1
|
Tan ZY, Wu B, Su X, Zhou Y, Ji YH. Differential expression of slow and fast-repriming tetrodotoxin-sensitive sodium currents in dorsal root ganglion neurons. Front Mol Neurosci 2024; 16:1336664. [PMID: 38273939 PMCID: PMC10808659 DOI: 10.3389/fnmol.2023.1336664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 12/11/2023] [Indexed: 01/27/2024] Open
Abstract
Sodium channel Nav1.7 triggers the generation of nociceptive action potentials and is important in sending pain signals under physiological and pathological conditions. However, studying endogenous Nav1.7 currents has been confounded by co-expression of multiple sodium channel isoforms in dorsal root ganglion (DRG) neurons. In the current study, slow-repriming (SR) and fast-repriming (FR) tetrodotoxin-sensitive (TTX-S) currents were dissected electrophysiologically in small DRG neurons of both rats and mice. Three subgroups of small DRG neurons were identified based on the expression pattern of SR and FR TTX-S currents. A majority of rat neurons only expressed SR TTX-S currents, while a majority of mouse neurons expressed additional FR TTX-S currents. ProTx-II inhibited SR TTX-S currents with variable efficacy among DRG neurons. The expression of both types of TTX-S currents was higher in Isolectin B4-negative (IB4-) compared to Isolectin B4-positive (IB4+) neurons. Paclitaxel selectively increased SR TTX-S currents in IB4- neurons. In simulation experiments, the Nav1.7-expressing small DRG neuron displayed lower rheobase and higher frequency of action potentials upon threshold current injections compared to Nav1.6. The results suggested a successful dissection of endogenous Nav1.7 currents through electrophysiological manipulation that may provide a useful way to study the functional expression and pharmacology of endogenous Nav1.7 channels in DRG neurons.
Collapse
Affiliation(s)
- Zhi-Yong Tan
- Department of Pathophysiology, Hebei University School of Basic Medicine, Baoding, China
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Bin Wu
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Institute of Special Environment Medicine, Nantong University, Nantong, China
| | - Xiaolin Su
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - You Zhou
- Department of Physiology, Hebei University School of Basic Medicine, Baoding, China
| | - Yong-Hua Ji
- Department of Physiology, Hebei University School of Basic Medicine, Baoding, China
| |
Collapse
|
2
|
Makau CM, Towett PK, Kanui TI, Abelson KSP. Effects of inhibition of Nav1.3, Nav1.7, and Nav1.8 channels on pain-related behavior in Speke's hinge-back tortoise (Kinixys spekii). J Neurosci Res 2024; 102:e25274. [PMID: 38284848 DOI: 10.1002/jnr.25274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 10/10/2023] [Accepted: 10/25/2023] [Indexed: 01/30/2024]
Abstract
Comparative studies using reptiles as experimental animals in pain research could expand our knowledge on the evolution and adaptation of pain mechanisms. Currently, there are no data reported on the involvement of voltage-gated sodium ion channels on nociception in reptiles. The aim of this study was to investigate the involvement of Nav1.3, Nav1.7, and Nav1.8 ion channels in nociception in Speke's hinge-back tortoise. ICA 121341 (selective blocker for Nav1.1/Nav1.3), NAV 26 (selective blocker for Nav1.7), and A803467 (selective blocker for Nav1.8) were used to investigate the involvement of Nav1.3, Nav1.7, and Nav1.8, respectively. The chemicals were administered intracoelomically thirty minutes before the start of nociceptive tests. ICA 121341 did not cause a significant decrease in the time spent in pain-related behavior in all the nociceptive tests. NAV 26 and A8034667 caused a statistically significant decrease in the mean time spent in pain-related behavior in the formalin and capsaicin tests. Only A803467 caused a statistically significant increase in the mean latency to pain-related behavior in the hot plate test. NAV 26 and A803467 had no observable side effects. In conclusion, Nav1.7 and Nav1.8 are involved in the processing of chemically induced inflammatory pain in Speke's hinge back tortoise. In addition, Nav1.8 are also significantly involved in the development of thermal-induced pain-related behavior in this species of reptile. However, our results do not support the involvement of Nav1.3 on the development of chemical or thermal induced pain-related behavior in the Speke's hinge back tortoise.
Collapse
Affiliation(s)
- Christopher M Makau
- Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| | - Philemon K Towett
- Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| | - Titus I Kanui
- School of Agriculture and Veterinary Sciences, South Eastern Kenya University, Kitui, Kenya
| | - Klas S P Abelson
- Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
Zhao C, Zhou X, Shi X. The influence of Nav1.9 channels on intestinal hyperpathia and dysmotility. Channels (Austin) 2023; 17:2212350. [PMID: 37186898 DOI: 10.1080/19336950.2023.2212350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023] Open
Abstract
The Nav1.9 channel is a voltage-gated sodium channel. It plays a vital role in the generation of pain and the formation of neuronal hyperexcitability after inflammation. It is highly expressed in small diameter neurons of dorsal root ganglions and Dogiel II neurons in enteric nervous system. The small diameter neurons in dorsal root ganglions are the primary sensory neurons of pain conduction. Nav1.9 channels also participate in regulating intestinal motility. Functional enhancements of Nav1.9 channels to a certain extent lead to hyperexcitability of small diameter dorsal root ganglion neurons. The hyperexcitability of the neurons can cause visceral hyperalgesia. Intestinofugal afferent neurons and intrinsic primary afferent neurons in enteric nervous system belong to Dogiel type II neurons. Their excitability can also be regulated by Nav1.9 channels. The hyperexcitability of intestinofugal afferent neurons abnormally activate entero-enteric inhibitory reflexes. The hyperexcitability of intrinsic primary afferent neurons disturb peristaltic waves by abnormally activating peristaltic reflexes. This review discusses the role of Nav1.9 channels in intestinal hyperpathia and dysmotility.
Collapse
Affiliation(s)
- Chenyu Zhao
- Department of Gastroenterology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Medical Genetics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xi Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Xiaoliu Shi
- Department of Medical Genetics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
4
|
Yin JB, Liu HX, Dong QQ, Wu HH, Liang ZW, Fu JT, Zhao WJ, Hu HQ, Guo HW, Zhang T, Lu YC, Jin S, Wang XL, Cao BZ, Wang Z, Ding T. Correlative increasing expressions of KIF5b and Nav1.7 in DRG neurons of rats under neuropathic pain conditions. Physiol Behav 2023; 263:114115. [PMID: 36773735 DOI: 10.1016/j.physbeh.2023.114115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 02/11/2023]
Abstract
Nav1.7, one of tetrodotoxin-sensitive voltage-gated sodium channels, mainly expressed in the small diameter dorsal root ganglion (DRG) neurons. The expression and accumulation on neuronal membrane of Nav1.7 increased following peripheral tissue inflammation or nerve injury. However, the mechanisms for membrane accumulation of Nav1.7 remained unclear. We report that KIF5b, a highly expressed member of the kinesin-1 family in DRGs, promoted the translocation of Nav1.7 to the plasma membrane in DRG neurons of the rat. Following nociceptive behaviors in rats induced by peripheral spared nerve injury (SNI), synchronously increased KIF5b and Nav1.7 expressions were observed in DRGs. Immunohistochemistry staining demonstrated the co-expressions of KIF5b and Nav1.7 in the same DRG neurons. Immunoprecipitation experiments further confirmed the interactions between KIF5b and Nav1.7. Moreover, intrathecal injections of KIF5b shRNA moderated the SNI-induced both mechanical and thermal hyperalgesia. The rescued analgesic effects also alleviated SNI-induced anxiety-like behaviors. In sum, KIF5b was required for the membrane localizations of Nav1.7, which suggests a novel mechanism for the trafficking of Nav1.7 involved in neuropathic pain.
Collapse
Affiliation(s)
- Jun-Bin Yin
- Department of Neurology, the 960th Hospital of PLA, Jinan 250031, China; Institute of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China; Department of Anatomy, Histology and Embryology, The Fourth Military Medical University, Xi'an 710032, China
| | - Hai-Xia Liu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Jinan 250021, China
| | - Qin-Qin Dong
- Department of Neurology, the 960th Hospital of PLA, Jinan 250031, China; Department of Neurology, Jinzhou Medical University, Jinzhou 121000, China
| | - Huang-Hui Wu
- Department of Anesthesiology, Medical College of Xiamen University, Xiamen 361005, China
| | - Zhuo-Wen Liang
- Institute of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Jin-Tao Fu
- Department of Critical Care Medicine, Affiliated Yanzhou District Hospital of Jining Medical College, Jining 272100, China
| | - Wen-Jun Zhao
- Department of Anatomy, Histology and Embryology, The Fourth Military Medical University, Xi'an 710032, China
| | - Huai-Qiang Hu
- Department of Neurology, the 960th Hospital of PLA, Jinan 250031, China
| | - Hong-Wei Guo
- Department of Neurology, the 960th Hospital of PLA, Jinan 250031, China
| | - Ting Zhang
- Department of Anatomy, Histology and Embryology, The Fourth Military Medical University, Xi'an 710032, China
| | - Ya-Cheng Lu
- Department of Anatomy, Histology and Embryology, The Fourth Military Medical University, Xi'an 710032, China
| | - Shan Jin
- Department of Neurology, the 960th Hospital of PLA, Jinan 250031, China
| | - Xiao-Ling Wang
- Department of Neurology, the 960th Hospital of PLA, Jinan 250031, China
| | - Bing-Zhen Cao
- Department of Neurology, the 960th Hospital of PLA, Jinan 250031, China.
