1
|
Aiad AAE, El-Haggar SM, El-Barbary AM, El-Afify DR. Metformin as adjuvant therapy in obese knee osteoarthritis patients. Inflammopharmacology 2024; 32:2349-2359. [PMID: 38869746 PMCID: PMC11300470 DOI: 10.1007/s10787-024-01495-y] [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: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 06/14/2024]
Abstract
AIMS This study aimed at investigating the efficacy of metformin as adjuvant therapy for obese knee osteoarthritis (OA) patients, considering its anti-inflammatory and cartilage-protective effects. PATIENTS AND METHODS In this randomized, double-blind, placebo-controlled study, 50 obese knee OA patients were assigned randomly to two groups, the metformin group (n = 25) which was treated with metformin 500 mg orally BID plus celecoxib 200 mg orally once daily, and the placebo group (n = 25) which was treated with placebo tablets BID plus celecoxib 200 mg orally once daily for 12 weeks. Cartilage Oligomeric Matrix Protein (COMP), C-terminal cross-linked telopeptide of type I collagen (CTX-1), and Interleukin 1-beta (IL-1β) serum levels were measured, while Western Ontario and McMaster Universities Arthritis Index (WOMAC) score assessed knee pain, stiffness, and physical function at baseline and after 12 weeks. RESULTS Following a 12-week treatment, the metformin group exhibited significantly reduced levels of COMP, CTX-1, and IL-1β in the serum compared to the placebo group (p = 0.0081, p = 0.0106, and p = 0.0223, respectively). Furthermore, metformin group produced significant improvements in WOMAC total scale (p < 0.0001), specifically in knee pain, stiffness, and physical function compared to placebo group (p < 0.0001, p < 0.0001, and p < 0.0001, respectively). CONCLUSION Metformin as an adjuvant therapy in obese knee OA patients may have beneficial effects on cartilage degradation and inflammation, as evidenced by the significant decreases in serum COMP, CTX-1, and IL-1β levels. Additionally, metformin may improve clinical outcomes, as shown by the significant improvements in WOMAC scores. CLINICALTRIALS GOV ID NCT05638893/Registered December 6, 2022 - Retrospectively.
Collapse
Affiliation(s)
- Amany Abd Elaal Aiad
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | | | - Amal Mohamed El-Barbary
- Department of Physical Medicine, Rheumatology and Rehabilitation, Faculty of Medicine, Tanta University, Tanta, 31527, Egypt
| | - Dalia Refat El-Afify
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
2
|
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
|
3
|
Tang X, Balachandran RC, Aschner M, Bowman AB. IGF/mTORC1/S6 Signaling Is Potentiated and Prolonged by Acute Loading of Subtoxicological Manganese Ion. Biomolecules 2023; 13:1229. [PMID: 37627294 PMCID: PMC10452562 DOI: 10.3390/biom13081229] [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: 06/10/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
The insulin-like growth factor (IGF)/insulin signaling (IIS) pathway is involved in cellular responses against intracellular divalent manganese ion (Mn2+) accumulation. As a pathway where multiple nodes utilize Mn2+ as a metallic co-factor, how the IIS signaling patterns are affected by Mn2+ overload is unresolved. In our prior studies, acute Mn2+ exposure potentiated IIS kinase activity upon physiological-level stimulation, indicated by elevated phosphorylation of protein kinase B (PKB, also known as AKT). AKT phosphorylation is associated with IIS activity; and provides direct signaling transduction input for the mammalian target of rapamycin complex 1 (mTORC1) and its downstream target ribosomal protein S6 (S6). Here, to better define the impact of Mn2+ exposure on IIS function, Mn2+-induced IIS activation was evaluated with serial concentrations and temporal endpoints. In the wild-type murine striatal neuronal line STHdh, the acute treatment of Mn2+ with IGF induced a Mn2+ concentration-sensitive phosphorylation of S6 at Ser235/236 to as low as 5 μM extracellular Mn2+. This effect required both the essential amino acids and insulin receptor (IR)/IGF receptor (IGFR) signaling input. Similar to simultaneous stimulation of Mn2+ and IGF, when a steady-state elevation of Mn2+ was established via a 24-h pre-exposure, phosphorylation of S6 also displayed higher sensitivity to sub-cytotoxic Mn2+ when compared to AKT phosphorylation at Ser473. This indicates a synergistic effect of sub-cytotoxic Mn2+ on IIS and mTORC1 signaling. Furthermore, elevated intracellular Mn2+, with both durations, led to a prolonged activation in AKT and S6 upon stimulation. Our data demonstrate that the downstream regulator S6 is a highly sensitive target of elevated Mn2+ and is well below the established acute cytotoxicity thresholds (<50 μM). These findings indicate that the IIS/mTORC1 pathways, in which Mn2+ normally serves as an essential co-factor, are dually responsible for the cellular changes in exposures to real-world Mn2+ concentrations.
Collapse
Affiliation(s)
- Xueqi Tang
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA; (X.T.)
| | - Rekha C. Balachandran
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA; (X.T.)
- Exponent Inc., Alexandria, VA 22314, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA; (X.T.)
| |
Collapse
|
4
|
Lambova SN. Pleiotropic Effects of Metformin in Osteoarthritis. Life (Basel) 2023; 13:life13020437. [PMID: 36836794 PMCID: PMC9960992 DOI: 10.3390/life13020437] [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: 12/26/2022] [Revised: 01/15/2023] [Accepted: 01/25/2023] [Indexed: 02/09/2023] Open
Abstract
The involvement of the knee joint is the most common localization of the pathological process in osteoarthritis (OA), which is associated with obesity in over 50% of the patients and is mediated by mechanical, inflammatory, and metabolic mechanisms. Obesity and the associated conditions (hyperglycemia, dyslipidemia, and hypertension) have been found to be risk factors for the development of knee OA, which has led to the emerging concept of the existence of a distinct phenotype, i.e., metabolic knee OA. Combined assessment of markers derived from dysfunctional adipose tissue, markers of bone and cartilage metabolism, as well as high-sensitivity inflammatory markers and imaging, might reveal prognostic signs for metabolic knee OA. Interestingly, it has been suggested that drugs used for the treatment of other components of the metabolic syndrome may also affect the clinical course and retard the progression of metabolic-associated knee OA. In this regard, significant amounts of new data are accumulating about the role of metformin-a drug, commonly used in clinical practice with suggested multiple pleiotropic effects. The aim of the current review is to analyze the current views about the potential pleiotropic effects of metformin in OA. Upon the analysis of the different effects of metformin, major mechanisms that might be involved in OA are the influence of inflammation, oxidative stress, autophagy, adipokine levels, and microbiome modulation. There is an increasing amount of evidence from in vitro studies, animal models, and clinical trials that metformin can slow OA progression by modulating inflammatory and metabolic factors that are summarized in the current up-to-date review. Considering the contemporary concept about the existence of metabolic type knee OA, in which the accompanying obesity and systemic low-grade inflammation are suggested to influence disease course, metformin could be considered as a useful and safe component of the personalized therapeutic approach in knee OA patients with accompanying type II diabetes or obesity.
Collapse
Affiliation(s)
- Sevdalina Nikolova Lambova
- Department of Propaedeutics of Internal Diseases “Prof Dr Anton Mitov”, Faculty of Medicine, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
- Department in Rheumatology, MHAT “Sveti Mina”, 4002 Plovdiv, Bulgaria
| |
Collapse
|
5
|
Yeo JH, Roh DH. The mTOR inhibitor rapamycin suppresses trigeminal neuropathic pain and p-MKK4/p-p38 mitogen-activated protein kinase-mediated microglial activation in the trigeminal nucleus caudalis of mice with infraorbital nerve injury. Front Mol Neurosci 2023; 16:1172366. [PMID: 37122619 PMCID: PMC10140572 DOI: 10.3389/fnmol.2023.1172366] [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: 02/23/2023] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
Neuropathic pain caused by trigeminal nerve injury is a typical refractory orofacial chronic pain accompanied by the development of hyperalgesia and allodynia. We previously demonstrated that the mammalian target of rapamycin (mTOR) inhibitor rapamycin suppressed orofacial formalin injection-induced nociception; however, the underlying mechanism is unclear, and it is unknown whether it can reduce trigeminal neuropathic pain. In mice, left infraorbital nerve and partial nerve ligation (ION-pNL) was performed using a silk suture (8-0). Fourteen days after surgery, neuropathic pain behavior was examined on a whisker pad and rapamycin (0.1, 0.3, and 1.0 mg/kg) was administered intraperitoneally. Mechanical and cold sensitivities in the orofacial region were quantified using von Frey filaments and acetone solution, respectively. Changes in mTOR and related proteins, such as p-MKK3/6, p-MKK4, p-JNK, p-ERK, p-p38 MAPK, GFAP, and Iba-1, in the trigeminal nucleus caudalis (TNC) or the trigeminal ganglia (TG) tissues were examined via western blot analysis or immunohistochemistry. Mice demonstrated significant mechanical and cold allodynia 2 weeks following ION-pNL injury, both of which were significantly reduced 1 h after the administration of high-dose rapamycin (1.0 mg/kg). In the TG tissue, ION-pNL surgery or rapamycin treatment did not change p-mTOR and p-4EBP1, but rapamycin reduced the increase of p-S6 and S6 induced by ION-pNL. In the TNC tissue, neither ION-pNL surgery nor rapamycin treatment altered p-mTOR, p-S6, and p-4EBP1 expressions, whereas rapamycin significantly decreased the ION-pNL-induced increase in Iba-1 expression. In addition, rapamycin suppressed the increase in p-p38 MAPK and p-MKK4 expressions but not p-MKK3/6 expression. Moreover, p-p38 MAPK-positive cells were colocalized with increased Iba-1 in the TNC. Our findings indicate that rapamycin treatment reduces both mechanical and cold orofacial allodynia in mice with trigeminal neuropathic pain, which is closely associated with the modulation of p-MKK4/p-p38 MAPK-mediated microglial activation in the TNC.
Collapse
|
6
|
Chen L, Hu Y, Wang S, Cao K, Mai W, Sha W, Ma H, Zeng LH, Xu ZZ, Gao YJ, Duan S, Wang Y, Gao Z. mTOR-neuropeptide Y signaling sensitizes nociceptors to drive neuropathic pain. JCI Insight 2022; 7:159247. [PMID: 36194480 DOI: 10.1172/jci.insight.159247] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 09/29/2022] [Indexed: 12/15/2022] Open
Abstract
Neuropathic pain is a refractory condition that involves de novo protein synthesis in the nociceptive pathway. The mTOR is a master regulator of protein translation; however, mechanisms underlying its role in neuropathic pain remain elusive. Using the spared nerve injury-induced neuropathic pain model, we found that mTOR was preferentially activated in large-diameter dorsal root ganglion (DRG) neurons and spinal microglia. However, selective ablation of mTOR in DRG neurons, rather than microglia, alleviated acute neuropathic pain in mice. We show that injury-induced mTOR activation promoted the transcriptional induction of neuropeptide Y (Npy), likely via signal transducer and activator of transcription 3 phosphorylation. NPY further acted primarily on Y2 receptors (Y2R) to enhance neuronal excitability. Peripheral replenishment of NPY reversed pain alleviation upon mTOR removal, whereas Y2R antagonists prevented pain restoration. Our findings reveal an unexpected link between mTOR and NPY/Y2R in promoting nociceptor sensitization and neuropathic pain.
Collapse
Affiliation(s)
- Lunhao Chen
- Spine Lab, Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yaling Hu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Siyuan Wang
- Spine Lab, Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kelei Cao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Weihao Mai
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
| | - Weilin Sha
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, China
| | - Huan Ma
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Ling-Hui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Zhen-Zhong Xu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yong-Jing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, China
| | - Shumin Duan
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yue Wang
- Spine Lab, Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihua Gao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| |
Collapse
|
7
|
Wang S, Dai Y. Roles of AMPK and Its Downstream Signals in Pain Regulation. Life (Basel) 2021; 11:life11080836. [PMID: 34440581 PMCID: PMC8401922 DOI: 10.3390/life11080836] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/06/2021] [Accepted: 08/12/2021] [Indexed: 12/20/2022] Open
Abstract
Pain is an unpleasant sensory and emotional state that decreases quality of life. A metabolic sensor, adenosine monophosphate-activated protein kinase (AMPK), which is ubiquitously expressed in mammalian cells, has recently attracted interest as a new target of pain research. Abnormal AMPK expression and function in the peripheral and central nervous systems are associated with various types of pain. AMPK and its downstream kinases participate in the regulation of neuron excitability, neuroinflammation and axonal and myelin regeneration. Numerous AMPK activators have reduced pain behavior in animal models. The current understanding of pain has been deepened by AMPK research, but certain issues, such as the interactions of AMPK at each step of pain regulation, await further investigation. This review examines the roles of AMPK and its downstream kinases in neurons and non-neuronal cells, as well as their contribution to pain regulation.
