51
|
Falowski SM, Conti KR, Mogilner AY. Analysis of S1 DRG Programming to Determine Location of the DRG and Ideal Anatomic Positioning of the Electrode. Neuromodulation 2019; 23:252-257. [DOI: 10.1111/ner.13039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/14/2019] [Accepted: 07/18/2019] [Indexed: 01/04/2023]
|
52
|
Shinoda M, Fukuoka T, Takeda M, Iwata K, Noguchi K. Spinal glial cell line-derived neurotrophic factor infusion reverses reduction of Kv4.1-mediated A-type potassium currents of injured myelinated primary afferent neurons in a neuropathic pain model. Mol Pain 2019; 15:1744806919841196. [PMID: 30868936 PMCID: PMC6463340 DOI: 10.1177/1744806919841196] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
High frequency spontaneous activity in injured primary afferents has been proposed as a pathological mechanism of neuropathic pain following nerve injury. Although spinal infusion of glial cell line-derived neurotrophic factor reduces the activity of injured myelinated A-fiber neurons after fifth lumbar (L5) spinal nerve ligation in rats, the implicated molecular mechanism remains undetermined. The fast-inactivating transient A-type potassium current (IA) is an important determinant of neuronal excitability, and five voltage-gated potassium channel (Kv) alpha-subunits, Kv1.4, Kv3.4, Kv4.1, Kv4.2, and Kv4.3, display IA in heterologous expression systems. Here, we examined the effect of spinal glial cell line-derived neurotrophic factor infusion on IA and the expression of these five Kv mRNAs in injured A-fiber neurons using the in vitro patch clamp technique and in situ hybridization histochemistry. Glial cell line-derived neurotrophic factor infusion reversed axotomy-induced reduction of the rheobase, elongation of first spike duration, and depolarization of the resting membrane potential. L5 spinal nerve ligation significantly reduced the current density of IA and glial cell line-derived neurotrophic factor treatment reversed the reduction. Among the examined Kv mRNAs, only the change in Kv4.1-expression was parallel with the change in IA after spinal nerve ligation and glial cell line-derived neurotrophic factor treatment. These findings suggest that glial cell line-derived neurotrophic factor should reduce the hyperexcitability of injured A-fiber primary afferents by IA recurrence. Among the five IA-related Kv channels, Kv4.1 should be a key channel, which account for this IA recurrence.
Collapse
Affiliation(s)
- Masamichi Shinoda
- 1 Department of Physiology, Nihon University School of Dentistry, Surugadai, Chiyoda-ku, Tokyo, Japan
| | - Tetsuo Fukuoka
- 2 Department of Anatomy and Neuroscience, Hyogo College of Medicine, Mukogawa-cho, Nishinomiya, Hyogo, Japan.,3 Fukuoka Clinic, Kasuga, Suita, Osaka, Japan
| | - Mamoru Takeda
- 4 Laboratory of Food and Physiological Sciences, Department of Life and Food Sciences, School of Life and Environmental Sciences, Azabu University, Fuchinobe, Chuo-ku, Sagamihara, Japan
| | - Koichi Iwata
- 1 Department of Physiology, Nihon University School of Dentistry, Surugadai, Chiyoda-ku, Tokyo, Japan
| | - Koichi Noguchi
- 2 Department of Anatomy and Neuroscience, Hyogo College of Medicine, Mukogawa-cho, Nishinomiya, Hyogo, Japan
| |
Collapse
|
53
|
Contribution of DNMT1 to Neuropathic Pain Genesis Partially through Epigenetically Repressing Kcna2 in Primary Afferent Neurons. J Neurosci 2019; 39:6595-6607. [PMID: 31182635 DOI: 10.1523/jneurosci.0695-19.2019] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 02/08/2023] Open
Abstract
Expressional changes of pain-associated genes in primary sensory neurons of DRG are critical for neuropathic pain genesis. DNA methyltransferase (DNMT)-triggered DNA methylation silences gene expression. We show here that DNMT1, a canonical maintenance methyltransferase, acts as the de novo DNMT and is required for neuropathic pain genesis likely through repressing at least DRG Kcna2 gene expression in male mice. Peripheral nerve injury upregulated DNMT1 expression in the injured DRG through the transcription factor cAMP response element binding protein-triggered transcriptional activation of Dnmt1 gene. Blocking this upregulation prevented nerve injury-induced DNA methylation within the promoter and 5'-untranslated region of Kcna2 gene, rescued Kcna2 expression and total Kv current, attenuated hyperexcitability in the injured DRG neurons, and alleviated nerve injury-induced pain hypersensitivities. Given that Kcna2 is a key player in neuropathic pain, our findings suggest that DRG DNMT1 may be a potential target for neuropathic pain management.SIGNIFICANCE STATEMENT In the present study, we reported that DNMT1, a canonical DNA maintenance methyltransferase, is upregulated via the activation of the transcription factor CREB in the injured DRG after peripheral nerve injury. This upregulation was responsible for nerve injury-induced de novo DNA methylation within the promoter and 5'-untranslated region of the Kcna2 gene, reductions in Kcna2 expression and Kv current and increases in neuronal excitability in the injured DRG. Since pharmacological inhibition or genetic knockdown of DRG DNMT1 alleviated nerve injury-induced pain hypersensitivities, DRG DNMT1 contributes to neuropathic pain genesis partially through repression of DRG Kcna2 gene expression.
Collapse
|
54
|
Jiang R, Li P, Yao YX, Li H, Liu R, Huang LE, Ling S, Peng Z, Yang J, Zha L, Xia LP, Chen X, Feng Z. Pulsed radiofrequency to the dorsal root ganglion or the sciatic nerve reduces neuropathic pain behavior, decreases peripheral pro-inflammatory cytokines and spinal β-catenin in chronic constriction injury rats. Reg Anesth Pain Med 2019; 44:rapm-2018-100032. [PMID: 31092705 DOI: 10.1136/rapm-2018-100032] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 04/15/2019] [Accepted: 04/22/2019] [Indexed: 11/04/2022]
Abstract
BACKGROUND AND OBJECTIVES Pulsed radiofrequency (PRF) is a minimal neurodestructive interventional pain therapy. However, its analgesic mechanism remains largely unclear. We aimed to investigate the peripheral and spinal mechanisms of PRF applied either adjacent to the ipsilateral L5 dorsal root ganglion (PRF-DRG) or PRF to the sciatic nerve (PRF-SN) in the neuropathic pain behavior induced by chronic constriction injury (CCI) in rats. METHODS On day 0, CCI or sham surgeries were performed. Rats then received either PRF-DRG, PRF-SN, or sham PRF treatment on day 4. Pain behavioral tests were conducted before surgeries and on days 1, 3, 5, 7, 9, 11, 13, and 14. After the behavioral tests, the rats were sacrificed. The venous blood or sciatic nerve samples were collected for ELISAs and the dorsal horns of the L4-L6 spinal cord were collected for western blot examination. RESULTS The mechanical allodynia and the thermal hyperalgesia has been relieved by a single PRF-DRG or PRF-SN application. In addition, the analgesic effect of PRF-DRG was superior to PRF-SN on CCI-induced neuropathic pain. Either PRF-DRG or PRF-SN reversed the enhancement of interleukin 1β (IL-1β) and tumor necrosis factor alpha (TNF-α) levels in the blood of CCI rats. PRF-DRG or PRF-SN also downregulated spinal β-catenin expression. CONCLUSIONS PRF treatment either to DRG or to sciatic nerve reduced neuropathic pain behavior, and reduced peripheral levels of pro-inflammatory cytokines and spinal β-catenin expression in CCI rats. PRF to DRG has a better analgesic effect than PRF to the nerve.
Collapse
Affiliation(s)
- Ren Jiang
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Anesthesiology, Yinzhou No. 2 Hospital, Ningbo, China
| | - Ping Li
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Anesthesiology, Yinzhou No. 2 Hospital, Ningbo, China
| | - Yong-Xing Yao
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Li
- Department of Anesthesiology, Yinzhou No. 2 Hospital, Ningbo, China
| | - Rongjun Liu
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Ling-Er Huang
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sunbin Ling
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiyou Peng
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Yang
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Leiqiong Zha
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li-Ping Xia
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaowei Chen
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Zhiying Feng
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
55
|
Pickles K. Is Electrical Nerve Stimulation the Answer for Management of Equine Headshaking? Vet Clin North Am Equine Pract 2019; 35:263-274. [PMID: 31047766 DOI: 10.1016/j.cveq.2019.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Horses with trigeminal mediated headshaking (TMHS) have a decreased activation threshold of the trigeminal nerve and clinical signs are suspected to be a manifestation of trigeminal neuralgia. Electrical nerve stimulation (ENS) is used for management of neuralgia in humans and appears to work via gate control theory. Use of an equine specific percutaneous ENS program in over 130 TMHS horses has resulted in approximately 50% success return to previous work. Electroacupuncture may also be useful in the management TMHS. Optimization of ENS procedures for TMHS is likely to require a greater understanding of the etiopathogenesis of the aberrant neurophysiology.
Collapse
Affiliation(s)
- Kirstie Pickles
- Chine House Veterinary Hospital, 12 Cossington Road, Sileby, Leicestershire LE12 7RS, UK.
| |
Collapse
|
56
|
Suppression of neuropathic pain by selective silencing of dorsal root ganglion ectopia using nonblocking concentrations of lidocaine. Pain 2019; 160:2105-2114. [DOI: 10.1097/j.pain.0000000000001602] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
57
|
Bennett DL, Clark AJ, Huang J, Waxman SG, Dib-Hajj SD. The Role of Voltage-Gated Sodium Channels in Pain Signaling. Physiol Rev 2019; 99:1079-1151. [DOI: 10.1152/physrev.00052.2017] [Citation(s) in RCA: 256] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Acute pain signaling has a key protective role and is highly evolutionarily conserved. Chronic pain, however, is maladaptive, occurring as a consequence of injury and disease, and is associated with sensitization of the somatosensory nervous system. Primary sensory neurons are involved in both of these processes, and the recent advances in understanding sensory transduction and human genetics are the focus of this review. Voltage-gated sodium channels (VGSCs) are important determinants of sensory neuron excitability: they are essential for the initial transduction of sensory stimuli, the electrogenesis of the action potential, and neurotransmitter release from sensory neuron terminals. Nav1.1, Nav1.6, Nav1.7, Nav1.8, and Nav1.9 are all expressed by adult sensory neurons. The biophysical characteristics of these channels, as well as their unique expression patterns within subtypes of sensory neurons, define their functional role in pain signaling. Changes in the expression of VGSCs, as well as posttranslational modifications, contribute to the sensitization of sensory neurons in chronic pain states. Furthermore, gene variants in Nav1.7, Nav1.8, and Nav1.9 have now been linked to human Mendelian pain disorders and more recently to common pain disorders such as small-fiber neuropathy. Chronic pain affects one in five of the general population. Given the poor efficacy of current analgesics, the selective expression of particular VGSCs in sensory neurons makes these attractive targets for drug discovery. The increasing availability of gene sequencing, combined with structural modeling and electrophysiological analysis of gene variants, also provides the opportunity to better target existing therapies in a personalized manner.