| | - Zhe Wang
- Institute of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China.
| | - Tan Ding
- Institute of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China; Department of Anatomy, Histology and Embryology, The Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
5
|
Xu L, Yang L, Wu Y, Wan X, Tang X, Xu Y, Chen Q, Liu Y, Liu S. Rac1/PAK1 signaling contributes to bone cancer pain by Regulation dendritic spine remodeling in rats. Mol Pain 2023; 19:17448069231161031. [PMID: 36938611 PMCID: PMC10028669 DOI: 10.1177/17448069231161031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
Bone cancer pain (BCP) is severe chronic pain caused by tumor metastasis to the bones, often resulting in significant skeletal remodeling and fractures. Currently, there is no curative treatment. Therefore, insight into the underlying mechanisms could guide the development of mechanism-based therapeutic strategies for BCP. We speculated that Rac1/PAK1 signaling plays a critical role in the development of BCP. Tumor cells implantation (TCI) into the tibial cavity resulted in bone cancer-associated mechanical allodynia. Golgi staining revealed changes in the excitatory synaptic structure of WDR (Wide-dynamic range) neurons in the spinal cord, including increased postsynaptic density (PSD) length and thickness, and width of the cleft. Behavioral and western blotting test revealed that the development and persistence of pain correlated with Rac1/PAK1 signaling activation in primary sensory neurons. Intrathecal injection of NSC23766, a Rac1 inhibitor, reduced the persistence of BCP as well as reversed the remodeling of dendrites. Therefore, we concluded that activation of the Rac1/PAK1 signaling pathway in the spinal cord plays an important role in the development of BCP through remodeling of dendritic spines. Modulation of the Rac1/PAK1 pathway may be a potential strategy for BCP treatment.
Collapse
Affiliation(s)
- Lingfei Xu
- Jiangsu Province Key Laboratory of
Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia
Application Technology, NMPA Key Laboratory for Research and Evaluation of
Narcotic and Psychotropic Drugs, Xuzhou Medical
University, Jiangsu, China
| | - Long Yang
- Jiangsu Province Key Laboratory of
Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia
Application Technology, NMPA Key Laboratory for Research and Evaluation of
Narcotic and Psychotropic Drugs, Xuzhou Medical
University, Jiangsu, China
| | - Yan Wu
- Jiangsu Province Key Laboratory of
Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia
Application Technology, NMPA Key Laboratory for Research and Evaluation of
Narcotic and Psychotropic Drugs, Xuzhou Medical
University, Jiangsu, China
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou
Medical University, Jiangsu, China
| | - Xinxin Wan
- Department of Anesthesiology, Nanjing Drum Tower
Hospital, Jiangsu, China
| | - Xihui Tang
- Jiangsu Province Key Laboratory of
Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia
Application Technology, NMPA Key Laboratory for Research and Evaluation of
Narcotic and Psychotropic Drugs, Xuzhou Medical
University, Jiangsu, China
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou
Medical University, Jiangsu, China
| | - Yuqing Xu
- Jiangsu Province Key Laboratory of
Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia
Application Technology, NMPA Key Laboratory for Research and Evaluation of
Narcotic and Psychotropic Drugs, Xuzhou Medical
University, Jiangsu, China
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou
Medical University, Jiangsu, China
| | - Qingsong Chen
- Jiangsu Province Key Laboratory of
Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia
Application Technology, NMPA Key Laboratory for Research and Evaluation of
Narcotic and Psychotropic Drugs, Xuzhou Medical
University, Jiangsu, China
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou
Medical University, Jiangsu, China
| | - Yuepeng Liu
- Institute of Xuzhou Medical
Science, Jiangsu, China
| | - Su Liu
- Jiangsu Province Key Laboratory of
Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia
Application Technology, NMPA Key Laboratory for Research and Evaluation of
Narcotic and Psychotropic Drugs, Xuzhou Medical
University, Jiangsu, China
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou
Medical University, Jiangsu, China
| |
Collapse
|
6
|
Elleman AV, Du Bois J. Chemical and Biological Tools for the Study of Voltage-Gated Sodium Channels in Electrogenesis and Nociception. Chembiochem 2022; 23:e202100625. [PMID: 35315190 PMCID: PMC9359671 DOI: 10.1002/cbic.202100625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/22/2022] [Indexed: 12/17/2022]
Abstract
The malfunction and misregulation of voltage-gated sodium channels (NaV s) underlie in large part the electrical hyperexcitability characteristic of chronic inflammatory and neuropathic pain. NaV s are responsible for the initiation and propagation of electrical impulses (action potentials) in cells. Tissue and nerve injury alter the expression and localization of multiple NaV isoforms, including NaV 1.1, 1.3, and 1.6-1.9, resulting in aberrant action potential firing patterns. To better understand the role of NaV regulation, localization, and trafficking in electrogenesis and pain pathogenesis, a number of chemical and biological reagents for interrogating NaV function have been advanced. The development and application of such tools for understanding NaV physiology are the focus of this review.
Collapse
Affiliation(s)
- Anna V Elleman
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - J Du Bois
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
7
|
Wei N, Yu Y, Yang Y, Wang XL, Zhong ZJ, Chen XF, Yu YQ. Inhibitions and Down-Regulation of Motor Protein Eg5 Expression in Primary Sensory Neurons Reveal a Novel Therapeutic Target for Pathological Pain. Neurotherapeutics 2022; 19:1401-1413. [PMID: 35764763 PMCID: PMC9587155 DOI: 10.1007/s13311-022-01263-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2022] [Indexed: 10/17/2022] Open
Abstract
The motor protein Eg5, known as kif11 or kinesin-5, interacts with adjacent microtubules in the mitotic spindle and plays essential roles in cell division, yet the function of Eg5 in mature postmitotic neurons remains largely unknown. In this study, we investigated the contribution and molecular mechanism of Eg5 in pathological pain. Pharmacological inhibition of Eg5 and a specific shRNA-expressing viral vector reversed complete Freund's adjuvant (CFA)-induced pain and abrogated vanilloid receptor subtype 1 (VR1) expression in dorsal root ganglion (DRG) neurons. In the dorsal root, Eg5 inhibition promoted VR1 axonal transport and decreased VR1 expression. In the spinal cord, Eg5 inhibition suppressed VR1 expression in axon terminals and impaired synapse formation in superficial laminae I/II. Finally, we showed that Eg5 is necessary for PI3K/Akt signalling-mediated VR1 membrane trafficking and pathological pain. The present study provides compelling evidence of a noncanonical function of Eg5 in primary sensory neurons. These results suggest that Eg5 may be a potential therapeutic target for intractable pain.
Collapse
Affiliation(s)
- Na Wei
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Baqiao, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Yang Yu
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Baqiao, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Yan Yang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Baqiao, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Xiao-Liang Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Baqiao, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Zhen-Juan Zhong
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Baqiao, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Xue-Feng Chen
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Baqiao, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Yao-Qing Yu
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Baqiao, Xi'an, 710038, China.
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China.
| |
Collapse
|
8
|
Kaburagi H, Nagata T, Enomoto M, Hirai T, Ohyagi M, Ihara K, Yoshida-Tanaka K, Ebihara S, Asada K, Yokoyama H, Okawa A, Yokota T. Systemic DNA/RNA heteroduplex oligonucleotide administration for regulating the gene expression of dorsal root ganglion and sciatic nerve. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 28:910-919. [PMID: 35694210 PMCID: PMC9167871 DOI: 10.1016/j.omtn.2022.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 05/03/2022] [Indexed: 11/24/2022]
Abstract
Neuropathic pain, a heterogeneous condition, affects 7%–10% of the general population. To date, efficacious and safe therapeutic approaches remain limited. Antisense oligonucleotide (ASO) therapy has opened the door to treat spinal muscular atrophy, with many ongoing clinical studies determining its therapeutic utility. ASO therapy for neuropathic pain and peripheral nerve disease requires efficient gene delivery and knockdown in both the dorsal root ganglion (DRG) and sciatic nerve, key tissues for pain signaling. We previously developed a new DNA/RNA heteroduplex oligonucleotide (HDO) technology that achieves highly efficient gene knockdown in the liver. Here, we demonstrated that intravenous injection of HDO, comprising an ASO and its complementary RNA conjugated to α-tocopherol, silences endogenous gene expression more than 2-fold in the DRG, and sciatic nerve with higher potency, efficacy, and broader distribution than ASO alone. Of note, we observed drastic target suppression in all sizes of neuronal DRG populations by in situ hybridization. Our findings establish HDO delivery as an investigative and potentially therapeutic platform for neuropathic pain and peripheral nerve disease.