Collapse
Affiliation(s)
- Shenglan Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe 650-8530, Japan
- Correspondence: (S.W.); (Y.D.); Tel.: +86-10-53912197 (S.W.); +81-78-304-3147 (Y.D.)
| | - Yi Dai
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe 650-8530, Japan
- Traditional Medicine Research Center, Chinese Medicine Confucius Institute, Hyogo College of Medicine, Kobe 663-8501, Japan
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Nishinomiya 663-8501, Japan
- Correspondence: (S.W.); (Y.D.); Tel.: +86-10-53912197 (S.W.); +81-78-304-3147 (Y.D.)
| |
Collapse
|
8
|
Acupuncture alleviates chronic pain and comorbid conditions in a mouse model of neuropathic pain: the involvement of DNA methylation in the prefrontal cortex. Pain 2021; 162:514-530. [PMID: 32796318 PMCID: PMC7808350 DOI: 10.1097/j.pain.0000000000002031] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022]
Abstract
ABSTRACT Chronic pain reduces life quality and is an important clinical problem associated with emotional and cognitive dysfunction. Epigenetic regulation of DNA methylation is involved in the induction of abnormal behaviors and pathological gene expression. We examined whether acupuncture can restore epigenetic changes caused by chronic pain, and identified the underlying mechanisms in neuropathic pain mice. Acupuncture treatment for 6 months (3 days/week) improved mechanical/cold allodynia and the emotional/cognitive dysfunction caused by left partial sciatic nerve ligation (PSNL)-induced neuropathic pain. The effects of acupuncture were associated with global DNA methylation recovery in the prefrontal cortex (PFC). Analysis of DNA methylation patterns in PFC indicated that 1364 overlapping genes among 4442 and 4416 methylated genes in the PSNL vs sham and PSNL vs acupuncture points groups, respectively, were highly associated with the DNA methylation process. Acupuncture restored the reduced expression of 5-methylcytosine, methyl-cytosine-phospho-guanine binding protein 2, and DNA methyltransferase family enzymes induced by PSNL in PFC. Methylation levels of Nr4a1 and Chkb associated with mitochondrial dysfunction were decreased in PFC of the PSNL mice, and increased by acupuncture. By contrast, high expression of Nr4a1 and Chkb mRNA in PSNL mice decreased after acupuncture. We also found that acupuncture inhibited the expression of Ras pathway-related genes such as Rasgrp1 and Rassf1. Finally, the expression of Nr4a1, Rasgrp1, Rassf1, and Chkb mRNA increased in the neuronal cells treated with Mecp2 small interfering RNA. These results suggest that acupuncture can relieve chronic pain-induced comorbid conditions by altering DNA methylation of Nr4a1, Rasgrp1, Rassf1, and Chkb in the PFC.
Collapse
|
9
|
Yousuf MS, Shiers SI, Sahn JJ, Price TJ. Pharmacological Manipulation of Translation as a Therapeutic Target for Chronic Pain. Pharmacol Rev 2021; 73:59-88. [PMID: 33203717 PMCID: PMC7736833 DOI: 10.1124/pharmrev.120.000030] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dysfunction in regulation of mRNA translation is an increasingly recognized characteristic of many diseases and disorders, including cancer, diabetes, autoimmunity, neurodegeneration, and chronic pain. Approximately 50 million adults in the United States experience chronic pain. This economic burden is greater than annual costs associated with heart disease, cancer, and diabetes combined. Treatment options for chronic pain are inadequately efficacious and riddled with adverse side effects. There is thus an urgent unmet need for novel approaches to treating chronic pain. Sensitization of neurons along the nociceptive pathway causes chronic pain states driving symptoms that include spontaneous pain and mechanical and thermal hypersensitivity. More than a decade of preclinical research demonstrates that translational mechanisms regulate the changes in gene expression that are required for ongoing sensitization of nociceptive sensory neurons. This review will describe how key translation regulation signaling pathways, including the integrated stress response, mammalian target of rapamycin, AMP-activated protein kinase (AMPK), and mitogen-activated protein kinase-interacting kinases, impact the translation of different subsets of mRNAs. We then place these mechanisms of translation regulation in the context of chronic pain states, evaluate currently available therapies, and examine the potential for developing novel drugs. Considering the large body of evidence now published in this area, we propose that pharmacologically manipulating specific aspects of the translational machinery may reverse key neuronal phenotypic changes causing different chronic pain conditions. Therapeutics targeting these pathways could eventually be first-line drugs used to treat chronic pain disorders. SIGNIFICANCE STATEMENT: Translational mechanisms regulating protein synthesis underlie phenotypic changes in the sensory nervous system that drive chronic pain states. This review highlights regulatory mechanisms that control translation initiation and how to exploit them in treating persistent pain conditions. We explore the role of mammalian/mechanistic target of rapamycin and mitogen-activated protein kinase-interacting kinase inhibitors and AMPK activators in alleviating pain hypersensitivity. Modulation of eukaryotic initiation factor 2α phosphorylation is also discussed as a potential therapy. Targeting specific translation regulation mechanisms may reverse changes in neuronal hyperexcitability associated with painful conditions.
Collapse
Affiliation(s)
- Muhammad Saad Yousuf
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas (M.S.Y., S.I.S., T.J.P.) and 4E Therapeutics Inc, Austin, Texas (J.J.S.)
| | - Stephanie I Shiers
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas (M.S.Y., S.I.S., T.J.P.) and 4E Therapeutics Inc, Austin, Texas (J.J.S.)
| | - James J Sahn
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas (M.S.Y., S.I.S., T.J.P.) and 4E Therapeutics Inc, Austin, Texas (J.J.S.)
| | - Theodore J Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas (M.S.Y., S.I.S., T.J.P.) and 4E Therapeutics Inc, Austin, Texas (J.J.S.)
| |
Collapse
|
10
|
Cha M, Choi S, Kim K, Lee BH. Manganese-enhanced MRI depicts a reduction in brain responses to nociception upon mTOR inhibition in chronic pain rats. Mol Brain 2020; 13:158. [PMID: 33267907 PMCID: PMC7713325 DOI: 10.1186/s13041-020-00687-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/17/2020] [Indexed: 11/30/2022] Open
Abstract
Neuropathic pain induced by a nerve injury can lead to chronic pain. Recent studies have reported hyperactive neural activities in the nociceptive-related area of the brain as a result of chronic pain. Although cerebral activities associated with hyperalgesia and allodynia in chronic pain models are difficult to represent with functional imaging techniques, advances in manganese (Mn)-enhanced magnetic resonance imaging (MEMRI) could facilitate the visualization of the activation of pain-specific neural responses in the cerebral cortex. In order to investigate the alleviation of pain nociception by mammalian target of rapamycin (mTOR) modulation, we observed cerebrocortical excitability changes and compared regional Mn2+ enhancement after mTOR inhibition. At day 7 after nerve injury, drugs were applied into the intracortical area, and drug (Vehicle, Torin1, and XL388) effects were compared within groups using MEMRI. Therein, signal intensities of the insular cortex (IC), primary somatosensory cortex of the hind limb region, motor cortex 1/2, and anterior cingulate cortex regions were significantly reduced after application of mTOR inhibitors (Torin1 and XL388). Furthermore, rostral-caudal analysis of the IC indicated that the rostral region of the IC was more strongly associated with pain perception than the caudal region. Our data suggest that MEMRI can depict pain-related signal changes in the brain and that mTOR inhibition is closely correlated with pain modulation in chronic pain rats.
Collapse
Affiliation(s)
- Myeounghoon Cha
- Department of Physiology, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, 03722, Seoul, Republic of Korea.
| | - Songyeon Choi
- Department of Physiology, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, 03722, Seoul, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 03722, Seoul, Republic of Korea
| | - Kyeongmin Kim
- Department of Physiology, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, 03722, Seoul, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 03722, Seoul, Republic of Korea
| | - Bae Hwan Lee
- Department of Physiology, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, 03722, Seoul, Republic of Korea. .,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 03722, Seoul, Republic of Korea.
| |
Collapse
|
11
|
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: 5.5] [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
|
12
|
Persistent Rheb-induced mTORC1 activation in spinal cord neurons induces hypersensitivity in neuropathic pain. Cell Death Dis 2020; 11:747. [PMID: 32920594 PMCID: PMC7487067 DOI: 10.1038/s41419-020-02966-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/21/2020] [Accepted: 08/31/2020] [Indexed: 12/21/2022]
Abstract
The small GTPase Ras homolog enriched in the brain (Rheb) can activate mammalian target of rapamycin (mTOR) and regulate the growth and cell cycle progression. We investigated the role of Rheb-mediated mTORC1 signaling in neuropathic pain. A chronic constriction injury (CCI) model was dopted. CCI induced obvious spinal Rheb expression and phosphorylation of mTOR, S6, and 4-E-BP1. Blocking mTORC1 signal with rapamycin alleviated the neuropathic pain and restored morphine efficacy in CCI model. Immunofluoresence showed a neuronal co-localization of CCI-induced Rheb and pS6. Rheb knockin mouse showed a similar behavioral phenotype as CCI. In spinal slice recording, CCI increased the firing frequency of neurons expressing HCN channels; inhibition of mTORC1 with rapamycin could reverse the increased spinal neuronal activity in neuropathic pain. Spinal Rheb is induced in neuropathic pain, which in turn active the mTORC1 signaling in CCI. Spinal Rheb-mTOR signal plays an important role in regulation of spinal sensitization in neuropathic pain, and targeting mTOR may give a new strategy for pain management.
Collapse
|
13
|
Ma X, Liu S, Liu D, Wang Q, Li H, Zhao Z. Exercise intervention attenuates neuropathic pain in diabetes via mechanisms of mammalian target of rapamycin (mTOR). Arch Physiol Biochem 2020; 126:41-48. [PMID: 30317878 DOI: 10.1080/13813455.2018.1489851] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
This study was to examine the role of exercise intervention in modulating neuropathic pain induced by diabetes. Diabetes was induced by streptozotocin (STZ, i.p.) in rats and mechanical hyperalgesia was observed three weeks after STZ. Mechanical withdrawal thresholds were increased after four to five weeks of exercise in STZ rats. We also examined the role of signal of mammalian target of rapamycin (mTOR) in regulating neuropathic pain. Inhibition of neuropathic pain by exercise in STZ rats was accompanied with decreases of p-mTOR, p-S6K1, and p-4E-BP1 in sensory nerves. Blocking mTOR also elevated mechanical withdrawal thresholds in STZ rats. Furthermore, pro-inflammatory IL-6 was greater in sensory nerves of STZ rats. Inhibition of IL-6 decreased mTOR and increased mechanical withdrawal thresholds in STZ rats. Overall, our data suggest the role played by exercise in improving neuropathic pain after STZ and that IL-6-mTOR signal is a part of mechanisms engaged in the effects of exercise.
Collapse
Affiliation(s)
- Xiaohui Ma
- Tumor Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Sitong Liu
- Tumor Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Dongxue Liu
- Tumor Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Qian Wang
- Tumor Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Hongwei Li
- Tumor Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhen Zhao
- Department of Blood Transfusion, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
14
|
He WY, Zhang B, Zhao WC, He J, Zhang L, Xiong QM, Wang J, Wang HB. Contributions of mTOR Activation-Mediated Upregulation of Synapsin II and Neurite Outgrowth to Hyperalgesia in STZ-Induced Diabetic Rats. ACS Chem Neurosci 2019; 10:2385-2396. [PMID: 30785256 DOI: 10.1021/acschemneuro.8b00680] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Painful diabetic neuropathy (PDN) is among the common complications in diabetes mellitus (DM), with its underlying mechanisms largely unknown. Synapsin II is primarily expressed in the spinal dorsal horn, and its upregulation mediates a superfluous release of glutamate and a deficiency of GABAergic interneuron synaptic transmission, which is directly implicated in the facilitation of pain signals in the hyperalgesic nociceptive response. Recently, synapsin II has been revealed to be associated with the modulation of neurite outgrowth, whereas the process of this neuronal structural neuroplasticity following neuronal hyperexcitability still remains unclear. In this study, we found that under conditions of elevated glucose, TNF-α induced the activation of mTOR, mediating the upregulation of synapsin II and neurite outgrowth in dorsal horn neurons. In vivo, we demonstrated that mTOR and synapsin II were upregulated and coexpressed in the spinal dorsal horn neurons in rats with streptozotocin (STZ)-induced diabetes. Furthermore, the intrathecal administration of the mTOR inhibitor rapamycin or synapsin II shRNA significantly diminished the expression of synapsin II, effectively mitigating hyperalgesia in PDN rats. We are the first to discover that in STZ-induced diabetic rats the activation of mTOR mediates the upregulation of synapsin II and neurite outgrowth, both contributing to hyperalgesia. These findings may benefit the clinical therapy of PDN by provision of a novel target.