Collapse
Affiliation(s)
- David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Alex J. Clark
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Jianying Huang
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Stephen G. Waxman
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Sulayman D. Dib-Hajj
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
58
|
Graham RD, Bruns TM, Duan B, Lempka SF. Dorsal root ganglion stimulation for chronic pain modulates Aβ-fiber activity but not C-fiber activity: A computational modeling study. Clin Neurophysiol 2019; 130:941-951. [PMID: 30981900 DOI: 10.1016/j.clinph.2019.02.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/23/2019] [Accepted: 02/16/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The goal of this project was to use computational models to investigate which types of primary sensory neurons are modulated by dorsal root ganglion stimulation (DRGS) to provide pain relief. METHODS We modeled DRGS by coupling an anatomical finite element model of a human L5 dorsal root ganglion to biophysical models of primary sensory neurons. We calculated the stimulation amplitude needed to elicit an action potential in each neuron, and examined how DRGS affected sensory neuron activity. RESULTS We showed that within clinical ranges of stimulation parameters, DRGS drives the activity of large myelinated Aβ-fibers but does not directly activate small nonmyelinated C-fibers. We also showed that the position of the active and return electrodes and the polarity of the stimulus pulse influence neural activation. CONCLUSIONS Our results indicate that DRGS may provide pain relief by activating pain-gating mechanisms in the dorsal horn via repeated activation of large myelinated afferents. SIGNIFICANCE Understanding the mechanisms of action of DRGS-induced pain relief may lead to innovations in stimulation technologies that improve patient outcomes.
Collapse
Affiliation(s)
- Robert D Graham
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Tim M Bruns
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Bo Duan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Scott F Lempka
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA; Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
59
|
Li ZH, Cui D, Qiu CJ, Song XJ. Cyclic nucleotide signaling in sensory neuron hyperexcitability and chronic pain after nerve injury. NEUROBIOLOGY OF PAIN 2019; 6:100028. [PMID: 31223142 PMCID: PMC6565612 DOI: 10.1016/j.ynpai.2019.100028] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 11/08/2022]
Abstract
Activation of cAMP-PKA and cGMP-PKG pathways contributes to injury-induced sensory neuron hyperexcitability. Activation of cAMP and cGMP contributes to the development of bone cancer pain. PAR2 activation mediates injury-induced cAMP-dependent sensory neuron hyperexcitability.
The cyclic nucleotide signaling, including cAMP-PKA and cGMP-PKG pathways, has been well known to play critical roles in regulating cellular growth, metabolism and many other intracellular processes. In recent years, more and more studies have uncovered the roles of cAMP and cGMP in the nervous system. The cAMP and cGMP signaling mediates chronic pain induced by different forms of injury and stress. Here we summarize the roles of cAMP-PKA and cGMP-PKG signaling pathways in the pathogenesis of chronic pain after nerve injury. In addition, acute dissociation and chronic compression of the dorsal root ganglion (DRG) neurons, respectively, leads to neural hyperexcitability possibly through PAR2 activation-dependent activation of cAMP-PKA pathway. Clinically, radiotherapy can effectively alleviate bone cancer pain at least partly through inhibiting the cancer cell-induced activation of cAMP-PKA pathway. Roles of cyclic nucleotide signaling in neuropathic and inflammatory pain are also seen in many other animal models and are involved in many pro-nociceptive mechanisms including the activation of hyperpolarization-activated cyclic nucleotide (HCN)-modulated ion channels and the exchange proteins directly activated by cAMP (EPAC). Further understanding the roles of cAMP and cGMP signaling in the pathogenesis of chronic pain is theoretically significant and clinically valuable for treatment of chronic pain.
Collapse
Affiliation(s)
- Ze-Hua Li
- Department of Biology, SUSTech Center for Pain Medicine, and Medical School, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.,Department of Anesthesiology and Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education of China), Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing 100142, China
| | - Dong Cui
- Department of Biology, SUSTech Center for Pain Medicine, and Medical School, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.,Department of Anesthesiology and Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education of China), Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing 100142, China
| | - Cheng-Jie Qiu
- Department of Biology, SUSTech Center for Pain Medicine, and Medical School, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Xue-Jun Song
- Department of Biology, SUSTech Center for Pain Medicine, and Medical School, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.,Department of Anesthesiology and Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education of China), Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
60
|
Chemokine CCL2 and its receptor CCR2 in the dorsal root ganglion contribute to oxaliplatin-induced mechanical hypersensitivity. Pain 2019; 159:1308-1316. [PMID: 29554018 DOI: 10.1097/j.pain.0000000000001212] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Activation of innate immune mechanisms within the dorsal root ganglion and spinal dorsal horn has been shown to play a key role in the development of neuropathic pain including paclitaxel-related chemotherapy-induced peripheral neuropathy (CIPN). Here, we tested whether similar mechanisms are generalizable to oxaliplatin-induced CIPN. After a single intraperitoneal injection of 3 mg/kg oxaliplatin, mechanical withdrawal threshold and the expression of C-C chemokine ligand 2 (CCL2) and its receptor, CCR2, in the dorsal root ganglion were measured by behavioral testing and immunohistochemical staining, respectively. Mechanical responsiveness increased from the first day after oxaliplatin injection and persisted until day 15, the last day of this experiment. Immunohistochemical showed that the expression of CCL2/CCR2 started to increase by 4 hours after oxaliplatin treatment, was significantly increased at day 4, and then both signals became normalized by day 15. Cotreatment with intrathecal anti-CCL2 antibodies prevented the development of oxaliplatin-induced mechanical hyperresponsiveness, and transiently reversed established hyperalgesia when given 1 week after chemotherapy. This is the first study to demonstrate CCL2/CCR2 signaling in a model of oxaliplatin-related CIPN; and it further shows that blocking of this signal can attenuate the development of oxaliplatin-induced mechanical hyperalgesia. Activation of innate immune mechanisms may therefore be a generalized basis for CIPN irrespective of the specific class of agent.
Collapse
|
61
|
Abstract
Voltage-gated potassium (Kv) channels are increasingly recognised as key regulators of nociceptive excitability. Kcns1 is one of the first potassium channels to be associated with neuronal hyperexcitability and mechanical sensitivity in the rat, as well as pain intensity and risk of developing chronic pain in humans. Here, we show that in mice, Kcns1 is predominantly expressed in the cell body and axons of myelinated sensory neurons positive for neurofilament-200, including Aδ-fiber nociceptors and low-threshold Aβ mechanoreceptors. In the spinal cord, Kcns1 was detected in laminae III to V of the dorsal horn where most sensory A fibers terminate, as well as large motoneurons of the ventral horn. To investigate Kcns1 function specifically in the periphery, we generated transgenic mice in which the gene is deleted in all sensory neurons but retained in the central nervous system. Kcns1 ablation resulted in a modest increase in basal mechanical pain, with no change in thermal pain processing. After neuropathic injury, Kcns1 KO mice exhibited exaggerated mechanical pain responses and hypersensitivity to both noxious and innocuous cold, consistent with increased A-fiber activity. Interestingly, Kcns1 deletion also improved locomotor performance in the rotarod test, indicative of augmented proprioceptive signalling. Our results suggest that restoring Kcns1 function in the periphery may be of some use in ameliorating mechanical and cold pain in chronic states.
Collapse
|
62
|
Peng Q, Mechanic J, Shoieb A, Pardo ID, Schaevitz L, Fenyk-Melody J, Vitsky A, Boucher M, Somps C, Cook JC, Liu CN. Circulating microRNA and automated motion analysis as novel methods of assessing chemotherapy-induced peripheral neuropathy in mice. PLoS One 2019; 14:e0210995. [PMID: 30677061 PMCID: PMC6345499 DOI: 10.1371/journal.pone.0210995] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 01/02/2019] [Indexed: 11/18/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CiPN) is a serious adverse effect in the clinic, but nonclinical assessment methods in animal studies are limited to labor intensive behavioral tests or semi-quantitative microscopic evaluation. Hence, microRNA (miRNA) biomarkers and automated in-life behavioral tracking were assessed for their utility as non-invasive methods. To address the lack of diagnostic biomarkers, we explored miR-124, miR-183 and miR-338 in a CiPN model induced by paclitaxel, a well-known neurotoxic agent. In addition, conventional and Vium's innovative Digital Vivarium technology-based in-life behavioral tests and postmortem microscopic examination of the dorsal root ganglion (DRG) and the sciatic nerve were performed. Terminal blood was collected on days 8 or 16, after 20 mg/kg paclitaxel was administered every other day for total of 4 or 7 doses, respectively, for plasma miRNA quantification by RT-qPCR. DRG and sciatic nerve samples were collected from mice sacrificed on day 16 for miRNA quantification. Among the three miRNAs analyzed, only miR-124 was statistically significantly increased (5 fold and 10 fold on day 8 and day 16, respectively). The increase in circulating miR-124 correlated with cold allodynia and axonal degeneration in both DRG and sciatic nerve. Automated home cage motion analysis revealed for the first time that nighttime motion was significantly decreased (P < 0.05) in paclitaxel-dosed animals. Although both increase in circulating miR-124 and decrease in nighttime motion are compelling, our results provide positive evidence warranting further testing using additional peripheral nerve toxicants and diverse experimental CiPN models.
Collapse
Affiliation(s)
- Qinghai Peng
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, La Jolla, California, United States of America
| | | | - Ahmed Shoieb
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, Groton, Connecticut, United States of America
| | - Ingrid D. Pardo
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, Groton, Connecticut, United States of America
| | | | - Judith Fenyk-Melody
- Comparative Medicine, Pfizer Worldwide Research & Development, Cambridge, Massachusetts, United States of America
| | - Allison Vitsky
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, La Jolla, California, United States of America
| | - Magalie Boucher
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, Cambridge, Massachusetts, United States of America
| | - Chris Somps
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, Groton, Connecticut, United States of America
| | - Jon C. Cook
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, Groton, Connecticut, United States of America
| | - Chang-Ning Liu
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, Groton, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
63
|
Peripheral oxytocin restores light touch and nociceptor sensory afferents towards normal after nerve injury. Pain 2019; 160:1146-1155. [DOI: 10.1097/j.pain.0000000000001495] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
64
|
Joubert F, Acosta MDC, Gallar J, Fakih D, Sahel JA, Baudouin C, Bodineau L, Mélik Parsadaniantz S, Réaux-Le Goazigo A. Effects of corneal injury on ciliary nerve fibre activity and corneal nociception in mice: A behavioural and electrophysiological study. Eur J Pain 2018; 23:589-602. [DOI: 10.1002/ejp.1332] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 09/18/2018] [Accepted: 10/20/2018] [Indexed: 01/18/2023]
Affiliation(s)
- Fanny Joubert
- INSERM; CNRS; Institut de la Vision; Sorbonne Université; Paris France
| | - Maria del Carmen Acosta
- Instituto de Neurosciencas; Universidad Miguel Hernández-CSIC; San Juan de Alicante; Alicante Spain
| | - Juana Gallar
- Instituto de Neurosciencas; Universidad Miguel Hernández-CSIC; San Juan de Alicante; Alicante Spain
| | - Darine Fakih
- INSERM; CNRS; Institut de la Vision; Sorbonne Université; Paris France
- Laboratoires Théa; Clermont-Ferrand France
| | - José-Alain Sahel
- INSERM; CNRS; Institut de la Vision; Sorbonne Université; Paris France
- INSERM-DGOS CIC 1423; CHNO des Quinze-Vingts; DHU Sight Restore; Paris France
| | - Christophe Baudouin
- INSERM; CNRS; Institut de la Vision; Sorbonne Université; Paris France
- INSERM-DGOS CIC 1423; CHNO des Quinze-Vingts; DHU Sight Restore; Paris France
- University Versailles Saint Quentin en Yvelines; Versailles France
| | - Laurence Bodineau
- INSERM; Sorbonne Université; UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique; Paris France
| | | | | |
Collapse
|
65
|
Devor M. Rethinking the causes of pain in herpes zoster and postherpetic neuralgia: the ectopic pacemaker hypothesis. Pain Rep 2018; 3:e702. [PMID: 30706041 PMCID: PMC6344138 DOI: 10.1097/pr9.0000000000000702] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/10/2018] [Indexed: 01/29/2023] Open
Abstract
INTRODUCTION Pain in herpes zoster (HZ) and postherpetic neuralgia (PHN) is traditionally explained in terms of 2 processes: irritable nociceptors in the rash-inflamed skin and, later, deafferentation due to destruction of sensory neurons in one virally infected dorsal root ganglion. OBJECTIVES AND METHODS Consideration of the evidence supporting this explanation in light of contemporary understanding of the pain system finds it wanting. An alternative hypothesis is proposed as a replacement. RESULTS This model, the ectopic pacemaker hypothesis of HZ and PHN, proposes that pain in both conditions is driven by hyperexcitable ectopic pacemaker sites at various locations in primary sensory neurons affected by the causative varicella zoster virus infection. This peripheral input is exacerbated by central sensitization induced and maintained by the ectopic activity. CONCLUSIONS The shift in perspective regarding the pain mechanism in HZ/PHN has specific implications for clinical management.