Collapse
|
9
|
Glutamine Maintains Satellite Glial Cells Growth and Survival in Culture. Neurochem Res 2022; 47:3635-3646. [PMID: 35522367 DOI: 10.1007/s11064-022-03614-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/10/2022] [Accepted: 04/19/2022] [Indexed: 10/18/2022]
Abstract
Satellite glial cells (SGCs) tightly surround neurons and modulate sensory transmission in dorsal root ganglion (DRG). At present, the biological property of primary SGCs in culture deserves further investigation. To reveal the key factor for SGCs growth and survival, we examined the effects of different culture supplementations containing Dulbecco's Modified Eagle Medium (DMEM)/F12, DMEM high glucose (HG) or Neurobasal-A (NB). CCK-8 proliferation assay showed an increased proliferation of SGCs in DMEM/F12 and DMEM/HG, but not in NB medium. Bax, AnnexinV, and propidium iodide (PI) staining results showed that NB medium caused cell death and apoptosis. We showed that glutamine was over 2.5 mM in DMEM/F12 and DMEM/HG, whereas it was absence in NB medium. Interestingly, exogenous glutamine application significantly reversed the poor proliferation and cell death of SGCs in NB medium. These findings demonstrated that DMEM/F12 medium was optimal to get high-purity SGCs. Glutamine was the key molecule to maintain SGCs growth and survival in culture. Here, we provided a novel approach to get high-purity SGCs by changing the key component of culture medium. Our study shed a new light on understanding the biological property and modulation of glial cells of primary sensory ganglia.
Collapse
|
10
|
Scorpion Venom peptide, AGAP inhibits TRPV1 and potentiates the analgesic effect of lidocaine. Heliyon 2021; 7:e08560. [PMID: 35005265 PMCID: PMC8715296 DOI: 10.1016/j.heliyon.2021.e08560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/04/2021] [Accepted: 12/01/2021] [Indexed: 11/20/2022] Open
Abstract
The current study was designed to test the hypothesis that BmK AGAP (AGAP) potentiates the analgesic effect of lidocaine. The chronic constrictive injury was performed on 72 rats to induce a rapid onset and long-lasting pain. The rats were randomly assigned to one of six groups; Group A (n = 12) received an intrathecal administration of saline, Group B (n = 12) received an intrathecal injection of lidocaine, Group C (n = 12) received an intrathecal administration of AGAP, Group D, E, and F (n = 12 each) received an intrathecal administration of lidocaine 0.005 mg/ml + AGAP 25, 50, 100 μg/kg respectively. The von Frey filaments were used to assess mechanical allodynia. Nav1.7 and TRPV1 currents were recorded by the whole-cell aspiration patch-clamp technique, and KCNQ2/3 currents were recorded by the whole-cell drilling patch-clamp technique. The whole-cell aspiration patch-clamp technique showed that AGAP inhibited TRPV1and KCNQ2/3 currents and increased the analgesic effect of lidocaine. AGAP may have a synergistic effect with lidocaine which demonstrates a potential therapeutic approach for optimizing post-operative analgesia.
Collapse
|
11
|
Chávez-Castillo M, Ortega Á, Cudris-Torres L, Duran P, Rojas M, Manzano A, Garrido B, Salazar J, Silva A, Rojas-Gomez DM, De Sanctis JB, Bermúdez V. Specialized Pro-Resolving Lipid Mediators: The Future of Chronic Pain Therapy? Int J Mol Sci 2021; 22:ijms221910370. [PMID: 34638711 PMCID: PMC8509014 DOI: 10.3390/ijms221910370] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 12/15/2022] Open
Abstract
Chronic pain (CP) is a severe clinical entity with devastating physical and emotional consequences for patients, which can occur in a myriad of diseases. Often, conventional treatment approaches appear to be insufficient for its management. Moreover, considering the adverse effects of traditional analgesic treatments, specialized pro-resolving lipid mediators (SPMs) have emerged as a promising alternative for CP. These include various bioactive molecules such as resolvins, maresins, and protectins, derived from ω-3 polyunsaturated fatty acids (PUFAs); and lipoxins, produced from ω-6 PUFAs. Indeed, SPMs have been demonstrated to play a central role in the regulation and resolution of the inflammation associated with CP. Furthermore, these molecules can modulate neuroinflammation and thus inhibit central and peripheral sensitizations, as well as long-term potentiation, via immunomodulation and regulation of nociceptor activity and neuronal pathways. In this context, preclinical and clinical studies have evidenced that the use of SPMs is beneficial in CP-related disorders, including rheumatic diseases, migraine, neuropathies, and others. This review integrates current preclinical and clinical knowledge on the role of SPMs as a potential therapeutic tool for the management of patients with CP.
Collapse
Affiliation(s)
- Mervin Chávez-Castillo
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.C.-C.); (Á.O.); (P.D.); (M.R.); (A.M.); (B.G.); (J.S.); (A.S.)
| | - Ángel Ortega
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.C.-C.); (Á.O.); (P.D.); (M.R.); (A.M.); (B.G.); (J.S.); (A.S.)
| | - Lorena Cudris-Torres
- Programa de Psicología, Fundación Universitaria del Área Andina sede Valledupar, Valledupar 200001, Colombia;
| | - Pablo Duran
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.C.-C.); (Á.O.); (P.D.); (M.R.); (A.M.); (B.G.); (J.S.); (A.S.)
| | - Milagros Rojas
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.C.-C.); (Á.O.); (P.D.); (M.R.); (A.M.); (B.G.); (J.S.); (A.S.)
| | - Alexander Manzano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.C.-C.); (Á.O.); (P.D.); (M.R.); (A.M.); (B.G.); (J.S.); (A.S.)
| | - Bermary Garrido
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.C.-C.); (Á.O.); (P.D.); (M.R.); (A.M.); (B.G.); (J.S.); (A.S.)
| | - Juan Salazar
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.C.-C.); (Á.O.); (P.D.); (M.R.); (A.M.); (B.G.); (J.S.); (A.S.)
| | - Aljadis Silva
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.C.-C.); (Á.O.); (P.D.); (M.R.); (A.M.); (B.G.); (J.S.); (A.S.)
| | - Diana Marcela Rojas-Gomez
- Escuela de Nutrición y Dietética, Facultad de Medicina, Universidad Andres Bello, Santiago 8370035, Chile;
| | - Juan B. De Sanctis
- Institute of Molecular and Translational Medicine, Palacký University Olomouc, 77900 Olomouc, Czech Republic;
| | - Valmore Bermúdez
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 080002, Colombia
- Correspondence:
| |
Collapse
|
12
|
A Buthus martensii Karsch scorpion sting targets Nav1.7 in mice and mimics a phenotype of human chronic pain. Pain 2021; 163:e202-e214. [PMID: 34252912 DOI: 10.1097/j.pain.0000000000002397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/28/2021] [Indexed: 11/25/2022]
Abstract
GAIN and loss-of-function mutations in Nav1.7 cause chronic pain and pain insensitivity, respectively. The preferential expression of Nav1.7 in peripheral nervous system and its role in human pain signaling make Nav1.7 a promising target for next-generation pain therapeutics. However, pharmacological agents have not fully recapitulated these pain phenotypes, and, due to the lack of subtype-selective molecular modulators, the role of Nav1.7 in the perception of pain remains poorly understood. Scorpion venom is an excellent source of bioactive peptides that modulate various ion channels, including voltage-gated sodium (Nav) channels . Here, we demonstrate that Buthus martensii Karsch scorpion venom (BV) elicits pain responses in mice through direct enhancement of Nav1.7 activity, and have identified that Makatoxin-3, an α-like toxin as a critical component for BV-mediated effects on Nav1.7. Blocking other Nav subtypes did not eliminate BV-evoked pain responses, supporting the pivotal role of Nav1.7 in BV-induced pain . Makatoxin-3 acts on the S3-S4 loop of voltage sensor domain IV (VSD4) of Nav1.7, which causes a hyperpolarizing shift in the steady-state fast inactivation and impairs inactivation kinetics. We also determined the key residues and structure-function relationships for the toxin-channel interactions, which are distinct from those of other well-studied α-toxins. This study not only reveals a new mechanism underlying BV-evoked pain, but also enriches our knowledge of key structural elements of scorpion toxins that are pivotal for toxin-Nav1.7 interaction, which facilitates the design of novel Nav1.7 selective modulators.
Collapse
|
13
|
Lin W, Zhang WW, Lyu N, Cao H, Xu WD, Zhang YQ. Growth Differentiation Factor-15 Produces Analgesia by Inhibiting Tetrodotoxin-Resistant Nav1.8 Sodium Channel Activity in Rat Primary Sensory Neurons. Neurosci Bull 2021; 37:1289-1302. [PMID: 34076854 PMCID: PMC8423960 DOI: 10.1007/s12264-021-00709-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/10/2021] [Indexed: 01/01/2023] Open
Abstract
Growth differentiation factor 15 (GDF-15) is a member of the transforming growth factor-β superfamily. It is widely distributed in the central and peripheral nervous systems. Whether and how GDF-15 modulates nociceptive signaling remains unclear. Behaviorally, we found that peripheral GDF-15 significantly elevated nociceptive response thresholds to mechanical and thermal stimuli in naïve and arthritic rats. Electrophysiologically, we demonstrated that GDF-15 decreased the excitability of small-diameter dorsal root ganglia (DRG) neurons. Furthermore, GDF-15 concentration-dependently suppressed tetrodotoxin-resistant sodium channel Nav1.8 currents, and shifted the steady-state inactivation curves of Nav1.8 in a hyperpolarizing direction. GDF-15 also reduced window currents and slowed down the recovery rate of Nav1.8 channels, suggesting that GDF-15 accelerated inactivation and slowed recovery of the channel. Immunohistochemistry results showed that activin receptor-like kinase-2 (ALK2) was widely expressed in DRG medium- and small-diameter neurons, and some of them were Nav1.8-positive. Blockade of ALK2 prevented the GDF-15-induced inhibition of Nav1.8 currents and nociceptive behaviors. Inhibition of PKA and ERK, but not PKC, blocked the inhibitory effect of GDF-15 on Nav1.8 currents. These results suggest a functional link between GDF-15 and Nav1.8 in DRG neurons via ALK2 receptors and PKA associated with MEK/ERK, which mediate the peripheral analgesia of GDF-15.