Collapse
Affiliation(s)
- Wan-you He
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Bin Zhang
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Wei-cheng Zhao
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Jian He
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Lei Zhang
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Qing-ming Xiong
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Jing Wang
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Han-bing Wang
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| |
Collapse
|
15
|
Li S, Guan S, Wang Y, Cheng L, Yang Q, Tian Z, Zhao M, Wang X, Feng B. Nicotine inhibits rapamycin-induced pain through activating mTORC1/S6K/IRS-1-related feedback inhibition loop. Brain Res Bull 2019; 149:75-85. [PMID: 31005665 DOI: 10.1016/j.brainresbull.2019.04.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/11/2019] [Accepted: 04/16/2019] [Indexed: 11/19/2022]
Abstract
Mammalian target of rapamycin complex 1 (mTORC1) inhibitors increase the incidence of pain in patients, and this finding has been replicated in animal models. However, reports on possible analgesics for this condition are scant. Accumulating evidence finds that nicotinic acetylcholine receptors (nAChRs) are involved in mediating pain. However, whether nicotine, a full agonist of nAChRs, alleviates mTORC1 inhibition-induced pain and its underlying mechanisms remain unknown. In this study, pain was induced in naïve male C57BL/6J mice by intraperitoneally injecting rapamycin acutely or repeatedly. Subsequently, pain thresholds, including mechanical and thermal pain, were measured. The involving signaling pathway was tested using western blot analysis and immunofluorescent assay. Changes in neuronal excitability caused by different treatments were also analyzed using whole-cell recording. Microinjection into the anterior cingulate cortex (ACC) was used to test the role of nAChRs containing the α4β2 or α7 subtype in this brain region in pain modulation. Our results showed that nicotine significantly reduced hyperalgesia in mice that received acute or repeated rapamycin injections, and reversed the effects of rapamycin on the phosphorylation of S6K, 4E-BP1, insulin receptor substrate-1 (IRS-1) at Ser636/639, AKT at Ser473, and ERK at Thr202/Tyr204. Whole-cell recording results showed that nicotine reduced the firing rates of pyramidal neurons in the ACC, and a pharmacological blockade of nAChRs containing the α4β2 or α7 subtype in ACC inhibited the antinociceptive effects of nicotine in mice with rapamycin-induced pain. Our findings indicate that analgesics targeting nAChRs can be developed to help patients with rapamycin-induced pain.
Collapse
Affiliation(s)
- Shuo Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Shaoyu Guan
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Yurong Wang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Pharmacy, School of Stomatology, Fourth Military Medical University, Xi'an, China; College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Lifei Cheng
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Pharmacy, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Qi Yang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Zhen Tian
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Minggao Zhao
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Xiaojuan Wang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Pharmacy, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Bin Feng
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Pharmacy, School of Stomatology, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
16
|
Feng XL, Deng HB, Wang ZG, Wu Y, Ke JJ, Feng XB. Suberoylanilide Hydroxamic Acid Triggers Autophagy by Influencing the mTOR Pathway in the Spinal Dorsal Horn in a Rat Neuropathic Pain Model. Neurochem Res 2018; 44:450-464. [PMID: 30560396 DOI: 10.1007/s11064-018-2698-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 11/05/2018] [Accepted: 12/10/2018] [Indexed: 02/08/2023]
Abstract
Histone acetylation levels can be upregulated by treating cells with histone deacetylase inhibitors (HDACIs), which can induce autophagy. Autophagy flux in the spinal cord of rats following the left fifth lumber spinal nerve ligation (SNL) is involved in the progression of neuropathic pain. Suberoylanilide hydroxamic acid (SAHA), one of the HDACIs can interfere with the epigenetic process of histone acetylation, which has been shown to ease neuropathic pain. Recent research suggest that SAHA can stimulate autophagy via the mammalian target of rapamycin (mTOR) pathway in some types of cancer cells. However, little is known about the role of SAHA and autophagy in neuropathic pain after nerve injury. In the present study, we aim to investigate autophagy flux and the role of the mTOR pathway on spinal cells autophagy activation in neuropathic pain induced by SNL in rats that received SAHA treatment. Autophagy-related proteins and mTOR or its active form were assessed by using western blot, immunohistochemistry, double immunofluorescence staining and transmission electron microscopy (TEM). We found that SAHA decreased the paw mechanical withdrawal threshold (PMWT) of the lower compared with SNL. Autophagy flux was mainly disrupted in the astrocytes and neuronal cells of the spinal cord dorsal horn on postsurgical day 28 and was reversed by daily intrathecal injection of SAHA (n = 100 nmol/day or n = 200 nmol/day). SAHA also decreased mTOR and phosphorylated mTOR (p-mTOR) expression, especially p-mTOR expression in astrocytes and neuronal cells of the spinal dorsal horn. These results suggest that SAHA attenuates neuropathic pain and contributes to autophagy flux in astrocytes and neuronal cells of the spinal dorsal horn via the mTOR signaling pathway.
Collapse
Affiliation(s)
- Xiang-Lan Feng
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Rd, Wuhan, 430071, Hubei, China
| | - Hong-Bo Deng
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Rd, Wuhan, 430071, Hubei, China
| | - Zheng-Gang Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Rd, Wuhan, 430071, Hubei, China
| | - Yun Wu
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Rd, Wuhan, 430071, Hubei, China
| | - Jian-Juan Ke
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Rd, Wuhan, 430071, Hubei, China
| | - Xiao-Bo Feng
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Rd, Wuhan, 430071, Hubei, China.
| |
Collapse
|
17
|
Perspectives of RAS and RHEB GTPase Signaling Pathways in Regenerating Brain Neurons. Int J Mol Sci 2018; 19:ijms19124052. [PMID: 30558189 PMCID: PMC6321366 DOI: 10.3390/ijms19124052] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/05/2018] [Accepted: 12/13/2018] [Indexed: 12/29/2022] Open
Abstract
Cellular activation of RAS GTPases into the GTP-binding “ON” state is a key switch for regulating brain functions. Molecular protein structural elements of rat sarcoma (RAS) and RAS homolog protein enriched in brain (RHEB) GTPases involved in this switch are discussed including their subcellular membrane localization for triggering specific signaling pathways resulting in regulation of synaptic connectivity, axonal growth, differentiation, migration, cytoskeletal dynamics, neural protection, and apoptosis. A beneficial role of neuronal H-RAS activity is suggested from cellular and animal models of neurodegenerative diseases. Recent experiments on optogenetic regulation offer insights into the spatiotemporal aspects controlling RAS/mitogen activated protein kinase (MAPK) or phosphoinositide-3 kinase (PI3K) pathways. As optogenetic manipulation of cellular signaling in deep brain regions critically requires penetration of light through large distances of absorbing tissue, we discuss magnetic guidance of re-growing axons as a complementary approach. In Parkinson’s disease, dopaminergic neuronal cell bodies degenerate in the substantia nigra. Current human trials of stem cell-derived dopaminergic neurons must take into account the inability of neuronal axons navigating over a large distance from the grafted site into striatal target regions. Grafting dopaminergic precursor neurons directly into the degenerating substantia nigra is discussed as a novel concept aiming to guide axonal growth by activating GTPase signaling through protein-functionalized intracellular magnetic nanoparticles responding to external magnets.
Collapse
|
18
|
Khoutorsky A, Price TJ. Translational Control Mechanisms in Persistent Pain. Trends Neurosci 2018; 41:100-114. [PMID: 29249459 DOI: 10.1016/j.tins.2017.11.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/14/2017] [Accepted: 11/22/2017] [Indexed: 12/21/2022]
Abstract
Persistent pain, which is poorly treated and estimated to afflict one third of the world's population, is largely mediated by the sensitization of nociceptive neurons. This sensitization involves de novo gene expression to support biochemical and structural changes required to maintain amplified pain signaling that frequently persists even after injury to tissue resolves. While transcription-dependent changes in gene expression are important, recent work demonstrates that activity-dependent regulation of mRNA translation is key to controlling the cellular proteome and the development and maintenance of persistent pain. In this review, we highlight recent advances in translational regulation of gene expression in nociceptive circuits, with a focus on key signaling pathways and mRNA targets that may be tractable for the creation of next-generation pain therapeutics.
Collapse
Affiliation(s)
- Arkady Khoutorsky
- Department of Anesthesia and Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, H3A 0G1, Canada.
| | - Theodore J Price
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA.
| |
Collapse
|
19
|
Uttam S, Wong C, Price TJ, Khoutorsky A. eIF4E-Dependent Translational Control: A Central Mechanism for Regulation of Pain Plasticity. Front Genet 2018; 9:470. [PMID: 30459806 PMCID: PMC6232926 DOI: 10.3389/fgene.2018.00470] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/24/2018] [Indexed: 01/04/2023] Open
Abstract
Translational control of gene expression has emerged as a key mechanism in regulating different forms of long-lasting neuronal plasticity. Maladaptive plastic reorganization of peripheral and spinal nociceptive circuits underlies many chronic pain states and relies on new gene expression. Accordingly, downregulation of mRNA translation in primary afferents and spinal dorsal horn neurons inhibits tissue injury-induced sensitization of nociceptive pathways, supporting a central role for translation dysregulation in the development of persistent pain. Translation is primarily regulated at the initiation stage via the coordinated activity of translation initiation factors. The mRNA cap-binding protein, eukaryotic translation initiation factor 4E (eIF4E), is involved in the recruitment of the ribosome to the mRNA cap structure, playing a central role in the regulation of translation initiation. eIF4E integrates inputs from the mTOR and ERK signaling pathways, both of which are activated in numerous painful conditions to regulate the translation of a subset of mRNAs. Many of these mRNAs are involved in the control of cell growth, proliferation, and neuroplasticity. However, the full repertoire of eIF4E-dependent mRNAs in the nervous system and their translation regulatory mechanisms remain largely unknown. In this review, we summarize the current evidence for the role of eIF4E-dependent translational control in the sensitization of pain circuits and present pharmacological approaches to target these mechanisms. Understanding eIF4E-dependent translational control mechanisms and their roles in aberrant plasticity of nociceptive circuits might reveal novel therapeutic targets to treat persistent pain states.
Collapse
Affiliation(s)
- Sonali Uttam
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Calvin Wong
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Theodore J. Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX, United States
| | - Arkady Khoutorsky
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| |
Collapse
|
20
|
Chen XB, Wang ZL, Yang QY, Zhao FY, Qin XL, Tang XE, Du JL, Chen ZH, Zhang K, Huang FJ. Diosgenin Glucoside Protects against Spinal Cord Injury by Regulating Autophagy and Alleviating Apoptosis. Int J Mol Sci 2018; 19:ijms19082274. [PMID: 30072674 PMCID: PMC6121626 DOI: 10.3390/ijms19082274] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 08/01/2018] [Accepted: 08/01/2018] [Indexed: 12/25/2022] Open
Abstract
Spinal cord injury (SCI) is a severe traumatic lesion of central nervous system (CNS) with only a limited number of restorative therapeutic options. Diosgenin glucoside (DG), a major bioactive ingredient of Trillium tschonoskii Max., possesses neuroprotective effects through its antioxidant and anti-apoptotic functions. In this study, we investigated the therapeutic benefit and underlying mechanisms of DG treatment in SCI. We found that in Sprague-Dawley rats with traumatic SCI, the expressions of autophagy marker Light Chain 3 (LC3) and Beclin1 were decreased with concomitant accumulation of autophagy substrate protein p62 and ubiquitinated proteins, indicating an impaired autophagic activity. DG treatment, however, significantly attenuated p62 expression and upregulated the Rheb/mTOR signaling pathway (evidenced as Ras homolog enriched in brain) due to the downregulation of miR-155-3p. We also observed significantly less tissue injury and edema in the DG-treated group, leading to appreciable functional recovery compared to that of the control group. Overall, the observed neuroprotection afforded by DG treatment warrants further investigation on its therapeutic potential in SCI.
Collapse
Affiliation(s)
- Xian-Bing Chen
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China.