Collapse
Affiliation(s)
- Marshall Devor
- Department of Cell and Developmental Biology, Institute of Life Sciences, and Center for Research on Pain, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
66
|
Odem MA, Bavencoffe AG, Cassidy RM, Lopez ER, Tian J, Dessauer CW, Walters ET. Isolated nociceptors reveal multiple specializations for generating irregular ongoing activity associated with ongoing pain. Pain 2018; 159:2347-2362. [PMID: 30015712 PMCID: PMC6193853 DOI: 10.1097/j.pain.0000000000001341] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Ongoing pain has been linked to ongoing activity (OA) in human C-fiber nociceptors, but rodent models of pain-related OA have concentrated on allodynia rather than ongoing pain, and on OA generated in non-nociceptive Aβ fibers rather than C-fiber nociceptors. Little is known about how ongoing pain or nociceptor OA is generated. To define neurophysiological alterations underlying nociceptor OA, we have used isolated dorsal root ganglion neurons that continue to generate OA after removal from animals displaying ongoing pain. We subclassify OA as either spontaneous activity generated solely by alterations intrinsic to the active neuron or as extrinsically driven OA. Both types of OA were implicated previously in nociceptors in vivo and after isolation following spinal cord injury, which produces chronic ongoing pain. Using novel automated algorithms to analyze irregular changes in membrane potential, we have found, in a distinctive, nonaccommodating type of probable nociceptor, induction by spinal cord injury of 3 alterations that promote OA: (1) prolonged depolarization of resting membrane potential, (2) a hyperpolarizing shift in the voltage threshold for action potential generation, and (3) an increase in the incidence of large depolarizing spontaneous fluctuations (DSFs). Can DSFs also be enhanced acutely to promote OA in neurons from uninjured animals? A low dose of serotonin failed to change resting membrane potential but lowered action potential threshold. When combined with artificial depolarization to model inflammation, serotonin also strongly potentiated DSFs and OA. These findings reveal nociceptor specializations for generating OA that may promote ongoing pain in chronic and acute conditions.
Collapse
Affiliation(s)
- Max A. Odem
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Alexis G. Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Ryan M. Cassidy
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Elia R. Lopez
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Carmen W. Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Edgar T. Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| |
Collapse
|
67
|
Kang XJ, Chi YN, Chen W, Liu FY, Cui S, Liao FF, Cai J, Wan Y. Increased expression of Ca V3.2 T-type calcium channels in damaged DRG neurons contributes to neuropathic pain in rats with spared nerve injury. Mol Pain 2018; 14:1744806918765808. [PMID: 29592785 PMCID: PMC5888807 DOI: 10.1177/1744806918765808] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Ion channels are very important in the peripheral sensitization in neuropathic pain. Our present study aims to investigate the possible contribution of CaV3.2 T-type calcium channels in damaged dorsal root ganglion neurons in neuropathic pain. We established a neuropathic pain model of rats with spared nerve injury. In these model rats, it was easy to distinguish damaged dorsal root ganglion neurons (of tibial nerve and common peroneal nerve) from intact dorsal root ganglion neurons (of sural nerves). Our results showed that CaV3.2 protein expression increased in medium-sized neurons from the damaged dorsal root ganglions but not in the intact ones. With whole cell patch clamp recording technique, it was found that after-depolarizing amplitudes of the damaged medium-sized dorsal root ganglion neurons increased significantly at membrane potentials of −85 mV and −95 mV. These results indicate a functional up-regulation of CaV3.2 T-type calcium channels in the damaged medium-sized neurons after spared nerve injury. Behaviorally, blockade of CaV3.2 with antisense oligodeoxynucleotides could significantly reverse mechanical allodynia. These results suggest that CaV3.2 T-type calcium channels in damaged medium-sized dorsal root ganglion neurons might contribute to neuropathic pain after peripheral nerve injury.
Collapse
Affiliation(s)
- Xue-Jing Kang
- 1 Neuroscience Research Institute, Peking University, Beijing, China
| | - Ye-Nan Chi
- 1 Neuroscience Research Institute, Peking University, Beijing, China.,2 Department of Anesthesiology, Dongfang Hospital, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Wen Chen
- 1 Neuroscience Research Institute, Peking University, Beijing, China
| | - Feng-Yu Liu
- 1 Neuroscience Research Institute, Peking University, Beijing, China
| | - Shuang Cui
- 1 Neuroscience Research Institute, Peking University, Beijing, China
| | - Fei-Fei Liao
- 1 Neuroscience Research Institute, Peking University, Beijing, China
| | - Jie Cai
- 1 Neuroscience Research Institute, Peking University, Beijing, China
| | - You Wan
- 1 Neuroscience Research Institute, Peking University, Beijing, China.,3 Key Lab for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
| |
Collapse
|
68
|
Yamakita S, Horii Y, Takemura H, Matsuoka Y, Yamashita A, Yamaguchi Y, Matsuda M, Sawa T, Amaya F. Synergistic activation of ERK1/2 between A-fiber neurons and glial cells in the DRG contributes to pain hypersensitivity after tissue injury. Mol Pain 2018; 14:1744806918767508. [PMID: 29592783 PMCID: PMC5881964 DOI: 10.1177/1744806918767508] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background Intense nociceptive signaling arising from ongoing injury activates primary afferent nociceptive systems to generate peripheral sensitization. ERK1/2 phosphorylation in dorsal root ganglion can be used to visualize intracellular signal activity immediately after noxious stimulation. The aim of this study was to investigate spatiotemporal characteristics of ERK1/2 phosphorylation against tissue injury in the primary afferent neurons. Methods Plantar incisions were made in the hind paws of Sprague-Dawley rats (n =150). Levobupivacaine was injected into the plantar aspect of the paws and ankles, Mitogen-activated protein kinase kinase (MEK) inhibitor was injected into the paw, and carbenoxolone, dual inhibitor of the gap junction and pannexin channel, was intraperitoneally injected. Pain hypersensitivity was investigated by a behavioral study, while phosphorylated ERK1/2 was detected in dorsal root ganglion and hind paw using immunohistochemistry and Western blot. Results Phosphorylated ERK1/2 was induced in dorsal root ganglion (26.8 ± 2.9% at baseline, 65.6 ± 3.6% at 2 min, and 26.3 ± 3.4% at 2 h) after the incision. NF-200 positive A-fiber neurons and satellite glial cells were positive for phosphorylated ERK1/2. Injury-induced pain hypersensitivity was abolished by MEK inhibitor. Levobupivacaine treatment inhibited phosphorylated ERK1/2 induction, carbenoxolone treatment inhibited glial phosphorylated ERK1/2 at 2 min after the injury, and carbenoxolone inhibited pain hypersensitivity and neuronal phosphorylated ERK1/2 at 1 h after the injury. Conclusion ERK1/2 phosphorylation in A-fiber neurons and satellite glial cells immediately after injury contributes to the generation of pain hypersensitivity. Signal communication between neurons and satellite glial cells expands the duration of neuronal ERK1/2 phosphorylation and pain hypersensitivity at 1 h after tissue injury.
Collapse
Affiliation(s)
- Shunsuke Yamakita
- 1 Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,2 Research Unit for the Neurobiology of Pain, Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasuhiko Horii
- 1 Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,2 Research Unit for the Neurobiology of Pain, Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hitomi Takemura
- 1 Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,2 Research Unit for the Neurobiology of Pain, Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yutaka Matsuoka
- 1 Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,2 Research Unit for the Neurobiology of Pain, Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ayahiro Yamashita
- 1 Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,2 Research Unit for the Neurobiology of Pain, Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yosuke Yamaguchi
- 1 Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,2 Research Unit for the Neurobiology of Pain, Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Megumi Matsuda
- 1 Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,2 Research Unit for the Neurobiology of Pain, Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Teiji Sawa
- 1 Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Fumimasa Amaya
- 1 Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,2 Research Unit for the Neurobiology of Pain, Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
69
|
Zhu YF, Kwiecien JM, Dabrowski W, Ungard R, Zhu KL, Huizinga JD, Henry JL, Singh G. Cancer pain and neuropathic pain are associated with A β sensory neuronal plasticity in dorsal root ganglia and abnormal sprouting in lumbar spinal cord. Mol Pain 2018; 14:1744806918810099. [PMID: 30324862 PMCID: PMC6243409 DOI: 10.1177/1744806918810099] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Evidence suggests that there are both nociceptive and neuropathic components of cancer-induced pain. We have observed that changes in intrinsic membrane properties and excitability of normally non-nociceptive Aβ sensory neurons are consistent in rat models of peripheral neuropathic pain and cancer-induced pain. This has prompted a comparative investigation of the intracellular electrophysiological characteristics of sensory neurons and of the ultrastructural morphology of the dorsal horn in rat models of neuropathic pain and cancer-induced pain. Neuropathic pain model rats were induced with a polyethylene cuff implanted around a sciatic nerve. Cancer-induced pain model rats were induced with mammary rat metastasis tumour-1 rat breast cancer or MATLyLu rat prostate cancer cells implanted into the distal epiphysis of a femur. Behavioural evidence of nociception was detected using von Frey tactile assessment. Aβ-fibre low threshold mechanoreceptor neurons in both cancer-induced pain and neuropathic pain models exhibited slower dynamics of action potential genesis, including a wider action potential duration and lower action potential amplitude compared to those in control animals. Enhanced excitability of Aβ-fibre low threshold mechanoreceptor neurons was also observed in cancer-induced pain and neuropathic pain models. Furthermore, both cancer-induced pain and neuropathic pain models showed abundant abnormal axonal sprouting in bundles of myelinated axons in the ipsilateral spinal laminae IV and V. The patterns of changes show consistency between rat models of cancer-induced pain and neuropathic pain. These findings add to the body of evidence that animal models of cancer-induced pain and neuropathic pain share features that may contribute to the peripheral and central sensitization and tactile hypersensitivity in both pain states.