Collapse
Affiliation(s)
- Wei Lin
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Wen-Wen Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Ning Lyu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - Hong Cao
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Wen-Dong Xu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China. .,Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Yu-Qiu Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China. .,Department of Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
14
|
Zhang LL, Qiu J, Hong JR, Xu XQ, Zhang GQ, Li G. Magnolol attenuates inflammatory pain by inhibiting sodium currents in mouse dorsal root ganglion neurons. Inflammopharmacology 2021; 29:869-877. [PMID: 34021831 DOI: 10.1007/s10787-021-00809-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 04/05/2021] [Indexed: 10/21/2022]
Abstract
Voltage-gated sodium channels are currently recognized as one of the targets of analgesics. Magnolol (Mag), an active component isolated from Magnolia officinalis, has been reported to exhibit analgesic effects. The objective of this study was to investigate whether the analgesic effect of Mag was associated with blocking Na+ channels. Inflammatory pain was induced by the injection of carrageenan into the hind paw of mice. Mag was administered orally. Mechanical hyperanalgesia was evaluated by using von Frey filaments. Na+ currents and neuronal excitability in acutely isolated mouse dorsal root ganglion (DRG) neurons were recorded with the whole-cell patch clamp technique. Results showed that Mag (10 ~ 40 mg/kg) dose-dependently inhibited the paw edema and reduced mechanical pain in the inflammatory animal model. Injection of carrageenan significantly increased the amplitudes of TTX-sensitive and TTX-resistant Na+ currents. Compared with the carrageenan group, Mag inhibited the upregulation of two types of Na+ currents induced by carrageenan in a dose-dependent manner. Mag 40 mg/kg shifted the inactivation curves of two types of Na+ currents to hyperpolarization and returned to normal animal level without changing their activation curves. Mag 40 mg/kg significantly reduced the percentage of cells firing multiple spikes and inhibited the neuronal hyperexcitability induced by carrageenan. Our data suggest that the analgesic effect of Mag may be associated with a decreased neuronal excitability by blocking Na+ current.
Collapse
Affiliation(s)
- Lu-Lu Zhang
- Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jie Qiu
- Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jiang-Ru Hong
- Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiu-Qi Xu
- Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Guang-Qin Zhang
- Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Guang Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research of Southwest, Medical University, Luzhou, 646000, China.
| |
Collapse
|
15
|
Modulations of Na v1.8 and Na v1.9 Channels in Monosodium Urate-Induced Gouty Arthritis in Mice. Inflammation 2021; 44:1405-1415. [PMID: 33515125 DOI: 10.1007/s10753-021-01425-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/03/2021] [Accepted: 01/21/2021] [Indexed: 10/22/2022]
Abstract
The aim of the present study was to observe the changes of TTX-R, Nav1.8, and Nav1.9 Na+ currents in MSU-induced gouty arthritis mice, and to explore the possibility of Nav1.8 and Nav1.9 channels as potential targets for gout pain treatment. Acute gouty arthritis was induced by monosodium urate (MSU) in mice. Swelling degree was evaluated by measuring the circumference of the ankle joint. Mechanical allodynia was assessed by applying the electronic von Frey. Na+ currents were recorded by patch-clamp techniques in acute isolated dorsal root ganglion (DRG) neurons. MSU treatment significantly increased the swelling degree of ankle joint and decreased the mechanical pain threshold. The amplitude of TTX-R Na+ current was significantly increased and reached its peak on the 4th day after injection of MSU. For TTX-R Na+ channel subunits, Nav1.8 current density was significantly increased, but Nav1.9 current density was markedly decreased after MSU treatment. MSU treatment shifted the steady-state activation curves of TTX-R Na+ channel, Nav1.8 and Nav1.9 channels, and the inactivation curves of TTX-R Na+ channel and Nav1.8 channels to the depolarizing direction, but did not affect the inactivation curve of Nav1.9 channel. Compared with the normal group, the recovery of Nav1.8 channel was faster, while that of Nav1.9 channel was slower. The recovery of TTX-R Na+ channel remained unchanged after MSU treatment. Additionally, MSU treatment increased DRG neurons excitability by reducing action potential threshold. Nav1.8 channel, not Nav1.9 channel, may be involved in MSU-induced gout pain by increasing nerve excitability.
Collapse
|
16
|
Richard SA, Kampo S, Sackey M, Hechavarria ME, Buunaaim ADB. The Pivotal Potentials of Scorpion Buthus Martensii Karsch-Analgesic-Antitumor Peptide in Pain Management and Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:4234273. [PMID: 33178316 PMCID: PMC7647755 DOI: 10.1155/2020/4234273] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/13/2020] [Accepted: 10/20/2020] [Indexed: 01/26/2023]
Abstract
Scorpion Buthus martensii Karsch -analgesic-antitumor peptide (BmK AGAP) has been used to treat diseases like tetanus, tuberculosis, apoplexy, epilepsy, spasm, migraine headaches, rheumatic pain, and cancer in China. AGAP is a distinctive long-chain scorpion toxin with a molecular mass of 7142 Da and composed of 66 amino acids cross-linked by four disulfide bridges. Voltage-gated sodium channels (VGSCs) are present in excitable membranes and partakes in essential roles in action potentials generation as compared to the significant function of voltage-gated calcium channels (VGCCs). A total of nine genes (Nav1.1-Nav1.9) have been recognized to encode practical sodium channel isoforms. Nav1.3, Nav1.7, Nav1.8, and Nav1.9 have been recognized as potential targets for analgesics. Nav1.8 and Nav1.9 are associated with nociception initiated by inflammation signals in the neuronal pain pathway, while Nav1.8 is fundamental for neuropathic pain at low temperatures. AGAP has a sturdy inhibitory influence on both viscera and soma pain. AGAP potentiates the effects of MAPK inhibitors on neuropathic as well as inflammation-associated pain. AGAP downregulates the secretion of phosphorylated p38, phosphorylated JNK, and phosphorylated ERK 1/2 in vitro. AGAP has an analgesic activity which may be an effective therapeutic agent for pain management because of its downregulation of PTX3 via NF-κB and Wnt/beta-catenin signaling pathway. In cancers like colon cancer, breast cancer, lymphoma, and glioma, rAGAP was capable of blocking the proliferation. Thus, AGAP is a promising therapy for these tumors. Nevertheless, research is needed with other tumors.
Collapse
Affiliation(s)
- Seidu A. Richard
- Department of Medicine, Princefield University, P.O. Box MA128, Ho, Ghana
| | - Sylvanus Kampo
- Department of Anesthesia and Critical Care, School of Medicine, University of Health and Allied Sciences, Ho, Ghana
| | - Marian Sackey
- Department of Pharmacy, Ho Teaching Hospital, P.O. Box MA-374, Ho, Ghana
| | | | - Alexis D. B. Buunaaim
- Department of Surgery, School of Medicine and Health Science, University for Development Studies, Tamale, Ghana
| |
Collapse
|
17
|
Alsaloum M, Higerd GP, Effraim PR, Waxman SG. Status of peripheral sodium channel blockers for non-addictive pain treatment. Nat Rev Neurol 2020; 16:689-705. [PMID: 33110213 DOI: 10.1038/s41582-020-00415-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 12/15/2022]
Abstract
The effective and safe treatment of pain is an unmet health-care need. Current medications used for pain management are often only partially effective, carry dose-limiting adverse effects and are potentially addictive, highlighting the need for improved therapeutic agents. Most common pain conditions originate in the periphery, where dorsal root ganglion and trigeminal ganglion neurons feed pain information into the CNS. Voltage-gated sodium (NaV) channels drive neuronal excitability and three subtypes - NaV1.7, NaV1.8 and NaV1.9 - are preferentially expressed in the peripheral nervous system, suggesting that their inhibition might treat pain while avoiding central and cardiac adverse effects. Genetic and functional studies of human pain disorders have identified NaV1.7, NaV1.8 and NaV1.9 as mediators of pain and validated them as targets for pain treatment. Consequently, multiple NaV1.7-specific and NaV1.8-specific blockers have undergone clinical trials, with others in preclinical development, and the targeting of NaV1.9, although hampered by technical constraints, might also be moving ahead. In this Review, we summarize the clinical and preclinical literature describing compounds that target peripheral NaV channels and discuss the challenges and future prospects for the field. Although the potential of peripheral NaV channel inhibition for the treatment of pain has yet to be realized, this remains a promising strategy to achieve non-addictive analgesia for multiple pain conditions.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA.,Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT, USA
| | - Grant P Higerd
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA
| | - Philip R Effraim
- Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.,Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA. .,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA. .,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
18
|
Painful and painless mutations of SCN9A and SCN11A voltage-gated sodium channels. Pflugers Arch 2020; 472:865-880. [PMID: 32601768 PMCID: PMC7351857 DOI: 10.1007/s00424-020-02419-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/25/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022]
Abstract
Chronic pain is a global problem affecting up to 20% of the world’s population and has a significant economic, social and personal cost to society. Sensory neurons of the dorsal root ganglia (DRG) detect noxious stimuli and transmit this sensory information to regions of the central nervous system (CNS) where activity is perceived as pain. DRG neurons express multiple voltage-gated sodium channels that underlie their excitability. Research over the last 20 years has provided valuable insights into the critical roles that two channels, NaV1.7 and NaV1.9, play in pain signalling in man. Gain of function mutations in NaV1.7 cause painful conditions while loss of function mutations cause complete insensitivity to pain. Only gain of function mutations have been reported for NaV1.9. However, while most NaV1.9 mutations lead to painful conditions, a few are reported to cause insensitivity to pain. The critical roles these channels play in pain along with their low expression in the CNS and heart muscle suggest they are valid targets for novel analgesic drugs.