- College of Medicine, Hubei University for Nationalities, Enshi 445000, China.
| | - Zi-Li Wang
- College of Medicine, Hubei University for Nationalities, Enshi 445000, China.
| | - Qing-Yu Yang
- College of Medicine, Hubei University for Nationalities, Enshi 445000, China.
| | - Fang-Yu Zhao
- College of Medicine, Hubei University for Nationalities, Enshi 445000, China.
| | - Xiao-Li Qin
- College of Medicine, Hubei University for Nationalities, Enshi 445000, China.
| | - Xian-E Tang
- College of Medicine, Hubei University for Nationalities, Enshi 445000, China.
| | - Jun-Long Du
- College of Medicine, Hubei University for Nationalities, Enshi 445000, China.
| | - Zong-Hai Chen
- College of Medicine, Hubei University for Nationalities, Enshi 445000, China.
| | - Kui Zhang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China.
| | - Fei-Jun Huang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
21
|
Um SW, Kim MJ, Leem JW, Bai SJ, Lee BH. Pain-Relieving Effects of mTOR Inhibitor in the Anterior Cingulate Cortex of Neuropathic Rats. Mol Neurobiol 2018; 56:2482-2494. [PMID: 30032425 PMCID: PMC6459802 DOI: 10.1007/s12035-018-1245-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/15/2018] [Indexed: 01/22/2023]
Abstract
The anterior cingulate cortex (ACC) is a well-known brain area that is associated with pain perception. Previous studies reported that the ACC has a specific role in the emotional processing of pain. Chronic pain is characterized by long-term potentiation that is induced in pain pathways and contributes to hyperalgesia caused by peripheral nerve injury. The mammalian target of rapamycin (mTOR) signaling, which is involved in synaptic protein synthesis, could be a key factor controlling long-term potentiation in neuropathic pain conditions. Until now, there have been no reports that studied the role of mTOR signaling in the ACC involved in neuropathic pain. Therefore, this study was conducted to determine the relationship of mTOR signaling in the ACC and neuropathic pain. Male Sprague-Dawley rats were subjected to cannula implantation and nerve injury under pentobarbital anesthesia. Microinjection with rapamycin into the ACC was conducted under isoflurane anesthesia on postoperative day (POD) 7. A behavioral test was performed to evaluate mechanical allodynia, and optical imaging was conducted to observe the neuronal responses of the ACC to peripheral stimulation. Inhibition of mTOR by rapamycin reduced mechanical allodynia, down-regulated mTOR signaling in the ACC, and diminished the expressions of synaptic proteins which are involved in excitatory signaling, thereby reducing neuropathic pain-induced synaptic plasticity. These results suggest that inhibiting mTOR activity by rapamycin in the ACC could serve as a new strategy for treating or managing neuropathic pain before it develops into chronic pain.
Collapse
Affiliation(s)
- Sun Woo Um
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Jee Kim
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joong Woo Leem
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sun Joon Bai
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Bae Hwan Lee
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
22
|
Uttam S, Wong C, Amorim IS, Jafarnejad SM, Tansley SN, Yang J, Prager-Khoutorsky M, Mogil JS, Gkogkas CG, Khoutorsky A. Translational profiling of dorsal root ganglia and spinal cord in a mouse model of neuropathic pain. NEUROBIOLOGY OF PAIN 2018; 4:35-44. [PMID: 30906902 PMCID: PMC6428075 DOI: 10.1016/j.ynpai.2018.04.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Translational landscape in DRG and spinal cord in SNI assay of neuropathic pain was established. ERK is a central hub of both transcriptionally and translationally controlled genes. Changes in translation efficiency and mRNA levels occur in the opposite direction for multiple mRNAs.
Acute pain serves as a protective mechanism, guiding the organism away from actual or potential tissue injury. In contrast, chronic pain is a debilitating condition without any obvious physiological function. The transition to, and the maintenance of chronic pain require new gene expression to support biochemical and structural changes within the pain pathway. The regulation of gene expression at the level of mRNA translation has emerged as an important step in the control of protein expression in the cell. Recent studies show that signaling pathways upstream of mRNA translation, such as mTORC1 and ERK, are upregulated in chronic pain conditions, and their inhibition effectively alleviates pain in several animal models. Despite this progress, mRNAs whose translation is altered in chronic pain conditions remain largely unknown. Here, we performed genome-wide translational profiling of dorsal root ganglion (DRG) and spinal cord dorsal horn tissues in a mouse model of neuropathic pain, spared nerve injury (SNI), using the ribosome profiling technique. We identified distinct subsets of mRNAs that are differentially translated in response to nerve injury in both tissues. We discovered key converging upstream regulators and pathways linked to mRNA translational control and neuropathic pain. Our data are crucial for the understanding of mechanisms by which mRNA translation promotes persistent hypersensitivity after nerve injury.
Collapse
Affiliation(s)
- Sonali Uttam
- Department of Anesthesia, McGill University, Montreal, QC H3A 0G1, Canada
| | - Calvin Wong
- Department of Anesthesia, McGill University, Montreal, QC H3A 0G1, Canada
| | - Inês S Amorim
- Patrick Wild Centre and Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Seyed Mehdi Jafarnejad
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - Shannon N Tansley
- Department of Anesthesia, McGill University, Montreal, QC H3A 0G1, Canada.,Department of Psychology, McGill University, Montreal QC H3A 1B1, Canada
| | - Jieyi Yang
- Department of Anesthesia, McGill University, Montreal, QC H3A 0G1, Canada
| | | | - Jeffrey S Mogil
- Department of Anesthesia, McGill University, Montreal, QC H3A 0G1, Canada.,Department of Psychology, McGill University, Montreal QC H3A 1B1, Canada.,Alan Edwards Centre for Research on Pain, McGill University, Montreal QC H3A 0G1, Canada
| | - Christos G Gkogkas
- Patrick Wild Centre and Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Arkady Khoutorsky
- Department of Anesthesia, McGill University, Montreal, QC H3A 0G1, Canada.,Alan Edwards Centre for Research on Pain, McGill University, Montreal QC H3A 0G1, Canada
| |
Collapse
|
23
|
Tansley SN, Wong C, Uttam S, Mogil JS, Khoutorsky A. Translation regulation in the spinal dorsal horn - A key mechanism for development of chronic pain. NEUROBIOLOGY OF PAIN 2018; 4:20-26. [PMID: 30906901 PMCID: PMC6428080 DOI: 10.1016/j.ynpai.2018.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Spinal sensitization shares molecular mechanisms with hippocampal LTP and memory. Changes in mRNA translation are observed in many chronic pain conditions. Targeting translational control mechanisms is a promising strategy to inhibit pain. Targeting spinal reconsolidation can reverse established hypersensitivity.
Chronic pain is a pathological condition characterized by long-lasting pain after damaged tissue has healed. Chronic pain can be caused and maintained by changes in various components of the pain pathway, including sensory neurons, spinal cord and higher brain centers. Exaggerated sensitivity and responsiveness of spinal nociceptive circuits, representing maladaptive plasticity, play key roles in the amplification of peripheral signals in chronic pain conditions. This spinal amplification mechanism profoundly contributes to the development and maintenance of chronic pain hypersensitivity in response to peripheral injury, and in some cases occurs independently of the peripheral stimulus. Long-lasting changes in the activity of spinal neurons are caused by alterations in their cellular proteome, which relies on de novo gene expression. Recent evidence indicates that translational control of gene expression plays a major role in determining protein levels, and is intricately involved in different forms of intrinsic and synaptic plasticity. In this review, we summarize findings supporting a key role for translational control in spinal cord-dependent mechanisms of chronic pain, and present recent approaches to reverse persistent pain by targeting these mechanisms.
Collapse
Affiliation(s)
- Shannon N Tansley
- Department of Anesthesia, McGill University, Montréal, QC H3A 0G1, Canada.,Department of Psychology, McGill University, Montréal, QC H3A 1B1, Canada
| | - Calvin Wong
- Department of Anesthesia, McGill University, Montréal, QC H3A 0G1, Canada
| | - Sonali Uttam
- Department of Anesthesia, McGill University, Montréal, QC H3A 0G1, Canada
| | - Jeffrey S Mogil
- Department of Anesthesia, McGill University, Montréal, QC H3A 0G1, Canada.,Department of Psychology, McGill University, Montréal, QC H3A 1B1, Canada.,Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC H3A 0G1, Canada
| | - Arkady Khoutorsky
- Department of Anesthesia, McGill University, Montréal, QC H3A 0G1, Canada.,Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC H3A 0G1, Canada
| |
Collapse
|
24
|
Megat S, Price TJ. Therapeutic opportunities for pain medicines via targeting of specific translation signaling mechanisms. NEUROBIOLOGY OF PAIN 2018; 4:8-19. [PMID: 30211342 PMCID: PMC6130820 DOI: 10.1016/j.ynpai.2018.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A common underlying cause of chronic pain is a phenotypic change in nociceptors in the peripheral nervous system. Translation regulation signaling pathways control gene expression changes that drive chronic pain. We focus on developments in pharmacology around translation regulation signaling that may yield new pain therapeutics.
As the population of the world ages and as more and more people survive diseases that used to be primary causes of mortality, the incidence of severe chronic pain in most of the world has risen dramatically. This type of pain is very difficult to treat and the opioid overdose epidemic that has become a leading cause of death in the United States and other parts of the world highlights the urgent need to develop new pain therapeutics. A common underlying cause of severe chronic pain is a phenotypic change in pain-sensing neurons in the peripheral nervous system called nociceptors. These neurons play a vital role in detecting potentially injurious stimuli, but when these neurons start to detect very low levels of inflammatory meditators or become spontaneously active, they send spurious pain signals to the brain that are significant drivers of chronic pain. An important question is what drives this phenotypic shift in nociceptors from quiescence under most conditions to sensitization to a broad variety of stimuli and spontaneous activity. The goal of this review is to discuss the critical role that specific translation regulation signaling pathways play in controlling gene expression changes that drive nociceptor sensitization and may underlie the development of spontaneous activity. The focus will be on advances in technologies that allow for identification of such targets and on developments in pharmacology around translation regulation signaling that may yield new pain therapeutics. A key advantage of pharmacological manipulation of these signaling events is that they may reverse phenotypic shifts in nociceptors that drive chronic pain thereby creating the first generation of disease modifying drugs for chronic pain.
Collapse
Affiliation(s)
- Salim Megat
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, USA
| | - Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, USA
| |
Collapse
|
25
|
Zhang Y, Ji F, Wang G, He D, Yang L, Zhang M. BDNF Activates mTOR to Upregulate NR2B Expression in the Rostral Anterior Cingulate Cortex Required for Inflammatory Pain-Related Aversion in Rats. Neurochem Res 2018; 43:681-691. [PMID: 29353374 DOI: 10.1007/s11064-018-2470-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/02/2018] [Accepted: 01/08/2018] [Indexed: 12/11/2022]
Abstract
The mechanistic target of rapamycin (mTOR) has been demonstrated to mediate pain-related aversion induced by formalin in the rostral anterior cingulate cortex (rACC). However, it remains unclear the signaling pathways and regulatory proteins involved. In the rACC, brain-derived neurotrophic factor (BDNF), an activity-dependent neuromodulator, has been shown to play a role in the development and persistence of chronic pain. In this study, we used a rat formalin-induced inflammatory pain model to demonstrate BDNF up-regulation in the rACC. Stimulation with exogenous BDNF up-regulated mTOR, whilst cyclotraxin B (CTX-B), a tropomyosin receptor kinase B (TrkB) antagonist, down-regulated mTOR. Our results suggest BDNF could activate an mTOR signaling pathway. Subsequently, we used formalin-induced conditioned place avoidance (F-CPA) training in rat models to investigate if mTOR activation was required for pain-related aversion. We demonstrated that BDNF/mTOR signaling could activate the NMDA receptor subunit episilon-2 (NR2B), which is required for F-CPA. Our results reveal that BDNF activates mTOR to up-regulate NR2B expression, which is required for inflammatory pain-related aversion in the rACC of rats.
Collapse
Affiliation(s)
- Yuangui Zhang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
- Department of Anesthesiology, Weifang People's Hospital, Weifang, China
| | - Fanceng Ji
- Department of Anesthesiology, Weifang People's Hospital, Weifang, China
| | - Gongming Wang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Dong He
- Shandong University, Jinan, China
| | - Le Yang
- Shandong University, Jinan, China
| | - Mengyuan Zhang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.
| |
Collapse
|
26
|
Wang L, Du J, Zhao F, Chen Z, Chang J, Qin F, Wang Z, Wang F, Chen X, Chen N. Trillium tschonoskii maxim saponin mitigates D-galactose-induced brain aging of rats through rescuing dysfunctional autophagy mediated by Rheb-mTOR signal pathway. Biomed Pharmacother 2017; 98:516-522. [PMID: 29287199 DOI: 10.1016/j.biopha.2017.12.046] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 12/04/2017] [Accepted: 12/13/2017] [Indexed: 12/22/2022] Open
Abstract
During the expansion of aging population, the study correlated with brain aging is one of the important research topics. Developing novel and effective strategies for delaying brain aging is highly desired. Brain aging is characteristics of impaired cognitive capacity due to dysfunctional autophagy regulated by Rheb-mTOR signal pathway in hippocampal tissues. In the present study, we have established a rat model with brain aging through subcutaneous injection of D-galactose (D-gal). Upon the intervention of Trillium tschonoskii Maxim (TTM) saponin, one of bioactive components from local natural herbs in China, the learning and memory capacity of D-gal-induced aging rats was evaluated through Morris water maze test, and the regulation of Rheb-mTOR signal pathway and functional status of autophagy in hippocampal tissues of D-gal-induced aging rats was explored by Western blot. TTM saponin revealed an obvious function to improve learning and memory capacity of D-gal-induced aging rats through up-regulating Rheb and down-regulating mTOR, thereby rescuing dysfunctional autophagy to execute anti-aging role. Meanwhile, this study confirmed the function of TTM saponin for preventing and treating brain aging, and provided a reference for the development and utilization of natural products in health promotion and aging-associated disease treatment.