Collapse
Affiliation(s)
- Yong Fang Zhu
- 1 Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada.,2 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Jacek M Kwiecien
- 2 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.,3 Department of Clinical Pathomorphology, Medical University of Lublin, Lublin, Poland
| | - Wojciech Dabrowski
- 4 Department of Anaesthesiology and Intensive Therapy, Medical University of Lublin, Lublin, Poland
| | - Robert Ungard
- 1 Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada.,2 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Kan Lun Zhu
- 2 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Jan D Huizinga
- 5 Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - James L Henry
- 6 Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Gurmit Singh
- 1 Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada.,2 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
70
|
Petersen BA, Nanivadekar AC, Chandrasekaran S, Fisher LE. Phantom limb pain: peripheral neuromodulatory and neuroprosthetic approaches to treatment. Muscle Nerve 2018; 59:154-167. [DOI: 10.1002/mus.26294] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Bailey A. Petersen
- Department of Bioengineering; University of Pittsburgh; 3520 Fifth Avenue, Pittsburgh Pennsylvania 15213 USA
| | - Ameya C. Nanivadekar
- Department of Bioengineering; University of Pittsburgh; 3520 Fifth Avenue, Pittsburgh Pennsylvania 15213 USA
| | - Santosh Chandrasekaran
- Department of Physical Medicine and Rehabilitation; University of Pittsburgh; Pittsburgh Pennsylvania USA
| | - Lee E. Fisher
- Department of Bioengineering; University of Pittsburgh; 3520 Fifth Avenue, Pittsburgh Pennsylvania 15213 USA
- Department of Physical Medicine and Rehabilitation; University of Pittsburgh; Pittsburgh Pennsylvania USA
| |
Collapse
|
71
|
Chao THH, Chen JH, Yen CT. Plasticity changes in forebrain activity and functional connectivity during neuropathic pain development in rats with sciatic spared nerve injury. Mol Brain 2018; 11:55. [PMID: 30285801 PMCID: PMC6167811 DOI: 10.1186/s13041-018-0398-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/20/2018] [Indexed: 01/08/2023] Open
Abstract
Neuropathic pain is a major worldwide health problem. Although central sensitization has been reported in well-established neuropathic conditions, information on the acute brain activation patterns in response to peripheral nerve injury is lacking. This study first mapped the brain activity in rats immediately following spared nerve injury (SNI) of the sciatic nerve. Using blood-oxygenation-level-dependent functional magnetic resonance imaging (BOLD-fMRI), we observed sustained activation in the bilateral insular cortices (ICs), primary somatosensory cortex (S1), and cingulate cortex. Second, this study sought to link this sustained activation pattern with brain sensitization. Using manganese-enhanced magnetic resonance imaging (MEMRI), we observed enhanced activity in the ipsilateral anterior IC (AIC) in free-moving SNI rats on Days 1 and 8 post-SNI. Furthermore, enhanced functional connectivity between the ipsilateral AIC, bilateral rostral AIC, and S1 was observed on Day 8 post-SNI. Chronic electrophysiological recording experiments were conducted to confirm the tonic neuronal activation in selected brain regions. Our data provide evidence of tonic activation-dependent brain sensitization during neuropathic pain development and offer evidence that the plasticity changes in the IC and S1 may contribute to neuropathic pain development.
Collapse
Affiliation(s)
- Tzu-Hao Harry Chao
- Department of Life Science, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, Taipei, 10617, Taiwan
| | - Jyh-Horng Chen
- Interdisciplinary MRI/MRS Lab, Department of Electrical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, Taipei, 10617, Taiwan
| | - Chen-Tung Yen
- Department of Life Science, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, Taipei, 10617, Taiwan.
| |
Collapse
|
72
|
García G, Martínez-Rojas VA, Oviedo N, Murbartián J. Blockade of anoctamin-1 in injured and uninjured nerves reduces neuropathic pain. Brain Res 2018; 1696:38-48. [DOI: 10.1016/j.brainres.2018.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 04/12/2018] [Accepted: 06/01/2018] [Indexed: 12/28/2022]
|
73
|
Kitamura N, Nagami E, Matsushita Y, Kayano T, Shibuya I. Constitutive activity of transient receptor potential vanilloid type 1 triggers spontaneous firing in nerve growth factor-treated dorsal root ganglion neurons of rats. IBRO Rep 2018; 5:33-42. [PMID: 30211336 PMCID: PMC6132080 DOI: 10.1016/j.ibror.2018.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 08/16/2018] [Indexed: 12/30/2022] Open
Abstract
We examined the role of TRPV1 in the generation of spontaneous APs in NGF-treated cultured DRG neurons of rats. Spontaneous firing in the on-cell configuration was abolished by TRPV1 antagonists capsazepine and BCTC. Chronic treatment with NGF induced capsazepine- and BCTC-sensitive cation conductance. NGF-induced cation conductance through TRPV1 causes spontaneous firing.
Dorsal root ganglion (DRG) neurons cultured in the presence of nerve growth factor (NGF, 100 ng/ml) often show a spontaneous action potential. Underlying mechanisms of this spontaneous firing were examined using the patch clamp technique. The spontaneous firing in the on-cell configuration was abolished by a decrease in the Na+ concentration and by the TRPV1 antagonists capsazepine (10 μM) and BCTC (1 μM). These responses were accompanied by hyperpolarization of the resting potential. The holding current observed in neurons voltage clamped at –60 mV in the whole-cell configuration was significantly larger in the neurons that fired spontaneously, indicating that these neurons had an additional cation conductance that caused depolarization and triggered action potentials. The holding current in the firing neurons was decreased by extracellular Na+ reduction, capsazepine and BCTC. The amplitudes of the capsazepine- or BCTC-sensitive component of the holding current in the spontaneously firing neurons were ten times as large as those recorded in the other neurons showing no spontaneous firing. However, the amplitudes of the current responses to capsaicin (1 μM) were not different regardless of the presence of spontaneous firing or treatment with NGF. These results indicate that chronic NGF treatment of cultured DRG neurons in rats induces a constitutively active cation conductance through TRPV1, which depolarizes the neurons and triggers spontaneous action potentials in the absence of any stimuli. Since NGF in the DRG is reported to increase after nerve injury, this NGF-mediated regulation of TRPV1 may be a cause of the pathogenesis of neuropathic pain.
Collapse
Affiliation(s)
- Naoki Kitamura
- Laboratory of Veterinary Physiology, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101, Koyama-cho Minami, Tottori 680-8553, Japan
| | - Erika Nagami
- Laboratory of Veterinary Physiology, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101, Koyama-cho Minami, Tottori 680-8553, Japan
| | - Yumi Matsushita
- Laboratory of Veterinary Physiology, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101, Koyama-cho Minami, Tottori 680-8553, Japan
| | - Tomohiko Kayano
- Laboratory of Veterinary Physiology, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101, Koyama-cho Minami, Tottori 680-8553, Japan
| | - Izumi Shibuya
- Laboratory of Veterinary Physiology, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101, Koyama-cho Minami, Tottori 680-8553, Japan
| |
Collapse
|
74
|
North RY, Lazaro TT, Dougherty PM. Ectopic Spontaneous Afferent Activity and Neuropathic Pain. Neurosurgery 2018; 65:49-54. [PMID: 31076785 DOI: 10.1093/neuros/nyy119] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/04/2018] [Indexed: 12/17/2022] Open
Affiliation(s)
- Robert Y North
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Tyler T Lazaro
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Patrick M Dougherty
- The Departments of Pain Medicine Research, The Division of Anesthesia, Critical Care and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
75
|
Djouhri L, Smith T, Ahmeda A, Alotaibi M, Weng X. Hyperpolarization-activated cyclic nucleotide-gated channels contribute to spontaneous activity in L4 C-fiber nociceptors, but not Aβ-non-nociceptors, after axotomy of L5-spinal nerve in the rat in vivo. Pain 2018; 159:1392-1402. [PMID: 29578948 DOI: 10.1097/j.pain.0000000000001224] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Peripheral neuropathic pain associated with partial nerve injury is believed to be driven partly by aberrant spontaneous activity (SA) in both injured and uninjured dorsal root ganglion (DRG) neurons. The underlying ionic mechanisms are not fully understood, but hyperpolarization-activated cyclic nucleotide-gated (HCN) channels which underlie the excitatory Ih current have been implicated in SA generation in axotomized A-fiber neurons after L5-spinal nerve ligation/axotomy (SNL/SNA). Here, using a modified model of SNA (mSNA) which involves, in addition to L5-SNA, loose ligation of the L4-spinal nerve with neuroinflammation-inducing chromic gut, we examined whether HCN channels also contribute to SA in the adjacent L4-neurons. Intracellular recordings from L4-DRG neurons in control rats, and L4-DRG neurons in mSNA rats were made using in vivo voltage- and current-clamp techniques. Compared with control, L4 C-nociceptors and Aβ-low-threshold mechanoreceptors (LTMs) exhibited SA 7 days after mSNA. This was accompanied, in C-nociceptors, by a significant increase in Ih amplitude, the percentage of Ih-expressing neurons, and Ih activation rate. Hyperpolarization-activated cyclic nucleotide-gated channel blockade with ZD7288 (10 mg/kg, intravenously) suppressed SA in C-nociceptors, but not Aβ-LTMs, and caused in C-nociceptors, membrane hyperpolarization and a decrease in Ih activation rate. Furthermore, intraplantar injection of ZD7288 (100 μM) was found to be as effective as gabapentin (positive control) in attenuating cold hypersensitivity in mSNA rats. These findings suggest that HCN channels contribute to nerve injury-induced SA in L4 C-nociceptors, but not Aβ-LTMs, and that ZD7288 exerts its analgesic effects by altering Ih activation properties and/or causing membrane hyperpolarization in L4 C-nociceptors.
Collapse
Affiliation(s)
- Laiche Djouhri
- Department of Physiology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Clinical and Molecular Pharmacology, Institute of Translational Medicine, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Trevor Smith
- Wolfson CARD, Neurorestoration Group, King's College London, London, United Kingdom
| | - Ahmad Ahmeda
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad Alotaibi
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Xiechuan Weng
- State Key Laboratory of Proteomics, Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| |
Collapse
|
76
|
Black BJ, Atmaramani R, Kumaraju R, Plagens S, Romero-Ortega M, Dussor G, Price TJ, Campbell ZT, Pancrazio JJ. Adult mouse sensory neurons on microelectrode arrays exhibit increased spontaneous and stimulus-evoked activity in the presence of interleukin-6. J Neurophysiol 2018; 120:1374-1385. [PMID: 29947589 DOI: 10.1152/jn.00158.2018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Following inflammation or injury, sensory neurons located in the dorsal root ganglia (DRG) may exhibit increased spontaneous and/or stimulus-evoked activity, contributing to chronic pain. Current treatment options for peripherally mediated chronic pain are highly limited, driving the development of cell- or tissue-based phenotypic (function-based) screening assays for peripheral analgesic and mechanistic lead discovery. Extant assays are often limited by throughput, content, use of tumorigenic cell lines, or tissue sources from immature developmental stages (i.e., embryonic or postnatal). Here, we describe a protocol for culturing adult mouse DRG neurons on substrate-integrated multiwell microelectrode arrays (MEAs). This approach enables multiplexed measurements of spontaneous as well as stimulus-evoked extracellular action potentials from large populations of cells. The DRG cultures exhibit stable spontaneous activity from 9 to 21 days in vitro. Activity is readily evoked by known chemical and physical agonists of sensory neuron activity such as capsaicin, bradykinin, PGE2, heat, and electrical field stimulation. Most importantly, we demonstrate that both spontaneous and stimulus-evoked activity may be potentiated by incubation with the inflammatory cytokine interleukin-6 (IL-6). Acute responsiveness to IL-6 is inhibited by treatment with a MAPK-interacting kinase 1/2 inhibitor, cercosporamide. In total, these findings suggest that adult mouse DRG neurons on multiwell MEAs are applicable to ongoing efforts to discover peripheral analgesic and their mechanisms of action. NEW & NOTEWORTHY This work describes methodologies for culturing spontaneously active adult mouse dorsal root ganglia (DRG) sensory neurons on microelectrode arrays. We characterize spontaneous and stimulus-evoked adult DRG activity over durations consistent with pharmacological interventions. Furthermore, persistent hyperexcitability could be induced by incubation with inflammatory cytokine IL-6 and attenuated with cercosporamide, an inhibitor of the IL-6 sensitization pathway. This constitutes a more physiologically relevant, moderate-throughput in vitro model for peripheral analgesic screening as well as mechanistic lead discovery.