Collapse
|
19
|
Li C, Ban M, Bai F, Chen J, Jin X, Song Y. Anti-Nociceptive and Anti-Inflammation Effect Mechanisms of Mutants of Syb-prII, a Recombinant Neurotoxic Polypeptide. Toxins (Basel) 2019; 11:E699. [PMID: 31805689 PMCID: PMC6949983 DOI: 10.3390/toxins11120699] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/14/2019] [Accepted: 11/15/2019] [Indexed: 01/09/2023] Open
Abstract
Syb-prII, a recombinant neurotoxic polypeptide, has analgesic effects with medicinal value. Previous experiments indicated that Syb-prII displayed strong analgesic activities. Therefore, a series of in vivo and vitro experiments were designed to investigate the analgesic and anti-inflammatory properties and possible mechanisms of Syb-prII. The results showed that administered Syb-prII-1 and Syb-prII-2 (0.5, 1, 2.0 mg/kg, i.v.) to mice significantly reduced the time of licking, biting, or flicking of paws in two phases in formalin-induced inflammatory nociception. Syb-prII-1 inhibited xylene-induced auricular swelling in a dose-dependent manner. The inhibitory effect of 2.0 mg/kg Syb-prII-1 on the ear swelling model was comparable to that of 200 mg/kg aspirin. In addition, the ELISA and Western blot analysis suggested that Syb-prII-1 and Syb-prII-2 may exert an analgesic effect by inhibiting the expression of Nav1.8 and the phosphorylation of ERK, JNK, and P38. Syb-prII-1 markedly suppressed the expression of IL-1β, IL-6, and TNF-α of mice in formalin-induced inflammatory nociception. We used the patch-clamp technique and investigated the effect of Syb-prII-1 on TTX-resistant sodium channel currents in acutely isolated rat DRG neurons. The results showed that Syb-prII-1 can significantly down regulate TTX-resistant sodium channel currents. In conclusion, Syb-prII mutants may alleviate inflammatory pain by significantly inhibiting the expression of Nav1.8, mediated by the phosphorylation of MAPKs and significant inhibition of TTX-resistant sodium channel currents.
Collapse
Affiliation(s)
| | | | | | | | | | - Yongbo Song
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China; (C.L.); (M.B.); (F.B.); (J.C.); (X.J.)
| |
Collapse
|
20
|
The Role of Maresins in Inflammatory Pain: Function of Macrophages in Wound Regeneration. Int J Mol Sci 2019; 20:ijms20235849. [PMID: 31766461 PMCID: PMC6928948 DOI: 10.3390/ijms20235849] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/12/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023] Open
Abstract
Although acute inflammatory responses are host-protective and generally self-limited, unresolved and delayed resolution of acute inflammation can lead to further tissue damage and chronic inflammation. The mechanism of pain induction under inflammatory conditions has been studied extensively; however, the mechanism of pain resolution is not fully understood. The resolution of inflammation is a biosynthetically active process, involving specialized pro-resolving mediators (SPMs). In particular, maresins (MaRs) are synthesized from docosahexaenoic acid (DHA) by macrophages and have anti-inflammatory and pro-resolving capacities as well as tissue regenerating and pain-relieving properties. A new class of macrophage-derived molecules—MaR conjugates in tissue regeneration (MCTRs)—has been reported to regulate phagocytosis and the repair and regeneration of damaged tissue. Macrophages not only participate in the biosynthesis of SPMs, but also play an important role in phagocytosis. They exhibit different phenotypes categorized as proinflammatory M1-like phenotypes and anti-inflammatory M2 phenotypes that mediate both harmful and protective functions, respectively. However, the signaling mechanisms underlying macrophage functions and phenotypic changes have not yet been fully established. Recent studies report that MaRs help resolve inflammatory pain by enhancing macrophage phagocytosis and shifting cytokine release to the anti-inflammatory M2 phenotypes. Consequently, this review elucidated the characteristics of MaRs and macrophages, focusing on the potent action of MaRs to enhance the M2 macrophage phenotype profiles that possess the ability to alleviate inflammatory pain.
Collapse
|
21
|
Spider Knottin Pharmacology at Voltage-Gated Sodium Channels and Their Potential to Modulate Pain Pathways. Toxins (Basel) 2019; 11:toxins11110626. [PMID: 31671792 PMCID: PMC6891507 DOI: 10.3390/toxins11110626] [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] [Received: 09/24/2019] [Revised: 10/24/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated sodium channels (NaVs) are a key determinant of neuronal signalling. Neurotoxins from diverse taxa that selectively activate or inhibit NaV channels have helped unravel the role of NaV channels in diseases, including chronic pain. Spider venoms contain the most diverse array of inhibitor cystine knot (ICK) toxins (knottins). This review provides an overview on how spider knottins modulate NaV channels and describes the structural features and molecular determinants that influence their affinity and subtype selectivity. Genetic and functional evidence support a major involvement of NaV subtypes in various chronic pain conditions. The exquisite inhibitory properties of spider knottins over key NaV subtypes make them the best lead molecules for the development of novel analgesics to treat chronic pain.
Collapse
|
22
|
de la Peña JBI, Song JJ, Campbell ZT. RNA control in pain: Blame it on the messenger. WILEY INTERDISCIPLINARY REVIEWS-RNA 2019; 10:e1546. [PMID: 31090211 DOI: 10.1002/wrna.1546] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/17/2019] [Accepted: 04/22/2019] [Indexed: 12/12/2022]
Abstract
mRNA function is meticulously controlled. We provide an overview of the integral role that posttranscriptional controls play in the perception of painful stimuli by sensory neurons. These specialized cells, termed nociceptors, precisely regulate mRNA polarity, translation, and stability. A growing body of evidence has revealed that targeted disruption of mRNAs and RNA-binding proteins robustly diminishes pain-associated behaviors. We propose that the use of multiple independent regulatory paradigms facilitates robust temporal and spatial precision of protein expression in response to a range of pain-promoting stimuli. This article is categorized under: RNA in Disease and Development > RNA in Disease Translation > Translation Regulation RNA Turnover and Surveillance > Regulation of RNA Stability.
Collapse
Affiliation(s)
- June Bryan I de la Peña
- Department of Biological Sciences and the Center for Advanced Pain Studies, University of Texas, Dallas, Richardson, Texas
| | - Jane J Song
- Department of Biological Sciences and the Center for Advanced Pain Studies, University of Texas, Dallas, Richardson, Texas
| | - Zachary T Campbell
- Department of Biological Sciences and the Center for Advanced Pain Studies, University of Texas, Dallas, Richardson, Texas
| |
Collapse
|
23
|
Fattori V, Pinho-Ribeiro FA, Staurengo-Ferrari L, Borghi SM, Rossaneis AC, Casagrande R, Verri WA. The specialised pro-resolving lipid mediator maresin 1 reduces inflammatory pain with a long-lasting analgesic effect. Br J Pharmacol 2019; 176:1728-1744. [PMID: 30830967 DOI: 10.1111/bph.14647] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 01/30/2019] [Accepted: 02/11/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Maresin 1 (MaR1) is a specialised pro-resolving lipid mediator with anti-inflammatory and analgesic activities. In this study, we addressed the modulation of peripheral and spinal cord cells by MaR1 in the context of inflammatory pain. EXPERIMENTAL APPROACH Mice were treated with MaR1 before intraplantar injection of carrageenan or complete Freund's adjuvant (CFA). Mechanical hyperalgesia was assessed using the electronic von Frey and thermal hyperalgesia using a hot plate. Spinal cytokine production and NF-κB activation were determined by ELISA and astrocytes and microglia activation by RT-qPCR and immunofluorescence. CGRP release by dorsal root ganglia (DRG) neurons was determined by EIA. Neutrophil and macrophage recruitment were determined by immunofluorescence, flow cytometry, and colorimetric methods. Trpv1 and Nav1.8 expression and calcium imaging of DRG neurons were determined by RT-qPCR and Fluo-4AM respectively. KEY RESULTS MaR1 reduced carrageenan- and CFA-induced mechanical and thermal hyperalgesia and neutrophil and macrophage recruitment proximal to CGRP+ fibres in the paw skin. Moreover, MaR1 reduced NF-κB activation, IL-1β and TNF-α production, and spinal cord glial cells activation. In the DRG, MaR1 reduced CFA-induced Nav1.8 and Trpv1 mRNA expression and calcium influx and capsaicin-induced release of CGRP by DRG neurons. CONCLUSIONS AND IMPLICATIONS MaR1 reduced DRG neurons activation and CGRP release explaining, at least in part, its analgesic and anti-inflammatory effects. The enduring analgesic and anti-inflammatory effects and also post-treatment activity of MaR1 suggest that specialised pro-resolving lipid mediators have potential as a new class of drugs for the treatment of inflammatory pain.