Collapse
Affiliation(s)
- Lingjie Wang
- College of Medicine, Hubei University for Nationalities, Enshi, 445000, China
| | - Junlong Du
- College of Medicine, Hubei University for Nationalities, Enshi, 445000, China
| | - Fangyu Zhao
- College of Medicine, Hubei University for Nationalities, Enshi, 445000, China
| | - Zonghai Chen
- College of Medicine, Hubei University for Nationalities, Enshi, 445000, China
| | - Jingru Chang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, College of Health Science, Wuhan Sports University, Wuhan, 430079, China
| | - Furong Qin
- College of Medicine, Hubei University for Nationalities, Enshi, 445000, China
| | - Zili Wang
- College of Medicine, Hubei University for Nationalities, Enshi, 445000, China
| | - Fengjie Wang
- College of Medicine, Hubei University for Nationalities, Enshi, 445000, China
| | - Xianbing Chen
- College of Medicine, Hubei University for Nationalities, Enshi, 445000, China; Key Laboratory of Biological Resource Prevention and Utilization of Hubei Province, Enshi, 445000, China.
| | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, College of Health Science, Wuhan Sports University, Wuhan, 430079, China.
| |
Collapse
|
27
|
Price TJ, Das V, Dussor G. Adenosine Monophosphate-activated Protein Kinase (AMPK) Activators For the Prevention, Treatment and Potential Reversal of Pathological Pain. Curr Drug Targets 2017; 17:908-20. [PMID: 26521775 DOI: 10.2174/1389450116666151102095046] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 10/20/2015] [Accepted: 10/29/2015] [Indexed: 12/23/2022]
Abstract
Pathological pain is an enormous medical problem that places a significant burden on patients and can result from an injury that has long since healed or be due to an unidentifiable cause. Although treatments exist, they often either lack efficacy or have intolerable side effects. More importantly, they do not reverse the changes in the nervous system mediating pathological pain, and thus symptoms often return when therapies are discontinued. Consequently, novel therapies are urgently needed that have both improved efficacy and disease-modifying properties. Here we highlight an emerging target for novel pain therapies, adenosine monophosphate-activated protein kinase (AMPK). AMPK is capable of regulating a variety of cellular processes including protein translation, activity of other kinases, and mitochondrial metabolism, many of which are thought to contribute to pathological pain. Consistent with these properties, preclinical studies show positive, and in some cases disease-modifying effects of either pharmacological activation or genetic regulation of AMPK in models of nerve injury, chemotherapy-induced peripheral neuropathy (CIPN), postsurgical pain, inflammatory pain, and diabetic neuropathy. Given the AMPK-activating ability of metformin, a widely prescribed and well-tolerated drug, these preclinical studies provide a strong rationale for both retrospective and prospective human pain trials with this drug. They also argue for the development of novel AMPK activators, whether orthosteric, allosteric, or modulators of events upstream of the kinase. Together, this review will present the case for AMPK as a novel therapeutic target for pain and will discuss future challenges in the path toward development of AMPK-based pain therapeutics.
Collapse
Affiliation(s)
- Theodore J Price
- School of Behavioral and Brain Sciences, University of Texas at Dallas, JO 4.212 800 W Campbell Rd, Richardson TX 75080, USA.
| | | | | |
Collapse
|
28
|
Pecikoza UB, Tomić MA, Micov AM, Stepanović-Petrović RM. Metformin Synergizes With Conventional and Adjuvant Analgesic Drugs to Reduce Inflammatory Hyperalgesia in Rats. Anesth Analg 2017; 124:1317-1329. [PMID: 27669556 DOI: 10.1213/ane.0000000000001561] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Metformin is a widely used and safe antidiabetic drug that has recently been shown to possess analgesic properties in models of inflammatory pain. Because various arthritic inflammatory disorders are highly prevalent in diabetic patients, we aimed to examine the type of interaction between metformin and several conventional and adjuvant analgesic drugs (ibuprofen, aspirin, tramadol, and pregabalin) in a rat model of somatic inflammatory hyperalgesia. METHODS Inflammation of the rat hind paw was induced by an intraplantar injection of carrageenan (0.1 mL, 1%). The antihyperalgesic effects of metformin (intraperitoneally), analgesics (orally or intraperitoneally), and 2-drug metformin-analgesic combinations were assessed with an electronic Von Frey anesthesiometer, by measuring the change in paw withdrawal thresholds induced by carrageenan (n = 6 rats in drug/drug combination-treated groups). First, we determined the doses of individual drugs needed to produce an antihyperalgesic effect of 50% (ED50 values). In combination experiments, drugs were coadministered in fixed-dose fractions (1/16, 1/8, 1/4, and 1/2) of their individual ED50 values and the type of interaction between components was determined by isobolographic analysis. RESULTS Metformin (50-200 mg/kg) significantly and dose-dependently reduced carrageenan-induced hyperalgesia with a maximal antihyperalgesic effect (mean ± SEM) of 62 ± 6% (all P ≤ .024). Ibuprofen (25-150 mg/kg), aspirin (100-400 mg/kg), tramadol (0.5-5 mg/kg), and pregabalin (2.5-20 mg/kg) also produced significant and dose-dependent antihyperalgesic effects (all P ≤ .042) of similar magnitude to metformin (the maximal antihyperalgesic effects were 73 ± 4% for ibuprofen, 62 ± 4.2% for aspirin, 69 ± 5.9% for tramadol, and 56 ± 3.9% for pregabalin). In combination experiments, administration of 2-drug metformin-analgesic combinations led to a significant and dose-dependent reduction of carrageenan-induced hyperalgesia (all P ≤ .027). The isobolographic analysis revealed that metformin interacted synergistically with the examined analgesics (experimental ED50 values of 2-drug combinations were significantly lower than theoretical additive ED50 values; all P < .05) and that there was a similar, approximately 5-fold, reduction of doses of both drugs in all tested combinations. CONCLUSIONS Our results suggest that in patients who are already receiving metformin therapy, lower doses of ibuprofen/aspirin/tramadol/pregabalin might be sufficient for achieving satisfactory pain relief. Metformin-aspirin combination might be particularly useful because it may achieve multiple therapeutic goals (glucoregulation, pain relief, and cardioprotection).
Collapse
Affiliation(s)
- Uroš B Pecikoza
- From the Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | | | | | | |
Collapse
|
29
|
Kwon M, Han J, Kim UJ, Cha M, Um SW, Bai SJ, Hong SK, Lee BH. Inhibition of Mammalian Target of Rapamycin (mTOR) Signaling in the Insular Cortex Alleviates Neuropathic Pain after Peripheral Nerve Injury. Front Mol Neurosci 2017; 10:79. [PMID: 28377693 PMCID: PMC5359287 DOI: 10.3389/fnmol.2017.00079] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/06/2017] [Indexed: 12/30/2022] Open
Abstract
Injury of peripheral nerves can trigger neuropathic pain, producing allodynia and hyperalgesia via peripheral and central sensitization. Recent studies have focused on the role of the insular cortex (IC) in neuropathic pain. Because the IC is thought to store pain-related memories, translational regulation in this structure may reveal novel targets for controlling chronic pain. Signaling via mammalian target of rapamycin (mTOR), which is known to control mRNA translation and influence synaptic plasticity, has been studied at the spinal level in neuropathic pain, but its role in the IC under these conditions remains elusive. Therefore, this study was conducted to determine the role of mTOR signaling in neuropathic pain and to assess the potential therapeutic effects of rapamycin, an inhibitor of mTORC1, in the IC of rats with neuropathic pain. Mechanical allodynia was assessed in adult male Sprague-Dawley rats after neuropathic surgery and following microinjections of rapamycin into the IC on postoperative days (PODs) 3 and 7. Optical recording was conducted to observe the neural responses of the IC to peripheral stimulation. Rapamycin reduced mechanical allodynia and downregulated the expression of postsynaptic density protein 95 (PSD95), decreased neural excitability in the IC, thereby inhibiting neuropathic pain-induced synaptic plasticity. These findings suggest that mTOR signaling in the IC may be a critical molecular mechanism modulating neuropathic pain.
Collapse
Affiliation(s)
- Minjee Kwon
- Department of Physiology, Yonsei University College of MedicineSeoul, South Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of MedicineSeoul, South Korea
| | - Jeongsoo Han
- Department of Physiology, Yonsei University College of MedicineSeoul, South Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of MedicineSeoul, South Korea
| | - Un Jeng Kim
- Department of Physiology, Yonsei University College of Medicine Seoul, South Korea
| | - Myeounghoon Cha
- Department of Physiology, Yonsei University College of Medicine Seoul, South Korea
| | - Sun Woo Um
- Department of Physiology, Yonsei University College of MedicineSeoul, South Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of MedicineSeoul, South Korea
| | - Sun Joon Bai
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine Seoul, South Korea
| | - Seong-Karp Hong
- Division of Bio and Health Sciences, Mokwon University Daejeon, South Korea
| | - Bae Hwan Lee
- Department of Physiology, Yonsei University College of MedicineSeoul, South Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of MedicineSeoul, South Korea; Brain Research Institute and Epilepsy Research Institute, Yonsei University College of MedicineSeoul, South Korea
| |
Collapse
|
30
|
Li S, Han J, Wang DS, Yang Q, Feng B, Kang WB, Yang L, Liu G, Zhao MG. Sinomenine attenuates chronic inflammatory pain in mice. Metab Brain Dis 2017; 32:211-219. [PMID: 27585465 DOI: 10.1007/s11011-016-9889-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 08/01/2016] [Indexed: 01/27/2023]
Abstract
Sinomenine, an alkaloid originally isolated from the roots of Sinomeniumacutum, is used as a traditional Chinese medicine for rheumatic arthritis. However, little is known about the neuronal mechanisms underlying the analgesic effects of sinomenine in animals with chronic inflammatory pain. In this study, we investigated the persistent inflammatory pain induced by hind paw injection of complete Freund's adjuvant (CFA) in mice, which was reversed by sinomenine administration. In the anterior cingulate cortex (ACC), a region highly associated with chronic pain processing, the upregulation of GluN2B-containing N-methyl-D-aspartate (NMDA) receptors and Ca2+/calmodulin-dependent protein kinase II, total levels of GluA1, and phosphorylation of GluA1 at Ser831 (p-GluA1-Ser831) were reversed by systemically administrating sinomenine. Furthermore, sinomenine treatment downregulated the mammalian target of rapamycin (mTOR) pathway. Increases in p-mTOR, p-p70S6k, p-S6, and p-4EBP, which were induced by chronic inflammation, were all changed. However, sinomenine did not affect the levels of GluN2A-containing NMDA receptors and p-GluA1-Ser845, as well as the total levels of mTOR, p70S6k, S6, and 4EBP. In conclusion, results indicated that sinomenine reduced the chronic inflammatory pain induced by CFA, at least partially by regulating the GluN2B receptors and mTOR signals in the ACC.