Collapse
Affiliation(s)
- Bryan J Black
- Department of Bioengineering, The University of Texas at Dallas , Richardson, Texas
| | - Rahul Atmaramani
- Department of Bioengineering, The University of Texas at Dallas , Richardson, Texas
| | - Rajeshwari Kumaraju
- Department of Bioengineering, The University of Texas at Dallas , Richardson, Texas
| | - Sarah Plagens
- Department of Bioengineering, The University of Texas at Dallas , Richardson, Texas
| | - Mario Romero-Ortega
- Department of Bioengineering, The University of Texas at Dallas , Richardson, Texas
| | - Gregory Dussor
- School of Behavioral and Brain Sciences, The University of Texas at Dallas , Richardson, Texas
| | - Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas , Richardson, Texas
| | - Zachary T Campbell
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Richardson, Texas
| | - Joseph J Pancrazio
- Department of Bioengineering, The University of Texas at Dallas , Richardson, Texas
| |
Collapse
|
77
|
Nerve Injury-Induced Chronic Pain Is Associated with Persistent DNA Methylation Reprogramming in Dorsal Root Ganglion. J Neurosci 2018; 38:6090-6101. [PMID: 29875269 DOI: 10.1523/jneurosci.2616-17.2018] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 05/23/2018] [Accepted: 05/25/2018] [Indexed: 02/07/2023] Open
Abstract
Nerve injury-induced hyperactivity of primary sensory neurons in the dorsal root ganglion (DRG) contributes to chronic pain development, but the underlying epigenetic mechanisms remain poorly understood. Here we determined genome-wide changes in DNA methylation in the nervous system in neuropathic pain. Spinal nerve ligation (SNL), but not paclitaxel treatment, in male Sprague Dawley rats induced a consistent low-level hypomethylation in the CpG sites in the DRG during the acute and chronic phases of neuropathic pain. DNA methylation remodeling in the DRG occurred early after SNL and persisted for at least 3 weeks. SNL caused DNA methylation changes at 8% of CpG sites with prevailing hypomethylation outside of CpG islands, in introns, intergenic regions, and repetitive sequences. In contrast, SNL caused more gains of methylation in the spinal cord and prefrontal cortex. The DNA methylation changes in the injured DRGs recapitulated developmental reprogramming at the neonatal stage. Methylation reprogramming was correlated with increased gene expression variability. A diet deficient in methyl donors induced hypomethylation and pain hypersensitivity. Intrathecal administration of the DNA methyltransferase inhibitor RG108 caused long-lasting pain hypersensitivity. DNA methylation reprogramming in the DRG thus contributes to nerve injury-induced chronic pain. Restoring DNA methylation may represent a new therapeutic approach to treat neuropathic pain.SIGNIFICANCE STATEMENT Epigenetic mechanisms are critically involved in the transition from acute to chronic pain after nerve injury. However, genome-wide changes in DNA methylation in the nervous system and their roles in neuropathic pain development remain unclear. Here we used digital restriction enzyme analysis of methylation to quantitatively determine genome-wide DNA methylation changes caused by nerve injury. We showed that nerve injury caused DNA methylation changes at 8% of CpG sites with prevailing hypomethylation outside of CpG islands in the dorsal root ganglion. Reducing DNA methylation induced pain hypersensitivity, whereas increasing DNA methylation attenuated neuropathic pain. These findings extend our understanding of the epigenetic mechanism of chronic neuropathic pain and suggest new strategies to treat nerve injury-induced chronic pain.
Collapse
|
78
|
Djouhri L, Smith T, Alotaibi M, Weng X. Membrane potential oscillations are not essential for spontaneous firing generation in L4 Aβ-afferent neurons after L5 spinal nerve axotomy and are not mediated by HCN channels. Exp Physiol 2018; 103:1145-1156. [PMID: 29860719 DOI: 10.1113/ep087013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/23/2018] [Indexed: 01/01/2023]
Abstract
NEW FINDINGS What is the central question of this study? Is spontaneous activity (SA) in L4 dorsal root ganglion (DRG) neurons induced by L5 spinal nerve axotomy associated with membrane potential oscillations in these neurons, and if so, are these membrane oscillations mediated by HCN channels? What is the main finding and its importance? Unlike injured L5 DRG neurons, which have been shown to be incapable of firing spontaneously without membrane potential oscillations, membrane potential oscillations are not essential for SA generation in conducting 'uninjured' L4 neurons, and they are not mediated by HCN channels. These findings suggest that the underlying cellular mechanisms of SA in injured and 'uninjured' DRG neurons induced by spinal nerve injury are distinct. ABSTRACT The underlying cellular and molecular mechanisms of peripheral neuropathic pain are not fully understood. However, preclinical studies using animal models suggest that this debilitating condition is driven partly by aberrant spontaneous activity (SA) in injured and uninjured dorsal root ganglion (DRG) neurons, and that SA in injured DRG neurons is triggered by subthreshold membrane potential oscillations (SMPOs). Here, using in vivo intracellular recording from control L4-DRG neurons, and ipsilateral L4-DRG neurons in female Wistar rats that had previously undergone L5 spinal nerve axotomy (SNA), we examined whether conducting 'uninjured' L4-DRG neurons in SNA rats exhibit SMPOs, and if so, whether such SMPOs are associated with SA in those L4 neurons, and whether they are mediated by hyperpolarization-activated cyclic nucleotide gated (HCN) channels. We found that 7 days after SNA: (a) none of the control A- or C-fibre DRG neurons showed SMPOs or SA, but 50%, 43% and 0% of spontaneously active cutaneous L4 Aβ-low threshold mechanoreceptors, Aβ-nociceptors and C-nociceptors exhibited SMPOs, respectively, in SNA rats with established neuropathic pain behaviors; (b) neither SMPOs nor SA in L4 Aβ-neurons was suppressed by blocking HCN channels with ZD7288 (10 mg kg-1 , i.v.); and (c) there is a tendency for female rats to show greater pain hypersensitivity than male rats. These results suggest that SMPOs are linked to SA only in some of the conducting L4 Aβ-neurons, that such oscillations are not a prerequisite for SA generation in those L4 A- or C-fibre neurons, and that HCN channels are not involved in their electrogenesis.
Collapse
Affiliation(s)
- L Djouhri
- Department of Physiology, College of Medicine, Alfaisal University, PO Box 50927, Riyadh, 11533, Saudi Arabia
| | - T Smith
- Wolfson CARD, Neurorestoration Group, Hodgkin Building, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - M Alotaibi
- Department of Physiology, College of Medicine, King Saud University, PO Box 7805, Riyadh, 11472, Saudi Arabia
| | - X Weng
- Department of Neurobiology and State Key Laboratory of Proteomics, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| |
Collapse
|
79
|
Du X, Gao H, Jaffe D, Zhang H, Gamper N. M-type K + channels in peripheral nociceptive pathways. Br J Pharmacol 2018; 175:2158-2172. [PMID: 28800673 PMCID: PMC5980636 DOI: 10.1111/bph.13978] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 07/17/2017] [Accepted: 08/03/2017] [Indexed: 12/22/2022] Open
Abstract
Pathological pain is a hyperexcitability disorder. Since the excitability of a neuron is set and controlled by a complement of ion channels it expresses, in order to understand and treat pain, we need to develop a mechanistic insight into the key ion channels controlling excitability within the mammalian pain pathways and how these ion channels are regulated and modulated in various physiological and pathophysiological settings. In this review, we will discuss the emerging data on the expression in pain pathways, functional role and modulation of a family of voltage-gated K+ channels called 'M channels' (KCNQ, Kv 7). M channels are increasingly recognized as important players in controlling pain signalling, especially within the peripheral somatosensory system. We will also discuss the therapeutic potential of M channels as analgesic drug targets. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc/.
Collapse
Affiliation(s)
- Xiaona Du
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of EducationHebei Medical UniversityShijiazhuangChina
- The Key Laboratory of New Drug Pharmacology and ToxicologyShijiazhuangHebei ProvinceChina
| | - Haixia Gao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of EducationHebei Medical UniversityShijiazhuangChina
- The Key Laboratory of New Drug Pharmacology and ToxicologyShijiazhuangHebei ProvinceChina
- School of Biomedical Sciences, Faculty of Biological SciencesUniversity of LeedsLeedsUK
| | - David Jaffe
- Department of Biology, UTSA Neurosciences InstituteUniversity of Texas at San AntonioSan AntonioTXUSA
| | - Hailin Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of EducationHebei Medical UniversityShijiazhuangChina
- The Key Laboratory of New Drug Pharmacology and ToxicologyShijiazhuangHebei ProvinceChina
| | - Nikita Gamper
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of EducationHebei Medical UniversityShijiazhuangChina
- The Key Laboratory of New Drug Pharmacology and ToxicologyShijiazhuangHebei ProvinceChina
- School of Biomedical Sciences, Faculty of Biological SciencesUniversity of LeedsLeedsUK
| |
Collapse
|
80
|
Jayaraj ND, Bhattacharyya BJ, Belmadani AA, Ren D, Rathwell CA, Hackelberg S, Hopkins BE, Gupta HR, Miller RJ, Menichella DM. Reducing CXCR4-mediated nociceptor hyperexcitability reverses painful diabetic neuropathy. J Clin Invest 2018. [PMID: 29533926 DOI: 10.1172/jci92117] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [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 an intractable complication of diabetes that affects 25% of patients. PDN is characterized by neuropathic pain and small-fiber degeneration, accompanied by dorsal root ganglion (DRG) nociceptor hyperexcitability and loss of their axons within the skin. The molecular mechanisms underlying DRG nociceptor hyperexcitability and small-fiber degeneration in PDN are unknown. We hypothesize that chemokine CXCL12/CXCR4 signaling is central to this mechanism, as we have shown that CXCL12/CXCR4 signaling is necessary for the development of mechanical allodynia, a pain hypersensitivity behavior common in PDN. Focusing on DRG neurons expressing the sodium channel Nav1.8, we applied transgenic, electrophysiological, imaging, and chemogenetic techniques to test this hypothesis. In the high-fat diet mouse model of PDN, we were able to prevent and reverse mechanical allodynia and small-fiber degeneration by limiting CXCR4 signaling or neuronal excitability. This study reveals that excitatory CXCR4/CXCL12 signaling in Nav1.8-positive DRG neurons plays a critical role in the pathogenesis of mechanical allodynia and small-fiber degeneration in a mouse model of PDN. Hence, we propose that targeting CXCR4-mediated DRG nociceptor hyperexcitability is a promising therapeutic approach for disease-modifying treatments for this currently intractable and widespread affliction.