Collapse
Affiliation(s)
- Victor Fattori
- Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Brazil
| | - Felipe A Pinho-Ribeiro
- Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Brazil
| | | | - Sergio M Borghi
- Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Brazil
| | - Ana C Rossaneis
- Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Brazil
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Centre of Health Science, Londrina State University, Londrina, Brazil
| | - Waldiceu A Verri
- Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Brazil
| |
Collapse
|
24
|
Antisense oligonucleotides selectively suppress target RNA in nociceptive neurons of the pain system and can ameliorate mechanical pain. Pain 2019; 159:139-149. [PMID: 28976422 DOI: 10.1097/j.pain.0000000000001074] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
There is an urgent need for better treatments for chronic pain, which affects more than 1 billion people worldwide. Antisense oligonucleotides (ASOs) have proven successful in treating children with spinal muscular atrophy, a severe infantile neurological disorder, and several ASOs are currently being tested in clinical trials for various neurological disorders. Here, we characterize the pharmacodynamic activity of ASOs in spinal cord and dorsal root ganglia (DRG), key tissues for pain signaling. We demonstrate that activity of ASOs lasts up to 2 months after a single intrathecal bolus dose. Interestingly, comparison of subcutaneous, intracerebroventricular, and intrathecal administration shows that DRGs are targetable by systemic and central delivery of ASOs, while target reduction in the spinal cord is achieved only after direct central delivery. Upon detailed characterization of ASO activity in individual cell populations in DRG, we observe robust target suppression in all neuronal populations, thereby establishing that ASOs are effective in the cell populations involved in pain propagation. Furthermore, we confirm that ASOs are selective and do not modulate basal pain sensation. We also demonstrate that ASOs targeting the sodium channel Nav1.7 induce sustained analgesia up to 4 weeks. Taken together, our findings support the idea that ASOs possess the required pharmacodynamic properties, along with a long duration of action beneficial for treating pain.
Collapse
|
25
|
Zhang XL, Cao XY, Lai RC, Xie MX, Zeng WA. Puerarin Relieves Paclitaxel-Induced Neuropathic Pain: The Role of Na v1.8 β1 Subunit of Sensory Neurons. Front Pharmacol 2019; 9:1510. [PMID: 30666203 PMCID: PMC6330330 DOI: 10.3389/fphar.2018.01510] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
Currently there is no effective treatment available for clinical patients suffering from neuropathic pain induced by chemotherapy paclitaxel. Puerarin is a major isoflavonoid extracted from the Chinese medical herb kudzu root, which has been used for treatment of cardiovascular disorders and brain injury. Here, we found that puerarin dose-dependently alleviated paclitaxel-induced neuropathic pain. At the same time, puerarin preferentially reduced the excitability and blocked the voltage-gated sodium (Nav) channels of dorsal root ganglion (DRG) neurons from paclitaxel-induced neuropathic pain rats. Furthermore, puerarin was a more potent blocker of tetrodotoxin-resistant (TTX-R) Nav channels than of tetrodotoxin-sensitive (TTX-S) Nav channels in chronic pain rats’ DRG neurons. In addition, puerarin had a stronger blocking effect on Nav1.8 channels in DRG neurons of neuropathic pain rats and β1 subunit siRNA can abolish this selective blocking effect on Nav1.8. Together, these results suggested that puerarin may preferentially block β1 subunit of Nav1.8 in sensory neurons contributed to its anti-paclitaxel induced neuropathic pain effect.
Collapse
Affiliation(s)
- Xiao-Long Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xian-Ying Cao
- College of Food Science and Technology, Hainan University, Haikou, China.,State Key Laboratory of Marine Resources Utilization in South China Sea, Hainan University, Haikou, China
| | - Ren-Chun Lai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Man-Xiu Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wei-An Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
26
|
Li CL, Yu Y, He T, Wang RR, Geng KW, Du R, Luo WJ, Wei N, Wang XL, Wang Y, Yang Y, Yu YQ, Chen J. Validating Rat Model of Empathy for Pain: Effects of Pain Expressions in Social Partners. Front Behav Neurosci 2018; 12:242. [PMID: 30386220 PMCID: PMC6199527 DOI: 10.3389/fnbeh.2018.00242] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/26/2018] [Indexed: 01/09/2023] Open
Abstract
Pain can be socially transferred between familiar rats due to empathic responses. To validate rat model of empathy for pain, effects of pain expressions in a cagemate demonstrator (CD) in pain on empathic pain responses in a naïve cagemate observer (CO) after 30 min priming dyadic social interactions (PDSI) were evaluated. The CD rats were prepared with four pain models: bee venom (BV), formalin, complete Freund's adjuvant (CFA), and spared nerve injury (SNI). Both BV and formalin tests are characterized by displayable and eye-identifiable spontaneous pain-related behaviors (SPRB) immediately after treatment, while CFA and SNI models are characterized by delayed occurrence of evoked pain hypersensitivity but with less eye-identifiable SPRB. After 30 min PDSI with a CD immediately after BV and formalin, respectively, the empathic mechanical pain hypersensitivity (EMPH) could be identified at both hind paws in CO rats. The BV—or formalin-induced EMPH in CO rats lasted for 4–5 h until full recovery. However, EMPH failed to develop in CO after socially interacting with a CD immediately after CFA, or 2 h after BV when SPRB completely disappeared. The CO's EMPH was partially relieved when socially interacting with an analgecized CD whose SPRB had been significantly suppressed. Moreover, repeated exposures to a CD in pain could enhance EMPH in CO. Finally, social transfer of pain hypersensitivity was also identified in CO who was being co-housed in pairs with a conspecific treated with CFA or SNI. The results suggest that development of EMPH in CO rats would be determined not only by extent of familiarity but also by visually identifiable pain expressions in the social partners during short period of PDSI. However, the visually unidentifiable pain can also be transferred to naïve cagemate when being co-housed in pairs with a distressed conspecific. In summary, the vicariously social contagion of pain between familiar rats is dependent upon not only expressions of pain in social partners but also the time that dyads spent in social communications. The rat model of empathy for pain is a highly stable, reproducible and valid model for studying the neural mechanisms of empathy in lower animals.
Collapse
Affiliation(s)
- Chun-Li Li
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, China
| | - Yang Yu
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, China
| | - Ting He
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, China
| | - Rui-Rui Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, China
| | - Kai-Wen Geng
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Rui Du
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wen-Jun Luo
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Na Wei
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, China
| | - Xiao-Liang Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, China
| | - Yang Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, China
| | - Yan Yang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, China
| | - Yao-Qing Yu
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, China
| | - Jun Chen
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, China
| |
Collapse
|
27
|
Nencini S, Ringuet M, Kim DH, Chen YJ, Greenhill C, Ivanusic JJ. Mechanisms of nerve growth factor signaling in bone nociceptors and in an animal model of inflammatory bone pain. Mol Pain 2017; 13:1744806917697011. [PMID: 28326938 PMCID: PMC5407668 DOI: 10.1177/1744806917697011] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Sequestration of nerve growth factor has been used successfully in the management of pain in animal models of bone disease and in human osteoarthritis. However, the mechanisms of nerve growth factor-induced bone pain and its role in modulating inflammatory bone pain remain to be determined. In this study, we show that nerve growth factor receptors (TrkA and p75) and some other nerve growth factor-signaling molecules (TRPV1 and Nav1.8, but not Nav1.9) are expressed in substantial proportions of rat bone nociceptors. We demonstrate that nerve growth factor injected directly into rat tibia rapidly activates and sensitizes bone nociceptors and produces acute behavioral responses with a similar time course. The nerve growth factor-induced changes in the activity and sensitivity of bone nociceptors we report are dependent on signaling through the TrkA receptor, but are not affected by mast cell stabilization. We failed to show evidence for longer term changes in expression of TrkA, TRPV1, Nav1.8 or Nav1.9 in the soma of bone nociceptors in a rat model of inflammatory bone pain. Thus, retrograde transport of NGF/TrkA and increased expression of some of the common nerve growth factor signaling molecules do not appear to be important for the maintenance of inflammatory bone pain. The findings are relevant to understand the basis of nerve growth factor sequestration and other therapies directed at nerve growth factor signaling, in managing pain in bone disease.