Collapse
Affiliation(s)
- Shuo Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Jing Han
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Dong-Sheng Wang
- Jinling Hospital, Department of Orthopedics, Nanjing University, School of Medicine, Nanjing, 210002, China
| | - Qi Yang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Bin Feng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Wen-Bo Kang
- Jinling Hospital, Department of Orthopedics, Nanjing University, School of Medicine, Nanjing, 210002, China
| | - Le Yang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Gang Liu
- Jinling Hospital, Department of Orthopedics, Nanjing University, School of Medicine, Nanjing, 210002, China.
| | - Ming-Gao Zhao
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
31
|
Klingebiel M, Dinekov M, Köhler C. Analysis of ribosomal protein S6 baseline phosphorylation and effect of tau pathology in the murine brain and human hippocampus. Brain Res 2017; 1659:121-135. [PMID: 28119058 DOI: 10.1016/j.brainres.2017.01.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 01/17/2023]
Abstract
We examined the distribution pattern of the phosphorylated 40S ribosomal subunit protein S6, a downstream target of the mTOR pathway, in the brains of 24-months-old human tau transgenic pR5 mice, non-transgenic littermates and in human hippocampi. We studied baseline levels of phosphorylated S6 and a possible effect of tau pathology. S6 phosphorylated at Ser235/236 (pS6Ser235/236) or Ser240/244 (pS6Ser240/244) has been used as a read-out of mTOR activity in several studies. The mTOR pathway regulates a wide variety of cellular functions including cell growth, ribosome biosynthesis, translational control and autophagy. Its dysregulation might underlie the neurodegenerative pathology of Alzheimer's disease and other tauopathies. pS6Ser235/236 and pS6Ser240/244 immunoreactivity in the mouse brain were widespread and similar distributed, but intensive pS6Ser235/236 immunoreactivity was more selective, especially highlighting certain brainstem regions. In the human hippocampus mainly granulovacuolar inclusions in neurons displayed pS6Ser235/236 immunoreactivity. In contrast, a considerable number of neurons displayed pS6Ser240/244 immunoreactivity in the cytoplasm without labeling of granulovacuolar inclusions. Except for a tendency of lower numbers of intensely phosphorylated S6-positive neurons in pR5 mice, the pattern of distribution of pS6Ser235/236 and pS6Ser240/244 immunoreactivity was largely unchanged when compared with non-transgenic mice and also when human hippocampi from AD cases and controls were compared. Similar to pR5 mice most neurons with hyper-phosphorylated tau in human hippocampi displayed no or only weak labeling for phosphorylated S6, suggesting that phosphorylated S6 is not especially associated with pathological tau, but is rather a feature of unaffected neurons.
Collapse
Affiliation(s)
- Maria Klingebiel
- Institute II for Anatomy, Medical Faculty, University of Cologne, Kerpener Str. 62, 50924 Cologne, Germany
| | - Maja Dinekov
- Institute II for Anatomy, Medical Faculty, University of Cologne, Kerpener Str. 62, 50924 Cologne, Germany
| | - Christoph Köhler
- Institute II for Anatomy, Medical Faculty, University of Cologne, Kerpener Str. 62, 50924 Cologne, Germany.
| |
Collapse
|
32
|
Ko MH, Yang ML, Youn SC, Lan CT, Tseng TJ. Intact subepidermal nerve fibers mediate mechanical hypersensitivity via the activation of protein kinase C gamma in spared nerve injury. Mol Pain 2016; 12:12/0/1744806916656189. [PMID: 27296621 PMCID: PMC4956387 DOI: 10.1177/1744806916656189] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Spared nerve injury is an important neuropathic pain model for investigating the role of intact primary afferents in the skin on pain hypersensitivity. However, potential cellular mechanisms remain poorly understood. In phosphoinositide-3 kinase pathway, pyruvate dehydrogenase kinase 1 (PDK1) participates in the regulation of neuronal plasticity for central sensitization. The downstream cascades of PDK1 include: (1) protein kinase C gamma (PKCg) controls the trafficking and phosphorylation of ionotropic glutamate receptor; (2) protein kinase B (Akt)/the mammalian target of rapamycin (mTOR) signaling is responsible for local protein synthesis. Under these statements, we therefore hypothesized that an increase of PKCg activation and mTOR-dependent PKCg synthesis in intact primary afferents after SNI might contribute to pain hypersensitivity. RESULTS The variants of spared nerve injury were performed in Sprague-Dawley rats by transecting any two of the three branches of the sciatic nerve, leaving only one branch intact. Following SNIt (spared tibial branch), mechanical hyperalgesia and mechanical allodynia, but not thermal hyperalgesia, were significantly induced. In the first footpad, normal epidermal innervations were verified by the protein gene product 9.5 (PGP9.5)- and growth-associated protein 43 (GAP43)-immunoreactive (IR) intraepidermal nerve fibers (IENFs) densities. Furthermore, the rapid increases of phospho-PKCg- and phosphomTOR-IR subepidermal nerve fibers (SENFs) areas were distinct gathered from the results of PGP9.5-, GAP43-, and neurofilament 200 (NF200)-IR SENFs areas. The efficacy of PKC inhibitor (GF 109203X) or mTOR complex 1 inhibitor (rapamycin) for attenuating mechanical hyperalgesia and mechanical allodynia by intraplantar injection was dose-dependent. CONCLUSIONS From results obtained in this study, we strongly recommend that the intact SENFs persistently increase PKCg activation and mTOR-dependent PKCg synthesis participate in the initiation and maintenance of mechanical hypersensitivity in spared nerve injury, which represents as a novel insight into the therapeutic strategy of pain in the periphery.
Collapse
Affiliation(s)
- Miau-Hwa Ko
- Department of Anatomy, College of Medicine, China Medical University, Taichung, Taiwan
| | - Ming-Ling Yang
- Department of Anatomy, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Su-Chung Youn
- Department of Anatomy, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chyn-Tair Lan
- Department of Anatomy, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung, Taiwan
- Chyn-Tair Lan, Department of Anatomy, School of Medicine, Chung Shan Medical University, No. 110, Jian-Guo N. R., Sec. 1, Taichung 40201, Taiwan. To-Jung Tseng, Department of Anatomy, School of Medicine, Chung Shan Medical University, No. 110, Jian-Guo N. R., Sec. 1, Taichung 40201, Taiwan.
| | - To-Jung Tseng
- Department of Anatomy, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
33
|
Lu B, Jiang J, Sun J, Xiao C, Meng B, Zheng J, Li X, Wang R, Wu G, Chen J. Inhibition of mammalian target of rapamycin activation in the rostral anterior cingulate cortex attenuates pain-related aversion in rats. Behav Brain Res 2016; 310:51-8. [PMID: 27163752 DOI: 10.1016/j.bbr.2016.05.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 04/12/2016] [Accepted: 05/04/2016] [Indexed: 10/21/2022]
Abstract
Pain is a complex experience that comprises both sensory and affective dimensions. Mammalian target of rapamycin (mTOR) plays an important role in the modulation of neuronal plasticity associated with the pathogenesis of pain sensation. However, the role of mTOR in pain affect is unclear. Using a formalin-induced conditioned place avoidance (F-CPA) test, the current study investigated the effects of the mTOR specific inhibitor rapamycin on noxious stimulation induced aversion in the rostral anterior cingulate cortex (rACC). Intraplantar injection of 5% formalin was associated with significant activation of mTOR, as well as p70 ribosomal S6 protein (p70S6K), its downstream effector, in the rACC. The inhibition of mTOR activation with rapamycin disrupted pain-related aversion; however, this inhibition did not affect formalin-induced spontaneous nociceptive behaviors in rats. These findings demonstrated for the first time that mTOR and its downstream pathway in the rACC contribute to the induction of pain-related negative emotion.
Collapse
Affiliation(s)
- Bo Lu
- Department of Anesthesia, Ningbo NO. 2 Hospital, Ningbo 315010, PR China
| | - Jingyan Jiang
- Department of Anesthesia, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo 315040, PR China
| | - Jianliang Sun
- Department of Anesthesia, Hangzhou Hospital Affiliated to Nanjing Medical University, Hangzhou First People's Hospital, Hangzhou 310006, PR China
| | - Chun Xiao
- Department of Anesthesia, Hangzhou Hospital Affiliated to Nanjing Medical University, Hangzhou First People's Hospital, Hangzhou 310006, PR China
| | - Bo Meng
- Department of Anesthesia, Ningbo NO. 2 Hospital, Ningbo 315010, PR China
| | - Jinwei Zheng
- Department of Anesthesia, Ningbo NO. 2 Hospital, Ningbo 315010, PR China
| | - Xiaoyu Li
- Department of Anesthesia, Ningbo NO. 2 Hospital, Ningbo 315010, PR China
| | - Ruichun Wang
- Department of Anesthesia, Ningbo NO. 2 Hospital, Ningbo 315010, PR China
| | - Guorong Wu
- Department of Anesthesia, Ningbo NO. 2 Hospital, Ningbo 315010, PR China
| | - Junping Chen
- Department of Anesthesia, Ningbo NO. 2 Hospital, Ningbo 315010, PR China.
| |
Collapse
|
34
|
He WY, Zhang B, Xiong QM, Yang CX, Zhao WC, He J, Zhou J, Wang HB. Intrathecal administration of rapamycin inhibits the phosphorylation of DRG Nav1.8 and attenuates STZ-induced painful diabetic neuropathy in rats. Neurosci Lett 2016; 619:21-8. [DOI: 10.1016/j.neulet.2016.02.064] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 02/27/2016] [Accepted: 02/29/2016] [Indexed: 01/19/2023]
|
35
|
Abstract
Chronic pain is a major clinical problem that is poorly treated with available therapeutics. Adenosine monophosphate-activated protein kinase (AMPK) has recently emerged as a novel target for the treatment of pain with the exciting potential for disease modification. AMPK activators inhibit signaling pathways that are known to promote changes in the function and phenotype of peripheral nociceptive neurons and promote chronic pain. AMPK activators also reduce the excitability of these cells suggesting that AMPK activators may be efficacious for the treatment of chronic pain disorders, like neuropathic pain, where changes in the excitability of nociceptors is thought to be an underlying cause. In agreement with this, AMPK activators have now been shown to alleviate pain in a broad variety of preclinical pain models indicating that this mechanism might be engaged for the treatment of many types of pain in the clinic. A key feature of the effect of AMPK activators in these models is that they can lead to a long-lasting reversal of pain hypersensitivity even long after treatment cessation, indicative of disease modification. Here, we review the evidence supporting AMPK as a novel pain target pointing out opportunities for further discovery that are likely to have an impact on drug discovery efforts centered around potent and specific allosteric activators of AMPK for chronic pain treatment.