Collapse
Affiliation(s)
| | | | - Abdelhak A Belmadani
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Dongjun Ren
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Craig A Rathwell
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Brittany E Hopkins
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Herschel R Gupta
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Richard J Miller
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Daniela M Menichella
- Department of Neurology and.,Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
81
|
Dilute lidocaine suppresses ectopic neuropathic discharge in dorsal root ganglia without blocking axonal propagation: a new approach to selective pain control. Pain 2018. [DOI: 10.1097/j.pain.0000000000001205] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
82
|
Chen L, Huang J, Zhao P, Persson AK, Dib-Hajj FB, Cheng X, Tan A, Waxman SG, Dib-Hajj SD. Conditional knockout of Na V1.6 in adult mice ameliorates neuropathic pain. Sci Rep 2018; 8:3845. [PMID: 29497094 PMCID: PMC5832877 DOI: 10.1038/s41598-018-22216-w] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/19/2018] [Indexed: 01/09/2023] Open
Abstract
Voltage-gated sodium channels NaV1.7, NaV1.8 and NaV1.9 have been the focus for pain studies because their mutations are associated with human pain disorders, but the role of NaV1.6 in pain is less understood. In this study, we selectively knocked out NaV1.6 in dorsal root ganglion (DRG) neurons, using NaV1.8-Cre directed or adeno-associated virus (AAV)-Cre mediated approaches, and examined the specific contribution of NaV1.6 to the tetrodotoxin-sensitive (TTX-S) current in these neurons and its role in neuropathic pain. We report here that NaV1.6 contributes up to 60% of the TTX-S current in large, and 34% in small DRG neurons. We also show NaV1.6 accumulates at nodes of Ranvier within the neuroma following spared nerve injury (SNI). Although NaV1.8-Cre driven NaV1.6 knockout does not alter acute, inflammatory or neuropathic pain behaviors, AAV-Cre mediated NaV1.6 knockout in adult mice partially attenuates SNI-induced mechanical allodynia. Additionally, AAV-Cre mediated NaV1.6 knockout, mostly in large DRG neurons, significantly attenuates excitability of these neurons after SNI and reduces NaV1.6 accumulation at nodes of Ranvier at the neuroma. Together, NaV1.6 in NaV1.8-positive neurons does not influence pain thresholds under normal or pathological conditions, but NaV1.6 in large NaV1.8-negative DRG neurons plays an important role in neuropathic pain.
Collapse
Affiliation(s)
- Lubin Chen
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Jianying Huang
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Anna-Karin Persson
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Fadia B Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Xiaoyang Cheng
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Andrew Tan
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA. .,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA. .,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA.
| |
Collapse
|
83
|
Xie RG, Chu WG, Hu SJ, Luo C. Characterization of Different Types of Excitability in Large Somatosensory Neurons and Its Plastic Changes in Pathological Pain States. Int J Mol Sci 2018; 19:ijms19010161. [PMID: 29303989 PMCID: PMC5796110 DOI: 10.3390/ijms19010161] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 12/30/2017] [Accepted: 01/02/2018] [Indexed: 12/16/2022] Open
Abstract
Sensory neuron types have been distinguished by distinct morphological and transcriptional characteristics. Excitability is the most fundamental functional feature of neurons. Mathematical models described by Hodgkin have revealed three types of neuronal excitability based on the relationship between firing frequency and applied current intensity. However, whether natural sensory neurons display different functional characteristics in terms of excitability and whether this excitability type undergoes plastic changes under pathological pain states have remained elusive. Here, by utilizing whole-cell patch clamp recordings, behavioral and pharmacological assays, we demonstrated that large dorsal root ganglion (DRG) neurons can be classified into three classes and four subclasses based on their excitability patterns, which is similar to mathematical models raised by Hodgkin. Analysis of hyperpolarization-activated cation current (Ih) revealed different magnitude of Ih in different excitability types of large DRG neurons, with higher Ih in Class 2-1 than that in Class 1, 2-2 and 3. This indicates a crucial role of Ih in the determination of excitability type of large DRG neurons. More importantly, this pattern of excitability displays plastic changes and transition under pathological pain states caused by peripheral nerve injury. This study sheds new light on the functional characteristics of large DRG neurons and extends functional classification of large DRG neurons by integration of transcriptomic and morphological characteristics.
Collapse
Affiliation(s)
- Rou-Gang Xie
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an 710032, China.
| | - Wen-Guang Chu
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an 710032, China.
| | - San-Jue Hu
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an 710032, China.
| | - Ceng Luo
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
84
|
Chen Y, Huang LYM. A simple and fast method to image calcium activity of neurons from intact dorsal root ganglia using fluorescent chemical Ca 2+ indicators. Mol Pain 2017; 13:1744806917748051. [PMID: 29212403 PMCID: PMC5731619 DOI: 10.1177/1744806917748051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Chemical calcium indicators have been commonly used to monitor calcium (Ca2+) activity in cell bodies, i.e., somata, of isolated dorsal root ganglion neurons. Recent studies have shown that dorsal root ganglion somata play an essential role in soma–glia interactions and actively participate in the transmission of nociceptive signals. It is therefore desirable to develop methods to study Ca2+ activity in neurons and glia in intact dorsal root ganglia. In our previous studies, we found that incubation of intact dorsal root ganglia with acetoxymethyl dye resulted in efficient Ca2+ dye loading into glial cells but limited dye loading into neurons. Here, we introduce a useful method to load Ca2+ dyes in intact dorsal root ganglion neurons through electroporation. We found that electroporation greatly facilitated loading of Fluo-4 acetoxymethyl, Oregon green bapta-1-488 acetoxymethyl, and Fluo-4 pentapotassium salt into dorsal root ganglion neurons. In contrast, electroporation did not further facilitate dye loading into glia. Using electroporation followed by incubation of acetoxymethyl form Ca2+ dye, we can load acetoxymethyl Ca2+ dye well in both neurons and glia. With this approach, we found that inflammation induced by complete Freund’s adjuvant significantly increased the incidence of neuron–glia interactions in dorsal root ganglia. We also confirmed the actions of capsaicin and morphine on Ca2+ responses in dorsal root ganglion neurons. Thus, by promoting the loading of Ca2+ dye in neurons and glia through electroporation and incubation, Ca2+ activities in neurons and neuron–glia interactions can be well studied in intact dorsal root ganglia.
Collapse
Affiliation(s)
- Yong Chen
- 1 Department of Neuroscience, Cell Biology and Anatomy, 12338 University of Texas Medical Branch, Galveston , TX, USA
| | - Li-Yen M Huang
- 1 Department of Neuroscience, Cell Biology and Anatomy, 12338 University of Texas Medical Branch, Galveston , TX, USA
| |
Collapse
|
85
|
Adanina VO, Vesselkin NP. Efferent projections of dorsal root afferents in the spinal cord of the lamprey Lampetra fluviatilis. J EVOL BIOCHEM PHYS+ 2017. [DOI: 10.1134/s0022093017050088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
86
|
Cruccu G, Nurmikko TJ, Ernault E, Riaz FK, McBride WT, Haanpää M. Superiority of capsaicin 8% patch versus oral pregabalin on dynamic mechanical allodynia in patients with peripheral neuropathic pain. Eur J Pain 2017; 22:700-706. [PMID: 29194851 PMCID: PMC5887877 DOI: 10.1002/ejp.1155] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2017] [Indexed: 01/17/2023]
Abstract
Background Dynamic Mechanical Allodynia (DMA) is a typical symptom of neuropathic pain (NP). In a recent study, the capsaicin 8% patch was noninferior to pregabalin in overall peripheral NP relief. In this study, we report the comparison of the two treatments in relieving DMA. Methods In a randomized, open‐label, head‐to‐head, 8‐week study, 488 patients with peripheral NP were treated with the capsaicin 8% patch (one application) or an optimized dose of pregabalin. Assessments included the area and intensity of DMA, and the number of patients achieving complete resolution of DMA. Results At baseline, 253 patients in the capsaicin 8% patch group and 235 patients in the pregabalin group had DMA. From baseline to end of study, the change in DMA intensity was significantly in favour of the capsaicin 8% patch versus pregabalin [−0.63 (95% CI: −1.04, −0.23; p = 0.002)]. Similarly, the capsaicin 8% patch was superior to pregabalin in reducing the area of DMA [−39.5 cm2 (95% CI: −69.1, −10.0; p = 0.009)] from baseline to end of study. Overall, a greater proportion of patients had a complete resolution of allodynia with capsaicin 8% patch treatment compared with pregabalin treatment (24.1% vs. 12.3%; p = 0.001) at end of study. Conclusion Capsaicin 8% treatment was superior to pregabalin in reducing the intensity and area of DMA, and in the number of patients with complete resolution of DMA. Significance The superiority of a topical treatment over pregabalin in relieving DMA supports the view that both peripheral and central sensitization can mediate allodynia.
Collapse
Affiliation(s)
- G Cruccu
- Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | - T J Nurmikko
- The Walton Centre NHS Foundation Trust, Liverpool, UK
| | - E Ernault
- Astellas Pharma Inc., Leiden, The Netherlands
| | - F K Riaz
- Astellas Pharma Inc., Chertsey, UK
| | - W T McBride
- Belfast Health and Social Care Trust, Belfast, Northern Ireland
| | - M Haanpää
- Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
87
|
Liu CN, Berryman E, Zakur D, Shoieb AM, Pardo ID, Boucher M, Somps CJ, Bagi CM, Cook JC. A novel endpoint for the assessment of chemotherapy-induced peripheral neuropathy in rodents: biomechanical properties of peripheral nerve. J Appl Toxicol 2017; 38:193-200. [PMID: 28815646 DOI: 10.1002/jat.3513] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 01/09/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CiPN) is a frequent adverse effect in patients and a leading safety consideration in oncology drug development. Although behavioral assessment and microscopic examination of the nerves and dorsal root ganglia can be incorporated into toxicity studies to assess CiPN risk, more sensitive and less labor-intensive endpoints are often lacking. In this study, rats and mice administered vincristine (75 μg kg-1 day-1 , i.p., for 10 days in rats and 100 μg kg-1 day-1 , i.p., for 11 days in mice, respectively) were employed as the CiPN models. Behavioral changes were assessed during the dosing phase. At necropsy, the sural or sciatic nerve was harvested from the rats and mice, respectively, and assessed for mechanical and histopathological endpoints. It was found that the maximal load and the load/extension ratio were significantly decreased in the nerves collected from the animals dosed with vincristine compared with the vehicle-treated animals (P < 0.05). Additionally, the gait analysis revealed that the paw print areas were significantly increased in mice (P < 0.01), but not in rats following vincristine administration. Light microscopic histopathology of the nerves and dorsal root ganglia were unaffected by vincristine administration. We concluded that ex vivo mechanical properties of the nerves is a sensitive endpoint, providing a new method to predict CiPN in rodent. Gait analysis may also be a useful tool in these pre-clinical animal models.