Collapse
Affiliation(s)
- Sara Nencini
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Australia
| | - Mitchell Ringuet
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Australia
| | - Dong-Hyun Kim
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Australia
| | - Yu-Jen Chen
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Australia
| | - Claire Greenhill
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Australia
| | - Jason J Ivanusic
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Australia
| |
Collapse
|
28
|
Sodium Channel Na v1.8 Underlies TTX-Resistant Axonal Action Potential Conduction in Somatosensory C-Fibers of Distal Cutaneous Nerves. J Neurosci 2017; 37:5204-5214. [PMID: 28450535 DOI: 10.1523/jneurosci.3799-16.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 04/03/2017] [Accepted: 04/04/2017] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium (NaV) channels are responsible for the initiation and conduction of action potentials within primary afferents. The nine NaV channel isoforms recognized in mammals are often functionally divided into tetrodotoxin (TTX)-sensitive (TTX-s) channels (NaV1.1-NaV1.4, NaV1.6-NaV1.7) that are blocked by nanomolar concentrations and TTX-resistant (TTX-r) channels (NaV1.8 and NaV1.9) inhibited by millimolar concentrations, with NaV1.5 having an intermediate toxin sensitivity. For small-diameter primary afferent neurons, it is unclear to what extent different NaV channel isoforms are distributed along the peripheral and central branches of their bifurcated axons. To determine the relative contribution of TTX-s and TTX-r channels to action potential conduction in different axonal compartments, we investigated the effects of TTX on C-fiber-mediated compound action potentials (C-CAPs) of proximal and distal peripheral nerve segments and dorsal roots from mice and pigtail monkeys (Macaca nemestrina). In the dorsal roots and proximal peripheral nerves of mice and nonhuman primates, TTX reduced the C-CAP amplitude to 16% of the baseline. In contrast, >30% of the C-CAP was resistant to TTX in distal peripheral branches of monkeys and WT and NaV1.9-/- mice. In nerves from NaV1.8-/- mice, TTX-r C-CAPs could not be detected. These data indicate that NaV1.8 is the primary isoform underlying TTX-r conduction in distal axons of somatosensory C-fibers. Furthermore, there is a differential spatial distribution of NaV1.8 within C-fiber axons, being functionally more prominent in the most distal axons and terminal regions. The enrichment of NaV1.8 in distal axons may provide a useful target in the treatment of pain of peripheral origin.SIGNIFICANCE STATEMENT It is unclear whether individual sodium channel isoforms exert differential roles in action potential conduction along the axonal membrane of nociceptive, unmyelinated peripheral nerve fibers, but clarifying the role of sodium channel subtypes in different axonal segments may be useful for the development of novel analgesic strategies. Here, we provide evidence from mice and nonhuman primates that a substantial portion of the C-fiber compound action potential in distal peripheral nerves, but not proximal nerves or dorsal roots, is resistant to tetrodotoxin and that, in mice, this effect is mediated solely by voltage-gated sodium channel 1.8 (NaV1.8). The functional prominence of NaV1.8 within the axonal compartment immediately proximal to its termination may affect strategies targeting pain of peripheral origin.
Collapse
|
29
|
K + Channel Modulatory Subunits KChIP and DPP Participate in Kv4-Mediated Mechanical Pain Control. J Neurosci 2017; 37:4391-4404. [PMID: 28330877 DOI: 10.1523/jneurosci.1619-16.2017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 03/14/2017] [Accepted: 03/16/2017] [Indexed: 11/21/2022] Open
Abstract
The K+ channel pore-forming subunit Kv4.3 is expressed in a subset of nonpeptidergic nociceptors within the dorsal root ganglion (DRG), and knockdown of Kv4.3 selectively induces mechanical hypersensitivity, a major symptom of neuropathic pain. K+ channel modulatory subunits KChIP1, KChIP2, and DPP10 are coexpressed in Kv4.3+ DRG neurons, but whether they participate in Kv4.3-mediated pain control is unknown. Here, we show the existence of a Kv4.3/KChIP1/KChIP2/DPP10 complex (abbreviated as the Kv4 complex) in the endoplasmic reticulum and cell surface of DRG neurons. After intrathecal injection of a gene-specific antisense oligodeoxynucleotide to knock down the expression of each component in the Kv4 complex, mechanical hypersensitivity develops in the hindlimbs of rats in parallel with a reduction in all components in the lumbar DRGs. Electrophysiological data further indicate that the excitability of nonpeptidergic nociceptors is enhanced. The expression of all Kv4 complex components in DRG neurons is downregulated following spinal nerve ligation (SNL). To rescue Kv4 complex downregulation, cDNA constructs encoding Kv4.3, KChIP1, and DPP10 were transfected into the injured DRGs (defined as DRGs with injured spinal nerves) of living SNL rats. SNL-evoked mechanical hypersensitivity was attenuated, accompanied by a partial recovery of Kv4.3, KChIP1, and DPP10 surface levels in the injured DRGs. By showing an interdependent regulation among components in the Kv4 complex, this study demonstrates that K+ channel modulatory subunits KChIP1, KChIP2, and DPP10 participate in Kv4.3-mediated mechanical pain control. Thus, these modulatory subunits could be potential drug targets for neuropathic pain.SIGNIFICANCE STATEMENT Neuropathic pain, a type of moderate to severe chronic pain resulting from nerve injury or disorder, affects 6.9%-10% of the global population. However, less than half of patients report satisfactory pain relief from current treatments. K+ channels, which act to reduce nociceptor activity, have been suggested to be novel drug targets for neuropathic pain. This study is the first to show that K+ channel modulatory subunits KChIP1, KChIP2, and DPP10 are potential drug targets for neuropathic pain because they form a channel complex with the K+ channel pore-forming subunit Kv4.3 in a subset of nociceptors to selectively inhibit mechanical hypersensitivity, a major symptom of neuropathic pain.
Collapse
|
30
|
|
31
|
Li CL, Liu XF, Li GX, Ban MQ, Chen JZ, Cui Y, Zhang JH, Wu CF. Antinociceptive Effects of AGAP, a Recombinant Neurotoxic Polypeptide: Possible Involvement of the Tetrodotoxin-Resistant Sodium Channels in Small Dorsal Root Ganglia Neurons. Front Pharmacol 2016; 7:496. [PMID: 28066245 PMCID: PMC5168466 DOI: 10.3389/fphar.2016.00496] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 12/02/2016] [Indexed: 11/22/2022] Open
Abstract
Antitumor-analgesic peptide (AGAP) is a novel recombinant polypeptide. The primary study showed that AGAP 1.0 mg/kg exhibited strong analgesic and antitumor effects. The tail vein administration of AGAP potently reduced pain behaviors in mice induced by intraplantar injection of formalin or intraperitoneal injection of acetic acid, without affecting basal pain perception. To further assess the mechanisms of AGAP, the effects of AGAP on sodium channels were assessed using the whole-cell patch clamp recordings in dorsal root ganglia (DRG) neurons. The results showed that AGAP (3–1000 nM) inhibited the sodium currents in small-diameter DRG neurons in a dose-dependent manner. 1000 nM AGAP could inhibit the current density-voltage relationship curve of sodium channels in a voltage-dependent manner and negatively shift the activation curves. 1000 nM AGAP could reduce the tetrodotoxin-resistant (TTX-R) sodium currents by 42.8% in small-diameter DRG neurons. Further analysis revealed that AGAP potently inhibited NaV1.8 currents by 59.4%, and negatively shifted the activation and inactivation kinetics. 1000 nM AGAP also reduced the NaV1.9 currents by 33.7%, but had no significant effect on activation and inactivation kinetics. Thus, our results demonstrated that submicromolar concentrations of AGAP inhibited TTX-R sodium channel in rat small-diameter DRG neurons. It is concluded that these new results may better explain, at least in part, the analgesic properties of this polypeptide.
Collapse
Affiliation(s)
- Chun-Li Li
- Department of Pharmacology, Shenyang Pharmaceutical University Shenyang, China
| | - Xi-Fang Liu
- Department of Pharmacology, Shenyang Pharmaceutical University Shenyang, China
| | - Gui-Xia Li
- Department of Pharmacology, Shenyang Pharmaceutical University Shenyang, China
| | - Meng-Qi Ban
- Department of Pharmacology, Shenyang Pharmaceutical University Shenyang, China
| | - Jian-Zhao Chen
- Department of Pharmacology, Shenyang Pharmaceutical University Shenyang, China
| | - Yong Cui
- Department of Biochemistry, Shenyang Pharmaceutical University Shenyang, China
| | - Jing-Hai Zhang
- Department of Biochemistry, Shenyang Pharmaceutical University Shenyang, China
| | - Chun-Fu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University Shenyang, China
| |
Collapse
|
32
|
Voltage-gated sodium channels and pain-related disorders. Clin Sci (Lond) 2016; 130:2257-2265. [DOI: 10.1042/cs20160041] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 09/15/2016] [Indexed: 11/17/2022]
Abstract
Voltage-gated sodium channels (VGSCs) are heteromeric transmembrane protein complexes. Nine homologous members, SCN1A–11A, make up the VGSC gene family. Sodium channel isoforms display a wide range of kinetic properties endowing different neuronal types with distinctly varied firing properties. Among the VGSCs isoforms, Nav1.7, Nav1.8 and Nav1.9 are preferentially expressed in the peripheral nervous system. These isoforms are known to be crucial in the conduction of nociceptive stimuli with mutations in these channels thought to be the underlying cause of a variety of heritable pain disorders. This review provides an overview of the current literature concerning the role of VGSCs in the generation of pain and heritable pain disorders.