Collapse
|
36
|
Khoutorsky A, Bonin RP, Sorge RE, Gkogkas CG, Pawlowski SA, Jafarnejad SM, Pitcher MH, Alain T, Perez-Sanchez J, Salter EW, Martin L, Ribeiro-da-Silva A, De Koninck Y, Cervero F, Mogil JS, Sonenberg N. Translational control of nociception via 4E-binding protein 1. eLife 2015; 4. [PMID: 26678009 PMCID: PMC4695384 DOI: 10.7554/elife.12002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 11/01/2015] [Indexed: 12/31/2022] Open
Abstract
Activation of the mechanistic/mammalian target of rapamycin (mTOR) kinase in models of acute and chronic pain is strongly implicated in mediating enhanced translation and hyperalgesia. However, the molecular mechanisms by which mTOR regulates nociception remain unclear. Here we show that deletion of the eukaryotic initiation factor 4E-binding protein 1 (4E-BP1), a major mTOR downstream effector, which represses eIF4E activity and cap-dependent translation, leads to mechanical, but not thermal pain hypersensitivity. Mice lacking 4E-BP1 exhibit enhanced spinal cord expression of neuroligin 1, a cell-adhesion postsynaptic protein regulating excitatory synapse function, and show increased excitatory synaptic input into spinal neurons, and a lowered threshold for induction of synaptic potentiation. Pharmacological inhibition of eIF4E or genetic reduction of neuroligin 1 levels normalizes the increased excitatory synaptic activity and reverses mechanical hypersensitivity. Thus, translational control by 4E-BP1 downstream of mTOR effects the expression of neuroligin 1 and excitatory synaptic transmission in the spinal cord, and thereby contributes to enhanced mechanical nociception. DOI:http://dx.doi.org/10.7554/eLife.12002.001 Despite the unpleasant feeling it causes, pain is necessary for survival as it helps individuals to avoid objects, environments and situations that cause damage to their body. However, millions of people experience long-lasting “chronic” pain, or are hypersensitive to pain. There are few treatments available for these conditions, but these treatments do not work well for the majority of patients, and can have serious side effects. To develop new treatments, researchers must first better understand how chronic pain develops. Pain is transmitted to the brain in the form of electrical signals “fired” along nerve fibers. Different nerves transmit information about different types of pain: for example, pain caused by a sharp object pressed against the skin activates a different set of neurons to those activated when touching something dangerously hot. Studies in mice have suggested that a protein called mTOR that is found inside neurons is important for them to fire pain signals. However, it is not clear exactly how mTOR contributes to pain signaling, although it is known to affect the activities of several other proteins in neurons. One protein that mTOR affects the activity of is called 4E-BP1. Now, Khoutorsky, Bonin, Sorge et al. show that mice that lack 4E-BP1 behave in ways that suggest they are hypersensitive to poking or pinching sensations. However, the mice did not show hypersensitivity when they touched a hot surface. Further investigation revealed that the neurons in the spinal cord of mice that lack 4E-BP1 produce abnormally high amounts of a molecule called neuroligin 1, which makes the neurons more likely to fire and thus signal pain. Khoutorsky, Bonin, Sorge et al. found that treating mice that lack 4E-BP1 with a compound that reduces neuroligin 1 production causes their neurons to fire more normally. This also reduces the animals’ apparent signs of hypersensitivity to pressure on their skin. It will be important in future studies to identify additional targets of 4E-BP1 in the spinal cord that could contribute to increased mechanical sensation, and also to study the role of 4E-BP1 in peripheral nerves. DOI:http://dx.doi.org/10.7554/eLife.12002.002
Collapse
Affiliation(s)
- Arkady Khoutorsky
- Department of Biochemistry, McGill University, Montréal, Canada.,Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Canada
| | - Robert P Bonin
- Unité de neurosciences cellulaires et moléculaire, Institut universitaire en santé mentale de Québec, Québec, Canada
| | - Robert E Sorge
- Department of Psychology, McGill University, Montréal, Canada.,Alan Edwards Centre for Research on Pain, McGill University, Montréal, Canada
| | - Christos G Gkogkas
- Department of Biochemistry, McGill University, Montréal, Canada.,Centre for Integrative Physiology and The Patrick Wild Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Sophie Anne Pawlowski
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Seyed Mehdi Jafarnejad
- Department of Biochemistry, McGill University, Montréal, Canada.,Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Canada
| | - Mark H Pitcher
- National Center for Complementary and Alternative Medicine, National Institutes of Health, Porter Neuroscience Research Center, Maryland, United States
| | - Tommy Alain
- Department of Biochemistry, McGill University, Montréal, Canada.,Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Canada
| | - Jimena Perez-Sanchez
- Unité de neurosciences cellulaires et moléculaire, Institut universitaire en santé mentale de Québec, Québec, Canada
| | - Eric W Salter
- Unité de neurosciences cellulaires et moléculaire, Institut universitaire en santé mentale de Québec, Québec, Canada
| | - Loren Martin
- Department of Psychology, McGill University, Montréal, Canada.,Alan Edwards Centre for Research on Pain, McGill University, Montréal, Canada
| | - Alfredo Ribeiro-da-Silva
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Yves De Koninck
- Unité de neurosciences cellulaires et moléculaire, Institut universitaire en santé mentale de Québec, Québec, Canada.,Alan Edwards Centre for Research on Pain, McGill University, Montréal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Québec, Canada
| | - Fernando Cervero
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, Canada.,Anesthesia Research Unit, McGill University, Montreal, Canada
| | - Jeffrey S Mogil
- Department of Psychology, McGill University, Montréal, Canada.,Alan Edwards Centre for Research on Pain, McGill University, Montréal, Canada
| | - Nahum Sonenberg
- Department of Biochemistry, McGill University, Montréal, Canada.,Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Canada
| |
Collapse
|
37
|
Biever A, Valjent E, Puighermanal E. Ribosomal Protein S6 Phosphorylation in the Nervous System: From Regulation to Function. Front Mol Neurosci 2015; 8:75. [PMID: 26733799 PMCID: PMC4679984 DOI: 10.3389/fnmol.2015.00075] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/23/2015] [Indexed: 01/31/2023] Open
Abstract
Since the discovery of the phosphorylation of the 40S ribosomal protein S6 (rpS6) about four decades ago, much effort has been made to uncover the molecular mechanisms underlying the regulation of this post-translational modification. In the field of neuroscience, rpS6 phosphorylation is commonly used as a readout of the mammalian target of rapamycin complex 1 signaling activation or as a marker for neuronal activity. Nevertheless, its biological role in neurons still remains puzzling. Here we review the pharmacological and physiological stimuli regulating this modification in the nervous system as well as the pathways that transduce these signals into rpS6 phosphorylation. Altered rpS6 phosphorylation observed in various genetic and pathophysiological mouse models is also discussed. Finally, we examine the current state of knowledge on the physiological role of this post-translational modification and highlight the questions that remain to be addressed.
Collapse
Affiliation(s)
- Anne Biever
- Centre National de la Recherche Scientifique, UMR5203, Institut de Génomique FonctionnelleMontpellier, France; Institut National de la Santé et de la Recherche Médicale, U1191Montpellier, France; Université de Montpellier, UMR-5203Montpellier, France
| | - Emmanuel Valjent
- Centre National de la Recherche Scientifique, UMR5203, Institut de Génomique FonctionnelleMontpellier, France; Institut National de la Santé et de la Recherche Médicale, U1191Montpellier, France; Université de Montpellier, UMR-5203Montpellier, France
| | - Emma Puighermanal
- Centre National de la Recherche Scientifique, UMR5203, Institut de Génomique FonctionnelleMontpellier, France; Institut National de la Santé et de la Recherche Médicale, U1191Montpellier, France; Université de Montpellier, UMR-5203Montpellier, France
| |
Collapse
|
38
|
Activation of the Mammalian Target of Rapamycin in the Rostral Ventromedial Medulla Contributes to the Maintenance of Nerve Injury-Induced Neuropathic Pain in Rat. Neural Plast 2015; 2015:394820. [PMID: 26770837 PMCID: PMC4684879 DOI: 10.1155/2015/394820] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/06/2015] [Indexed: 11/20/2022] Open
Abstract
The mammalian target of rapamycin (mTOR), a serine-threonine protein kinase, integrates extracellular signals, thereby modulating several physiological and pathological processes, including pain. Previous studies have suggested that rapamycin (an mTOR inhibitor) can attenuate nociceptive behaviors in many pain models, most likely at the spinal cord level. However, the mechanisms of mTOR at the supraspinal level, particularly at the level of the rostral ventromedial medulla (RVM), remain unclear. Thus, the aim of this study was to elucidate the role of mTOR in the RVM, a key relay region for the descending pain control pathway, under neuropathic pain conditions. Phosphorylated mTOR was mainly expressed in serotonergic spinally projecting neurons and was significantly increased in the RVM after spared nerve injury- (SNI-) induced neuropathic pain. Moreover, in SNI rat brain slices, rapamycin infusion both decreased the amplitude instead of the frequency of spontaneous excitatory postsynaptic currents and reduced the numbers of action potentials in serotonergic neurons. Finally, intra-RVM microinjection of rapamycin effectively alleviated established mechanical allodynia but failed to affect the development of neuropathic pain. In conclusion, our data provide strong evidence for the role of mTOR in the RVM in nerve injury-induced neuropathic pain, indicating a novel mechanism of mTOR inhibitor-induced analgesia.
Collapse
|
39
|
Izumi Y, Sasaki M, Hashimoto S, Sawa T, Amaya F. mTOR signaling controls VGLUT2 expression to maintain pain hypersensitivity after tissue injury. Neuroscience 2015; 308:169-79. [DOI: 10.1016/j.neuroscience.2015.09.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/16/2015] [Accepted: 09/03/2015] [Indexed: 01/09/2023]
|
40
|
Abdelaziz DM, Stone LS, Komarova SV. Localized experimental bone metastasis drives osteolysis and sensory hypersensitivity at distant non-tumor-bearing sites. Breast Cancer Res Treat 2015. [PMID: 26208488 DOI: 10.1007/s10549-015-3517-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Patients with breast cancer metastasis to bone suffer from inadequate pain relief. Animal models provide increased understanding of cancer-induced bone and sensory alterations. The objective of this study was to investigate the measures of pain at distant non-tumor-bearing sites in animals with localized bone metastasis. Immunocompetent BALB/c mice are injected intra-tibially with murine mammary carcinoma cells (4T1) or saline, and the sensitivity to mechanical and thermal stimuli in the contralateral paw was examined. In addition to previously demonstrated development of osteolysis and hypersensitivity to mechanical and thermal stimuli in the cancer-injected tibia, these animals exhibited an increase in sensory hypersensitivity in the contralateral limb. No bone lesions were evident on radiographs of the contralateral limbs. Histomorphometry detected decreased bone volume per tissue volume and increased osteoclast number in the contralateral tibia and vertebral bones of cancer-bearing animals. Neuroplasticity was examined by immunofluorescence for calcitonin gene-related peptide (CGRP) in sensory neurons and glial fibrillary acidic protein (GFAP) in lumbar spinal cords. CGRP-immunoreactivity and GFAP-immunoreactivity were significantly elevated both ipsilateral and contralateral in tumor-bearing animals. The anti-inflammatory and osteolysis-targeting drug rapamycin reduced hypersensitivity to mechanical and cold stimuli, attenuated GFAP over-expression, and lowered osteoclast number. The osteoclast-targeting drug pamidronate reduced sensitivity to cold and protected against bone loss. Localized bone cancer drives hypersensitivity, bone remodeling, and sensory neuron plasticity at sites distant from the primary tumor area. Drugs targeting these mechanisms may be useful in the treatment of pain distant from the primary tumor site.
Collapse
|
41
|
Lutz BM, Nia S, Xiong M, Tao YX, Bekker A. mTOR, a new potential target for chronic pain and opioid-induced tolerance and hyperalgesia. Mol Pain 2015; 11:32. [PMID: 26024835 PMCID: PMC4455918 DOI: 10.1186/s12990-015-0030-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 05/21/2015] [Indexed: 01/25/2023] Open
Abstract
Chronic pain is a major public health problem with limited treatment options. Opioids remain a routine treatment for chronic pain, but extended exposure to opioid therapy can produce opioid tolerance and hyperalgesia. Although the mechanisms underlying chronic pain, opioid-induced tolerance, and opioid-induced hyperalgesia remain to be uncovered, mammalian target of rapamycin (mTOR) is involved in these disorders. The mTOR complex 1 and its triggered protein translation are required for the initiation and maintenance of chronic pain (including cancer pain) and opioid-induced tolerance/hyperalgesia. Given that mTOR inhibitors are FDA-approved drugs and an mTOR inhibitor is approved for the treatment of several cancers, these findings suggest that mTOR inhibitors will likely have multiple clinical benefits, including anticancer, antinociception/anti-cancer pain, and antitolerance/hyperalgesia. This paper compares the role of mTOR complex 1 in chronic pain, opioid-induced tolerance, and opioid-induced hyperalgesia.
Collapse
Affiliation(s)
- Brianna Marie Lutz
- Rutgers Graduate School of Biomedical Sciences, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA.,Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Sam Nia
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Ming Xiong
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA.
| | - Alex Bekker
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA.
| |
Collapse
|
42
|
Lv J, Li Z, She S, Xu L, Ying Y. Effects of intrathecal injection of rapamycin on pain threshold and spinal cord glial activation in rats with neuropathic pain. Neurol Res 2015; 37:739-43. [DOI: 10.1179/1743132815y.0000000052] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
43
|
Huang SY, Sung CS, Chen WF, Chen CH, Feng CW, Yang SN, Hung HC, Chen NF, Lin PR, Chen SC, Wang HMD, Chu TH, Tai MH, Wen ZH. Involvement of phosphatase and tensin homolog deleted from chromosome 10 in rodent model of neuropathic pain. J Neuroinflammation 2015; 12:59. [PMID: 25889774 PMCID: PMC4386079 DOI: 10.1186/s12974-015-0280-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 03/07/2015] [Indexed: 12/30/2022] Open
Abstract
Background Many cancer research studies have extensively examined the phosphatase and tensin homolog deleted from chromosome 10 (PTEN) pathway. There are only few reports that suggest that PTEN might affect pain; however, there is still a lack of evidence to show the role of PTEN for modulating pain. Here, we report a role for PTEN in a rodent model of neuropathic pain. Results We found that chronic constriction injury (CCI) surgery in rats could elicit downregulation of spinal PTEN as well as upregulation of phosphorylated PTEN (phospho-PTEN) and phosphorylated mammalian target of rapamycin (phospho-mTOR). After examining such changes in endogenous PTEN in neuropathic rats, we explored the effects of modulating the spinal PTEN pathway on nociceptive behaviors. The normal rats exhibited mechanical allodynia after intrathecal (i.t.) injection of adenovirus-mediated PTEN antisense oligonucleotide (Ad-antisense PTEN). These data indicate the importance of downregulation of spinal PTEN for nociception. Moreover, upregulation of spinal PTEN by i.t. adenovirus-mediated PTEN (Ad-PTEN) significantly prevented CCI-induced development of nociceptive sensitization, thermal hyperalgesia, mechanical allodynia, cold allodynia, and weight-bearing deficits in neuropathic rats. Furthermore, upregulation of spinal PTEN by i.t. Ad-PTEN significantly attenuated CCI-induced microglia and astrocyte activation, upregulation of tumor necrosis factor-α (TNF-α) and phospho-mTOR, and downregulation of PTEN in neuropathic rats 14 days post injury. Conclusions These findings demonstrate that PTEN plays a key, beneficial role in a rodent model of neuropathic pain.