Collapse
Affiliation(s)
- Chang-Ning Liu
- Worldwide Comparative Medicine, Pfizer Worldwide R&D, Groton, Connecticut, 06340, USA
| | - Edwin Berryman
- Worldwide Comparative Medicine, Pfizer Worldwide R&D, Groton, Connecticut, 06340, USA
| | - David Zakur
- Worldwide Comparative Medicine, Pfizer Worldwide R&D, Groton, Connecticut, 06340, USA
| | - Ahmed M Shoieb
- Drug Safety Research & Development, Pfizer Worldwide R&D, Groton, Connecticut, 06340, USA
| | - Ingrid D Pardo
- Drug Safety Research & Development, Pfizer Worldwide R&D, Groton, Connecticut, 06340, USA
| | - Magalie Boucher
- Drug Safety Research & Development, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139, USA
| | - Chris J Somps
- Drug Safety Research & Development, Pfizer Worldwide R&D, Groton, Connecticut, 06340, USA
| | - Chedo M Bagi
- Worldwide Comparative Medicine, Pfizer Worldwide R&D, Groton, Connecticut, 06340, USA
| | - Jon C Cook
- Drug Safety Research & Development, Pfizer Worldwide R&D, Groton, Connecticut, 06340, USA
| |
Collapse
|
88
|
Rat model of cancer-induced bone pain: changes in nonnociceptive sensory neurons in vivo. Pain Rep 2017; 2:e603. [PMID: 29392218 PMCID: PMC5741358 DOI: 10.1097/pr9.0000000000000603] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/15/2017] [Accepted: 04/25/2017] [Indexed: 01/31/2023] Open
Abstract
Nonnociceptive sensory neurons relate to transient episodes of intense pain that characterize neuropathic pain. They are involved in the peripheral sensitization and tactile hypersensitivity. Introduction: Clinical data on cancer-induced bone pain (CIBP) suggest extensive changes in sensory function. In a previous investigation of an animal model of CIBP, we have observed that changes in intrinsic membrane properties and excitability of dorsal root ganglion (DRG) nociceptive neurons correspond to mechanical allodynia and hyperalgesia. Objectives: To investigate the mechanisms underlying changes in nonnociceptive sensory neurons in this model, we have compared the electrophysiological properties of primary nonnociceptive sensory neurons at <1 and >2 weeks after CIBP model induction with properties in sham control animals. Methods: Copenhagen rats were injected with 106 MAT-LyLu rat prostate cancer cells into the distal femur epiphysis to generate a model of CIBP. After von Frey tactile measurement of mechanical withdrawal thresholds, the animals were prepared for acute electrophysiological recordings of mechanically sensitive neurons in the DRG in vivo. Results: The mechanical withdrawal threshold progressively decreased in CIBP model rats. At <1 week after model induction, there were no changes observed in nonnociceptive Aβ-fiber DRG neurons between CIBP model rats and sham rats. However, at >2 weeks, the Aβ-fiber low-threshold mechanoreceptors (LTMs) in CIBP model rats exhibited a slowing of the dynamics of action potential (AP) genesis, including wider AP duration and lower AP amplitude compared with sham rats. Furthermore, enhanced excitability of Aβ-fiber LTM neurons was observed as an excitatory discharge in response to intracellular injection of depolarizing current into the soma. Conclusion: After induction of the CIBP model, Aβ-fiber LTMs at >2 weeks but not <1 week had undergone changes in electrophysiological properties. Importantly, changes observed are consistent with observations in models of peripheral neuropathy. Thus, Aβ-fiber nonnociceptive primary sensory neurons might be involved in the peripheral sensitization and tumor-induced tactile hypersensitivity in CIBP.
Collapse
|
89
|
Liu DL, Wang X, Chu WG, Lu N, Han WJ, Du YK, Hu SJ, Bai ZT, Wu SX, Xie RG, Luo C. Chronic cervical radiculopathic pain is associated with increased excitability and hyperpolarization-activated current ( I h) in large-diameter dorsal root ganglion neurons. Mol Pain 2017; 13:1744806917707127. [PMID: 28587505 PMCID: PMC5466279 DOI: 10.1177/1744806917707127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cervical radiculopathic pain is a very common symptom that may occur with cervical
spondylosis. Mechanical allodynia is often associated with cervical radiculopathic pain
and is inadequately treated with current therapies. However, the precise mechanisms
underlying cervical radiculopathic pain-associated mechanical allodynia have remained
elusive. Compelling evidence from animal models suggests a role of large-diameter dorsal
root ganglion neurons and plasticity of spinal circuitry attached with Aβ fibers in
mediating neuropathic pain. Whether cervical radiculopathic pain condition induces plastic
changes of large-diameter dorsal root ganglion neurons and what mechanisms underlie these
changes are yet to be known. With combination of patch-clamp recording,
immunohistochemical staining, as well as behavioral surveys, we demonstrated that upon
chronic compression of C7/8 dorsal root ganglions, large-diameter cervical dorsal root
ganglion neurons exhibited frequent spontaneous firing together with hyperexcitability.
Quantitative analysis of hyperpolarization-activated cation current
(Ih) revealed that Ih was
greatly upregulated in large dorsal root ganglion neurons from cervical radiculopathic
pain rats. This increased Ih was supported by the enhanced
expression of hyperpolarization-activated, cyclic nucleotide-modulated channels subunit 3
in large dorsal root ganglion neurons. Blockade of Ih with
selective antagonist, ZD7288 was able to eliminate the mechanical allodynia associated
with cervical radiculopathic pain. This study sheds new light on the functional plasticity
of a specific subset of large-diameter dorsal root ganglion neurons and reveals a novel
mechanism that could underlie the mechanical allodynia associated with cervical
radiculopathy.
Collapse
Affiliation(s)
- Da-Lu Liu
- 1 Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an, China.,2 Department of Radiation Medicine, Faculty of Preventive Medicine, Fourth Military Medical University, Xi'an, China
| | - Xu Wang
- 1 Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an, China.,3 Research Center for Resource Polypeptide Drugs and College of Life Sciences, Yanan University, Yanan, China
| | - Wen-Guang Chu
- 1 Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an, China
| | - Na Lu
- 1 Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an, China.,4 ART Center, Northwest Women's and Children's Hospital, Xi'an, China
| | - Wen-Juan Han
- 1 Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an, China
| | - Yi-Kang Du
- 5 The First Brigade, Fourth Military Medical University, Xi'an, China
| | - San-Jue Hu
- 1 Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an, China
| | - Zhan-Tao Bai
- 3 Research Center for Resource Polypeptide Drugs and College of Life Sciences, Yanan University, Yanan, China
| | - Sheng-Xi Wu
- 1 Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an, China
| | - Rou-Gang Xie
- 1 Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an, China
| | - Ceng Luo
- 1 Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
90
|
Du X, Hao H, Yang Y, Huang S, Wang C, Gigout S, Ramli R, Li X, Jaworska E, Edwards I, Deuchars J, Yanagawa Y, Qi J, Guan B, Jaffe DB, Zhang H, Gamper N. Local GABAergic signaling within sensory ganglia controls peripheral nociceptive transmission. J Clin Invest 2017; 127:1741-1756. [PMID: 28375159 PMCID: PMC5409786 DOI: 10.1172/jci86812] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 02/02/2017] [Indexed: 01/05/2023] Open
Abstract
The integration of somatosensory information is generally assumed to be a function of the central nervous system (CNS). Here we describe fully functional GABAergic communication within rodent peripheral sensory ganglia and show that it can modulate transmission of pain-related signals from the peripheral sensory nerves to the CNS. We found that sensory neurons express major proteins necessary for GABA synthesis and release and that sensory neurons released GABA in response to depolarization. In vivo focal infusion of GABA or GABA reuptake inhibitor to sensory ganglia dramatically reduced acute peripherally induced nociception and alleviated neuropathic and inflammatory pain. In addition, focal application of GABA receptor antagonists to sensory ganglia triggered or exacerbated peripherally induced nociception. We also demonstrated that chemogenetic or optogenetic depolarization of GABAergic dorsal root ganglion neurons in vivo reduced acute and chronic peripherally induced nociception. Mechanistically, GABA depolarized the majority of sensory neuron somata, yet produced a net inhibitory effect on the nociceptive transmission due to the filtering effect at nociceptive fiber T-junctions. Our findings indicate that peripheral somatosensory ganglia represent a hitherto underappreciated site of somatosensory signal integration and offer a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Xiaona Du
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Han Hao
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Yuehui Yang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Sha Huang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Caixue Wang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Sylvain Gigout
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Rosmaliza Ramli
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- School of Dental Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Xinmeng Li
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Ewa Jaworska
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Ian Edwards
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Jim Deuchars
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine and Japan Science and Technology Agency, CREST, Maebashi, Japan
| | - Jinlong Qi
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Bingcai Guan
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - David B. Jaffe
- Department of Biology, UTSA Neurosciences Institute, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Nikita Gamper
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
91
|
Hunter CW, Yang A, Davis T. Selective Radiofrequency Stimulation of the Dorsal Root Ganglion (DRG) as a Method for Predicting Targets for Neuromodulation in Patients With Post Amputation Pain: A Case Series. Neuromodulation 2017; 20:708-718. [PMID: 28337820 DOI: 10.1111/ner.12595] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 01/10/2017] [Accepted: 01/30/2017] [Indexed: 12/17/2022]
Abstract
OBJECTIVE While spinal cord stimulation (SCS) has established itself as an accepted and validated treatment for neuropathic pain, there are a number of conditions where it has experienced less, long-term success: post amputee pain (PAP) being one of them. Dorsal root ganglion (DRG) stimulation has shown great promise, particularly in conditions where traditional SCS has fallen short. One major difference between DRG stimulation and traditional SCS is the ability to provide focal stimulation over targeted areas. While this may be a contributing factor to its superiority, it can also be a limitation insofar stimulating the wrong DRG(s) can lead to failure. This is particularly relevant in conditions like PAP where neuroplastic maladaptation occurs causing the pain to deviate from expected patterns, thus creating uncertainty and variability in predicting targets for stimulation. We propose selective radiofrequency (RF) stimulation of the DRG as a method for preoperatively predicting targets for neuromodulation in patients with PAP. METHODS We present four patients with PAP of the lower extremities. RF stimulation was used to selectively stimulate individual DRG's, creating areas of paresthesias to see which most closely correlated/overlapped with the painful area(s). RF stimulation to the DRG's that resulted in the desirable paresthesia coverage in the residual or the missing limb(s) was recorded as "positive." Trial DRG leads were placed based on the positive RF stimulation findings. RESULTS In each patient, stimulating one or more DRG(s) produced paresthesias patterns that were contradictory to know dermatomal patterns. Upon completion of a one-week trial all four patients reported 60-90% pain relief, with coverage over the painful areas, and opted for permanent implant. CONCLUSIONS Mapping the DRG via RF stimulation appears to provide improved accuracy for determining lead placement in the setting of PAP where pain patterns are known to deviate from conventional dermatomal mapping.
Collapse
Affiliation(s)
- Corey W Hunter
- Ainsworth Institute of Pain Management, New York, NY, USA
| | - Ajax Yang
- Department of Physical Medicine & Rehabilitation, Mount Sinai Hospital, New York, NY, USA
| | - Tim Davis
- Orthopedic Pain Specialists, Santa Monica, CA, USA
| |
Collapse
|
92
|
Senkowski D, Heinz A. Chronic pain and distorted body image: Implications for multisensory feedback interventions. Neurosci Biobehav Rev 2016; 69:252-9. [DOI: 10.1016/j.neubiorev.2016.08.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 08/01/2016] [Accepted: 08/06/2016] [Indexed: 12/14/2022]
|
93
|
HCN2 ion channels: basic science opens up possibilities for therapeutic intervention in neuropathic pain. Biochem J 2016; 473:2717-36. [DOI: 10.1042/bcj20160287] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/18/2016] [Indexed: 01/22/2023]
Abstract
Nociception — the ability to detect painful stimuli — is an invaluable sense that warns against present or imminent damage. In patients with chronic pain, however, this warning signal persists in the absence of any genuine threat and affects all aspects of everyday life. Neuropathic pain, a form of chronic pain caused by damage to sensory nerves themselves, is dishearteningly refractory to drugs that may work in other types of pain and is a major unmet medical need begging for novel analgesics. Hyperpolarisation-activated cyclic nucleotide (HCN)-modulated ion channels are best known for their fundamental pacemaker role in the heart; here, we review data demonstrating that the HCN2 isoform acts in an analogous way as a ‘pacemaker for pain’, in that its activity in nociceptive neurons is critical for the maintenance of electrical activity and for the sensation of chronic pain in pathological pain states. Pharmacological block or genetic deletion of HCN2 in sensory neurons provides robust pain relief in a variety of animal models of inflammatory and neuropathic pain, without any effect on normal sensation of acute pain. We discuss the implications of these findings for our understanding of neuropathic pain pathogenesis, and we outline possible future opportunities for the development of efficacious and safe pharmacotherapies in a range of chronic pain syndromes.
