism by which APPL1 deficiency facilitates the mTOR activation and thus exacerbates the hyperalgesia in streptozocin-induced diabetic rats, presumably via the regulation of Rab5/Akt and AMPK signaling pathway.
Collapse
Affiliation(s)
- Wan-You He
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| | - Bin Zhang
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| | - Wei-Cheng Zhao
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| | - Jian He
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| | - Yunhua Wang
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| | - Lei Zhang
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| | - Qing-Ming Xiong
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| | - Han-Bing Wang
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| |
Collapse
|