Collapse
Affiliation(s)
- Shi-Ying Huang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Center for Neuroscience, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan.
| | - Chun-Sung Sung
- Department of Anesthesiology, Taipei Veterans General Hospital, No. 201, Section 2, Shipai Road, Taipei, 11217, Taiwan. .,School of Medicine, National Yang-Ming University, No. 155, Section 2, Linong Street, Taipei, 11221, Taiwan.
| | - Wu-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, No. 123, DAPI Road, Kaohsiung, 83301, Taiwan. .,Department of Neurosurgery, Xiamen Chang Gung Memorial Hospital, No. 123, Xiafei Road, Fujian, 361026, China.
| | - Chun-Hong Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan.
| | - Chien-Wei Feng
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan.
| | - San-Nan Yang
- School of Medicine, College of Medicine and Department of Pediatrics, E-DA Hospital, I-Shou University, No. 1, Yida Road, Kaohsiung, 82445, Taiwan.
| | - Han-Chun Hung
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan.
| | - Nan-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, No. 2, Zhongzheng 1st Road, Kaohsiung, 80284, Taiwan.
| | - Pey-Ru Lin
- Institute of Biomedical Sciences, National Sun Yat-sen University, #70 Lienhai Road, Kaohsiung, 80424, Taiwan.
| | - San-Cher Chen
- Center for Neuroscience, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Institute of Biomedical Sciences, National Sun Yat-sen University, #70 Lienhai Road, Kaohsiung, 80424, Taiwan.
| | - Hui-Min David Wang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Department of Fragrance and Cosmetic Science, Kaohsiung Medical University, No. 100, Shiquan 1st Road, Kaohsiung, 80708, Taiwan. .,Graduate Institute of Natural Products, Kaohsiung Medical University, No. 100, Shiquan 1st Road, Kaohsiung, 80708, Taiwan. .,Center for Stem Cell Research, Kaohsiung Medical University, No. 100, Shiquan 1st Road, Kaohsiung, 80708, Taiwan.
| | - Tian-Huei Chu
- Institute of Biomedical Sciences, National Sun Yat-sen University, #70 Lienhai Road, Kaohsiung, 80424, Taiwan.
| | - Ming-Hong Tai
- Center for Neuroscience, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Institute of Biomedical Sciences, National Sun Yat-sen University, #70 Lienhai Road, Kaohsiung, 80424, Taiwan. .,Department of Biological Sciences, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan.
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Marine Biomedical Laboratory and Center for Translational Biopharmaceuticals, Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan.
| |
Collapse
|
44
|
Melemedjian OK, Khoutorsky A. Translational control of chronic pain. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 131:185-213. [PMID: 25744674 DOI: 10.1016/bs.pmbts.2014.11.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pain is a crucial physiological response to injury and pathologies. The development and maintenance of pain requires the expression of novel genes. The expression of such genes occurs in highly regulated and orchestrated manner where protein translation provides an exquisite temporal and spatial fidelity within the axons and dendrites of neurons. Signaling pathways that regulate local translation are activated by cytokines, neurotrophic factors, or neurotransmitters, which are released either due to tissue damage or neuronal activity. In recent years, the ERK and mTOR pathways have been demonstrated to be central in regulating local translation in neurons of both the peripheral and central nervous systems in diverse models of chronic pain. The ERK and mTOR pathways converge onto the cap-dependent translational machinery that regulates genes essential for the development of nociceptive sensitization. Moreover, inhibition of these pathways has proved to be effective in normalizing the biochemical changes and the associated pain in various preclinical models.
Collapse
Affiliation(s)
- Ohannes K Melemedjian
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, Maryland, USA.
| | - Arkady Khoutorsky
- Department of Biochemistry, Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
45
|
mTOR kinase: a possible pharmacological target in the management of chronic pain. BIOMED RESEARCH INTERNATIONAL 2015; 2015:394257. [PMID: 25685786 PMCID: PMC4313067 DOI: 10.1155/2015/394257] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 09/12/2014] [Indexed: 12/29/2022]
Abstract
Chronic pain represents a major public health problem worldwide. Current pharmacological treatments for chronic pain syndromes, including neuropathic pain, are only partially effective, with significant pain relief achieved in 40-60% of patients. Recent studies suggest that the mammalian target of rapamycin (mTOR) kinase and downstream effectors may be implicated in the development of chronic inflammatory, neuropathic, and cancer pain. The expression and activity of mTOR have been detected in peripheral and central regions involved in pain transmission. mTOR immunoreactivity was found in primary sensory axons, in dorsal root ganglia (DRG), and in dorsal horn neurons. This kinase is a master regulator of protein synthesis, and it is critically involved in the regulation of several neuronal functions, including the synaptic plasticity that is a major mechanism leading to the development of chronic pain. Enhanced activation of this pathway is present in different experimental models of chronic pain. Consistently, pharmacological inhibition of the kinase activity turned out to have significant antinociceptive effects in several experimental models of inflammatory and neuropathic pain. We will review the main evidence from animal and human studies supporting the hypothesis that mTOR may be a novel pharmacological target for the management of chronic pain.
Collapse
|
46
|
Rapamycin ameliorates neuropathic pain by activating autophagy and inhibiting interleukin-1β in the rat spinal cord. ACTA ACUST UNITED AC 2014; 34:830-837. [DOI: 10.1007/s11596-014-1361-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 09/22/2014] [Indexed: 12/13/2022]
|
47
|
Hua C, Zehou O, Ducassou S, Minard-Colin V, Hamel-Teillac D, Wolkenstein P, Valeyrie-Allanore L. Sirolimus improves pain in NF1 patients with severe plexiform neurofibromas. Pediatrics 2014; 133:e1792-7. [PMID: 24864177 DOI: 10.1542/peds.2013-1224] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Plexiform neurofibromas (PNs) are common and potentially debilitating complications of neurofibromatosis 1 (NF1). These benign nerve-sheath tumors are associated with significant pain and morbidity because they compress vital structures. The mammalian target of rapamycin (mTOR) pathway is a major mediator involved in tumor growth in NF1. We present 3 cases of patients with NF1, aged 8, 16, and 17 years, followed for inoperable and symptomatic PNs; patients received sirolimus for life-threatening and painful neurofibromas after multidisciplinary consultation. Epidemiologic, clinical, and radiologic data were retrospectively collected. The volume of PNs did not differ between baseline and 12-month follow-up and pain was alleviated, with withdrawal of analgesics in 2 cases at 6 months, and significantly decreased for the third case. Sirolimus for inoperable symptomatic PNs in patients with NF1 permitted stabilization of mass and produced unpredictable and important alleviation of pain in all cases with good tolerance. This treatment was proposed in extreme cases, in absence of therapeutic alternatives, after multidisciplinary consensus. The mTOR pathway may be both a major mediator of NF1 tumor growth and regulator of nociceptor sensitivity. mTOR inhibitors clinically used as anticancer and immunosuppressant drugs could be a potential treatment of chronic pain.
Collapse
Affiliation(s)
- Camille Hua
- AP-HP, Hôpital Henri-Mondor, Service de Dermatologie, Centre de référence des Neurofibromatoses, Créteil, France
| | - Ouidad Zehou
- AP-HP, Hôpital Henri-Mondor, Service de Dermatologie, Centre de référence des Neurofibromatoses, Créteil, France
| | - Stéphane Ducassou
- CHU Bordeaux, GH Pellegrin, Service d'Onco-Hématologie Pédiatrique, Bordeaux, France
| | - Véronique Minard-Colin
- Institut Gustave Roussy, Département de Cancérologie de l'Enfant et de l'Adolescent, Villejuif, France
| | | | - Pierre Wolkenstein
- AP-HP, Hôpital Henri-Mondor, Service de Dermatologie, Centre de référence des Neurofibromatoses, Créteil, France
- LIC EA 4393, Hôpital Henri Mondor, Assistance Publique Hôpitaux de Paris, Université Paris-Est Créteil, Créteil, France
| | - Laurence Valeyrie-Allanore
- AP-HP, Hôpital Henri-Mondor, Service de Dermatologie, Centre de référence des Neurofibromatoses, Créteil, France
- LIC EA 4393, Hôpital Henri Mondor, Assistance Publique Hôpitaux de Paris, Université Paris-Est Créteil, Créteil, France
| |
Collapse
|
48
|
Kjell J, Codeluppi S, Josephson A, Abrams MB. Spatial and cellular characterization of mTORC1 activation after spinal cord injury reveals biphasic increase mainly attributed to microglia/macrophages. Brain Pathol 2014; 24:557-67. [PMID: 24576152 PMCID: PMC4347463 DOI: 10.1111/bpa.12135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 02/17/2014] [Indexed: 01/07/2023] Open
Abstract
Mechanistic target of rapamycin complex 1 (mTORC1) is an intracellular kinase complex that regulates energy homeostasis and transcription. Modulation of mTORC1 has proven beneficial in experimental spinal cord injury, making this molecular target a candidate for therapeutic intervention in spinal cord injury. However, both inactivation and activation of mTORC1 have been reported beneficial for recovery. To obtain a more complete picture of mTORC1 activity, we aimed to characterize the spatiotemporal activation pattern of mTORC1 and identify activation in particular cell types after contusion spinal cord injury in rats. To be able to provide a spatial characterization of mTORC1 activation, we monitored activation of downstream target S6. We found robust mTORC1 activation both at the site of injury and in spinal segments rostral and caudal to the injury. There was constitutive mTORC1 activation in neurons that was biphasically reduced caudally after injury. We found biphasic mTORC1 activation in glial cells, primarily activated microglia/macrophages. Furthermore, we found mTORC1 activation in proliferating cells, suggesting this may be a function affected by mTORC1 modulation. Our results reveal potential windows of opportunity for therapeutic interference with mTORC1 signaling and immune cells as targets for inhibition of mTORC1 in spinal cord injury.
Collapse
Affiliation(s)
- Jacob Kjell
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | | | | |
Collapse
|
49
|
Iannitti T, Kerr BJ, Taylor BK. Mechanisms and pharmacology of neuropathic pain in multiple sclerosis. Curr Top Behav Neurosci 2014; 20:75-97. [PMID: 24590824 PMCID: PMC4464806 DOI: 10.1007/7854_2014_288] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The neuropathic pain of multiple sclerosis is quite prevalent and severely impacts quality of life. A few randomized, placebo-controlled, blinded clinical trials suggest that cannabis- and anticonvulsant-based treatments provide partial pain relief, but at the expense of adverse events. An even smaller, but emerging, number of translational studies are using rodent models of experimental autoimmune encephalomyelitis (EAE), which exhibit pain-like behaviors resembling those of Multiple sclerosis (MS) patients. These studies not only support the possible effectiveness of anticonvulsants, but also compel further clinical trials with serotonin-norepinephrine reuptake inhibitors, the immunosuppressant drug rapamycin, or drugs which interfere with glutamatergic neurotransmission. Future behavioral studies in EAE models are essential toward a new pharmacotherapy of multiple sclerosis pain.
Collapse
Affiliation(s)
- T Iannitti
- Department of Physiology, School of Medicine, University of Kentucky Medical Center, Lexington, KY, 40536-0298, USA
| | | | | |
Collapse
|
50
|
Abstract
The number of approved new molecular entity drugs has been decreasing as the pharmaceutical company investment in research and development is increasing. As we face this painful crisis, called an innovation gap, there is increasing awareness that development of new uses of existing drugs may be a powerful tool to help overcome this obstacle because it takes too long, costs too much and can be risky to release drugs developed de novo. Consequently, drug repositioning is emerging in different therapeutic areas, including the pain research area. Worldwide, pain is the main reason for seeking healthcare, and pain relief represents an unmet global clinical need. Therefore, development of analgesics with better efficacy, safety and cost effectiveness is of paramount importance. Despite the remarkable advancement in research on cellular and molecular mechanisms underlying pain pathophysiology over the past three decades, target-based therapeutic opportunities have not been pursued to the same extent. Phenotypic screening remains a more powerful tool for drug development than target-based screening so far. It sounds somewhat heretical, but some multi-action drugs, rather than very selective ones, have been developed intentionally. In the present review, we first critically discuss the utility of drug repositioning for analgesic drug development and then show examples of 'old' drugs that have been successfully repositioned or that are under investigation for their analgesic actions. We conclude that drug repositioning should be more strongly encouraged to help build a bridge between basic research and pain relief worldwide.
Collapse
Affiliation(s)
- Leandro Francisco Silva Bastos
- Laboratório de Imunofarmacologia, sala O4-202, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos 6627, Pampulha, Belo Horizonte, CEP 31.270-901, Brazil,
| | | |
Collapse
|