Collapse
|
94
|
Tibbs GR, Posson DJ, Goldstein PA. Voltage-Gated Ion Channels in the PNS: Novel Therapies for Neuropathic Pain? Trends Pharmacol Sci 2016; 37:522-542. [DOI: 10.1016/j.tips.2016.05.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/24/2016] [Accepted: 05/03/2016] [Indexed: 12/19/2022]
|
95
|
L5 spinal nerve axotomy induces sensitization of cutaneous L4 Aβ-nociceptive dorsal root ganglion neurons in the rat in vivo. Neurosci Lett 2016; 624:72-7. [DOI: 10.1016/j.neulet.2016.05.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 05/06/2016] [Accepted: 05/07/2016] [Indexed: 12/21/2022]
|
96
|
Dias QM, Prado WA. The lesion of dorsolateral funiculus changes the antiallodynic effect of the intrathecal muscimol and baclofen in distinct phases of neuropathic pain induced by spinal nerve ligation in rats. Brain Res Bull 2016; 124:103-15. [DOI: 10.1016/j.brainresbull.2016.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/31/2016] [Accepted: 04/04/2016] [Indexed: 12/20/2022]
|
97
|
Calvo M, Richards N, Schmid AB, Barroso A, Zhu L, Ivulic D, Zhu N, Anwandter P, Bhat MA, Court FA, McMahon SB, Bennett DLH. Altered potassium channel distribution and composition in myelinated axons suppresses hyperexcitability following injury. eLife 2016; 5:e12661. [PMID: 27033551 PMCID: PMC4841771 DOI: 10.7554/elife.12661] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 03/15/2016] [Indexed: 12/25/2022] Open
Abstract
Neuropathic pain following peripheral nerve injury is associated with hyperexcitability in damaged myelinated sensory axons, which begins to normalise over time. We investigated the composition and distribution of shaker-type-potassium channels (Kv1 channels) within the nodal complex of myelinated axons following injury. At the neuroma that forms after damage, expression of Kv1.1 and 1.2 (normally localised to the juxtaparanode) was markedly decreased. In contrast Kv1.4 and 1.6, which were hardly detectable in the naïve state, showed increased expression within juxtaparanodes and paranodes following injury, both in rats and humans. Within the dorsal root (a site remote from injury) we noted a redistribution of Kv1-channels towards the paranode. Blockade of Kv1 channels with α-DTX after injury reinstated hyperexcitability of A-fibre axons and enhanced mechanosensitivity. Changes in the molecular composition and distribution of axonal Kv1 channels, therefore represents a protective mechanism to suppress the hyperexcitability of myelinated sensory axons that follows nerve injury.
Collapse
Affiliation(s)
- Margarita Calvo
- Wolfson Centre for Age-Related Diseases, Kings College London, London, United Kingdom.,Departamento de Fisiologia, Facultad de Ciencias Biologicas- Pontificia Universidad Catolica de Chile, Santiago, Chile.,Departamento de Anestesiologia, Facultad de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Natalie Richards
- Wolfson Centre for Age-Related Diseases, Kings College London, London, United Kingdom
| | - Annina B Schmid
- School of Health and Rehabilitation Sciences, The University of Queensland, Brisbane, Australia.,Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Alejandro Barroso
- Wolfson Centre for Age-Related Diseases, Kings College London, London, United Kingdom.,Hospital Regional Universitario de Málaga. Servicio de Anestesiología, Málaga, Spain
| | - Lan Zhu
- Wolfson Centre for Age-Related Diseases, Kings College London, London, United Kingdom.,School of Allied Health Sciences, Faculty of Health and Life Sciences, De Montfort University, Leicester, United Kingdom
| | - Dinka Ivulic
- Departamento de Fisiologia, Facultad de Ciencias Biologicas- Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Ning Zhu
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Philipp Anwandter
- Departamento Ortopedia y Traumatologia, Facultad de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Manzoor A Bhat
- Department of Physiology, UT Health Science Center at San Antonio, San Antonio, United States.,School of Medicine, UT Health Science Center at San Antonio, San Antonio, United States
| | - Felipe A Court
- Center for Integrative Biology, Universidad Mayor, Santiago, Chile.,FONDAP, Geroscience Center for Brain Health and Metabolism, Santiago, Chile.,Millenium Nucleus for Regenerative Biology, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Stephen B McMahon
- Wolfson Centre for Age-Related Diseases, Kings College London, London, United Kingdom
| | - David L H Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
98
|
Radiotherapy Suppresses Bone Cancer Pain through Inhibiting Activation of cAMP Signaling in Rat Dorsal Root Ganglion and Spinal Cord. Mediators Inflamm 2016; 2016:5093095. [PMID: 26989332 PMCID: PMC4775803 DOI: 10.1155/2016/5093095] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 01/10/2016] [Accepted: 01/18/2016] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy is one of the major clinical approaches for treatment of bone cancer pain. Activation of cAMP-PKA signaling pathway plays important roles in bone cancer pain. Here, we examined the effects of radiotherapy on bone cancer pain and accompanying abnormal activation of cAMP-PKA signaling. Female Sprague-Dawley rats were used and received tumor cell implantation (TCI) in rat tibia (TCI cancer pain model). Some of the rats that previously received TCI treatment were treated with X-ray radiation (radiotherapy). Thermal hyperalgesia and mechanical allodynia were measured and used for evaluating level of pain caused by TCI treatment. PKA mRNA expression in dorsal root ganglion (DRG) was detected by RT-PCR. Concentrations of cAMP, IL-1β, and TNF-α as well as PKA activity in DRG and the spinal cord were measured by ELISA. The results showed that radiotherapy significantly suppressed TCI-induced thermal hyperalgesia and mechanical allodynia. The level of PKA mRNA in DRG, cAMP concentration and PKA activity in DRG and in the spinal cord, and concentrations of IL-1β and TNF-α in the spinal cord were significantly reduced by radiotherapy. In addition, radiotherapy also reduced TCI-induced bone loss. These findings suggest that radiotherapy may suppress bone cancer pain through inhibition of activation of cAMP-PKA signaling pathway in DRG and the spinal cord.
Collapse
|
99
|
Medici T, Shortland PJ. Effects of peripheral nerve injury on parvalbumin expression in adult rat dorsal root ganglion neurons. BMC Neurosci 2015; 16:93. [PMID: 26674138 PMCID: PMC4681077 DOI: 10.1186/s12868-015-0232-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 12/07/2015] [Indexed: 12/24/2022] Open
Abstract
Background Parvalbumin (PV) is a calcium binding protein that identifies a subpopulation of proprioceptive dorsal root ganglion (DRG) neurons. Calcitonin gene-related peptide (CGRP) is also expressed in a high proportion of muscle afferents but its relationship to PV is unclear. Little is known of the phenotypic responses of muscle afferents to nerve injury. Sciatic nerve axotomy or L5 spinal nerve ligation and section (SNL) lesions were used to explore these issues in adult rats using immunocytochemistry. Results In naive animals, the mean PV expression was 25 % of L4 or L5 dorsal root ganglion (DRG) neurons, and this was unchanged 2 weeks after sciatic nerve axotomy. Colocalization studies with the injury marker activating transcription factor 3 (ATF3) showed that approximately 24 % of PV neurons expressed ATF3 after sciatic nerve axotomy suggesting that PV may show a phenotypic switch from injured to uninjured neurons. This possibility was further assessed using the spinal nerve ligation (SNL) injury model where injured and uninjured neurons are located in different DRGs. Two weeks after L5 SNL there was no change in total PV staining and essentially all L5 PV neurons expressed ATF3. Additionally, there was no increase in PV-ir in the adjacent uninjured L4 DRG cells. Co-labelling of DRG neurons revealed that less than 2 % of PV neurons normally expressed CGRP and no colocalization was seen after injury. Conclusion These experiments clearly show that axotomy does not produce down regulation of PV protein in the DRG. Moreover, this lack of change is not due to a phenotypic switch in PV immunoreactive (ir) neurons, or de novo expression of PV-ir in uninjured neurons after nerve injury. These results further illustrate differences that occur when muscle afferents are injured as compared to cutaneous afferents.
Collapse
Affiliation(s)
- Tom Medici
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Newark Street, London, E1 2AT, UK. .,Queens Hospital, Romford, Essex, RM7 0AG, UK.
| | - Peter J Shortland
- School of Science and Health, Western Sydney University, Narellen Road, Campbelltown, NSW, 2560, Australia. .,Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Newark Street, London, E1 2AT, UK.
| |
Collapse
|
100
|
Laedermann CJ, Abriel H, Decosterd I. Post-translational modifications of voltage-gated sodium channels in chronic pain syndromes. Front Pharmacol 2015; 6:263. [PMID: 26594175 PMCID: PMC4633509 DOI: 10.3389/fphar.2015.00263] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/23/2015] [Indexed: 02/06/2023] Open
Abstract
In the peripheral sensory nervous system the neuronal expression of voltage-gated sodium channels (Navs) is very important for the transmission of nociceptive information since they give rise to the upstroke of the action potential (AP). Navs are composed of nine different isoforms with distinct biophysical properties. Studying the mutations associated with the increase or absence of pain sensitivity in humans, as well as other expression studies, have highlighted Nav1.7, Nav1.8, and Nav1.9 as being the most important contributors to the control of nociceptive neuronal electrogenesis. Modulating their expression and/or function can impact the shape of the AP and consequently modify nociceptive transmission, a process that is observed in persistent pain conditions. Post-translational modification (PTM) of Navs is a well-known process that modifies their expression and function. In chronic pain syndromes, the release of inflammatory molecules into the direct environment of dorsal root ganglia (DRG) sensory neurons leads to an abnormal activation of enzymes that induce Navs PTM. The addition of small molecules, i.e., peptides, phosphoryl groups, ubiquitin moieties and/or carbohydrates, can modify the function of Navs in two different ways: via direct physical interference with Nav gating, or via the control of Nav trafficking. Both mechanisms have a profound impact on neuronal excitability. In this review we will discuss the role of Protein Kinase A, B, and C, Mitogen Activated Protein Kinases and Ca++/Calmodulin-dependent Kinase II in peripheral chronic pain syndromes. We will also discuss more recent findings that the ubiquitination of Nav1.7 by Nedd4-2 and the effect of methylglyoxal on Nav1.8 are also implicated in the development of experimental neuropathic pain. We will address the potential roles of other PTMs in chronic pain and highlight the need for further investigation of PTMs of Navs in order to develop new pharmacological tools to alleviate pain.
Collapse
Affiliation(s)
- Cedric J. Laedermann
- F.M. Kirby Neurobiology Research Center, Boston Children’s Hospital, Harvard Medical School, BostonMA, USA
| | - Hugues Abriel
- Department of Clinical Research, University of BernBern, Switzerland
| | - Isabelle Decosterd
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV) and University of LausanneLausanne, Switzerland
- Department of Fundamental Neurosciences, University of LausanneLausanne, Switzerland
| |
Collapse
|