Collapse
|
33
|
Chen J, Guan SM, Sun W, Fu H. Melittin, the Major Pain-Producing Substance of Bee Venom. Neurosci Bull 2016; 32:265-72. [PMID: 26983715 DOI: 10.1007/s12264-016-0024-y] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 01/25/2016] [Indexed: 11/24/2022] Open
Abstract
Melittin is a basic 26-amino-acid polypeptide that constitutes 40-60% of dry honeybee (Apis mellifera) venom. Although much is known about its strong surface activity on lipid membranes, less is known about its pain-producing effects in the nervous system. In this review, we provide lines of accumulating evidence to support the hypothesis that melittin is the major pain-producing substance of bee venom. At the psychophysical and behavioral levels, subcutaneous injection of melittin causes tonic pain sensation and pain-related behaviors in both humans and animals. At the cellular level, melittin activates primary nociceptor cells through direct and indirect effects. On one hand, melittin can selectively open thermal nociceptor transient receptor potential vanilloid receptor channels via phospholipase A2-lipoxygenase/cyclooxygenase metabolites, leading to depolarization of primary nociceptor cells. On the other hand, algogens and inflammatory/pro-inflammatory mediators released from the tissue matrix by melittin's pore-forming effects can activate primary nociceptor cells through both ligand-gated receptor channels and the G-protein-coupled receptor-mediated opening of transient receptor potential canonical channels. Moreover, subcutaneous melittin up-regulates Nav1.8 and Nav1.9 subunits, resulting in the enhancement of tetrodotoxin-resistant Na(+) currents and the generation of long-term action potential firing. These nociceptive responses in the periphery finally activate and sensitize the spinal dorsal horn pain-signaling neurons, resulting in spontaneous nociceptive paw flinches and pain hypersensitivity to thermal and mechanical stimuli. Taken together, it is concluded that melittin is the major pain-producing substance of bee venom, by which peripheral persistent pain and hyperalgesia (or allodynia), primary nociceptive neuronal sensitization, and CNS synaptic plasticity (or metaplasticity) can be readily induced and the molecular and cellular mechanisms underlying naturally-occurring venomous biotoxins can be experimentally unraveled.
Collapse
Affiliation(s)
- Jun Chen
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.
- Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China.
- Beijing Institute for Brain Disorders, Beijing, 100069, China.
| | - Su-Min Guan
- School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Wei Sun
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Han Fu
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| |
Collapse
|
34
|
Optogenetic Silencing of Nav1.8-Positive Afferents Alleviates Inflammatory and Neuropathic Pain. eNeuro 2016; 3:eN-NWR-0140-15. [PMID: 27022626 PMCID: PMC4794527 DOI: 10.1523/eneuro.0140-15.2016] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/03/2016] [Accepted: 02/19/2016] [Indexed: 11/21/2022] Open
Abstract
We report a novel transgenic mouse model in which the terminals of peripheral nociceptors can be silenced optogenetically with high spatiotemporal precision, leading to the alleviation of inflammatory and neuropathic pain. Inhibitory archaerhodopsin-3 (Arch) proton pumps were delivered to Nav1.8+ primary afferents using the Nav1.8-Cre driver line. Arch expression covered both peptidergic and nonpeptidergic nociceptors and yellow light stimulation reliably blocked electrically induced action potentials in DRG neurons. Acute transdermal illumination of the hindpaws of Nav1.8-Arch+ mice significantly reduced mechanical allodynia under inflammatory conditions, while basal mechanical sensitivity was not affected by the optical stimulation. Arch-driven hyperpolarization of nociceptive terminals was sufficient to prevent channelrhodopsin-2 (ChR2)-mediated mechanical and thermal hypersensitivity in double-transgenic Nav1.8-ChR2+-Arch+mice. Furthermore, prolonged optical silencing of peripheral afferents in anesthetized Nav1.8-Arch+ mice led to poststimulation analgesia with a significant decrease in mechanical and thermal hypersensitivity under inflammatory and neuropathic conditions. These findings highlight the role of peripheral neuronal inputs in the onset and maintenance of pain hypersensitivity, demonstrate the plasticity of pain pathways even after sensitization has occurred, and support the involvement of Nav1.8+ afferents in both inflammatory and neuropathic pain. Together, we present a selective analgesic approach in which genetically identified subsets of peripheral sensory fibers can be remotely and optically inhibited with high temporal resolution, overcoming the compensatory limitations of genetic ablations.
Collapse
|
35
|
Wang Y, Lu YF, Li CL, Sun W, Li Z, Wang RR, He T, Yang F, Yang Y, Wang XL, Guan SM, Chen J. Involvement of Rac1 signalling pathway in the development and maintenance of acute inflammatory pain induced by bee venom injection. Br J Pharmacol 2016; 173:937-50. [PMID: 26700000 DOI: 10.1111/bph.13413] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 12/10/2015] [Accepted: 12/12/2015] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE The Rho GTPase, Rac1, is involved in the pathogenesis of neuropathic pain induced by malformation of dendritic spines in the spinal dorsal horn (sDH) neurons. In the present study, the contribution of spinal Rac1 to peripheral inflammatory pain was studied. EXPERIMENTAL APPROACH Effects of s.c. bee venom (BV) injection on cellular localization of Rac1 in the rat sDH was determined with double labelling immunofluorescence. Activation of Rac1 and its downstream effector p21-activated kinase (PAK), ERKs and p38 MAPK in inflammatory pain states was evaluated with a pull-down assay and Western blotting. The preventive and therapeutic analgesic effects of intrathecal administration of NSC23766, a selective inhibitor of Rac1, on BV-induced spontaneous nociception and pain hypersensitivity were investigated. KEY RESULTS Rac1 labelling was mainly localized within neurons in both the superficial and deep layers of the sDH in rats of naïve, vehicle-treated and inflamed (BV injected) groups. GTP-Rac1-PAK and ERKs/p38 were activated following s.c. BV injection. Post-treatment with intrathecal NSC23766 significantly inhibited GTP-Rac1 activity and phosphorylation of Rac1-PAK, ERKs and p38 MAPK in the sDH. Both pre-treatment and post-treatment with intrathecal NSC23766 dose-dependently attenuated the paw flinches, primary thermal and mechanical hyperalgesia and the mirror-image thermal hyperalgesia induced by BV injection, but without affecting the baseline pain sensitivity and motor coordination. CONCLUSIONS AND IMPLICATIONS The spinal GTP-Rac1-PAK-ERK/p38MAPK signalling pathway is involved in both the development and maintenance of peripheral inflammatory pain and can be used as a potential molecular target for developing a novel therapeutic strategy for clinical pain.
Collapse
Affiliation(s)
- Yan Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Yun-Fei Lu
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Chun-Li Li
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Wei Sun
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Zhen Li
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Rui-Rui Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Ting He
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Fan Yang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Yan Yang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Xiao-Liang Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Su-Min Guan
- School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jun Chen
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China.,Beijing Institute for Brain Disorders, Beijing, 100069, China
| |
Collapse
|
36
|
Ye P, Hua L, Jiao Y, Li Z, Qin S, Fu J, Jiang F, Liu T, Ji Y. Functional up-regulation of Nav1.8 sodium channel on dorsal root ganglia neurons contributes to the induction of scorpion sting pain. Acta Biochim Biophys Sin (Shanghai) 2016; 48:132-44. [PMID: 26764239 DOI: 10.1093/abbs/gmv123] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/02/2015] [Indexed: 12/19/2022] Open
Abstract
BmK I, purified from the venom of scorpion Buthus martensi Karsch (BmK), is a receptor site-3-specific modulator of voltage-gated sodium channels (VGSCs) and can induce pain-related behaviors in rats. The tetrodotoxin-resistant (TTX-R) sodium channel Nav1.8 contributes to most of the sodium current underlying the action potential upstroke in dorsal root ganglia (DRG) neurons and may serve as a critical ion channel targeted by BmK I. Herein, using electrophysiological, molecular, and behavioral approaches, we investigated whether the aberrant expression of Nav1.8 in DRG contributes to generation of pain induced by BmK I. The expression of Nav1.8 was found to be significantly increased at both mRNA and protein levels following intraplantar injection of BmK I in rats. In addition, the current density of TTX-R Nav1.8 sodium channel is significantly increased and the gating kinetics of Nav1.8 is also altered in DRG neurons from BmK I-treated rats. Furthermore, spontaneous pain and mechanical allodynia, but not thermal hyperalgesia induced by BmK I, are significantly alleviated through either blockade of the Nav1.8 sodium channel by its selective blocker A-803467 or knockdown of the Nav1.8 expression in DRG by antisense oligodeoxynucleotide (AS-ODN) targeting Nav1.8 in rats. Finally, BmK I was shown to induce enhanced pain behaviors in complete freund's adjuvant (CFA)-inflamed rats, which was partly due to the over-expression of Nav1.8 in DRG. Our results suggest that functional up-regulation of Nav1.8 channel on DRG neurons contributes to the development of BmK I-induced pain in rats.
Collapse
Affiliation(s)
- Pin Ye
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai 200436, China
| | - Liming Hua
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai 200436, China
| | - Yunlu Jiao
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai 200436, China
| | - Zhenwei Li
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai 200436, China
| | - Shichao Qin
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai 200436, China
| | - Jin Fu
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai 200436, China
| | - Feng Jiang
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai 200436, China
| | - Tong Liu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience, Soochow University, Suzhou 215021, China
| | - Yonghua Ji
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai 200436, China
| |
Collapse
|
37
|
Gabapentinoid Insensitivity after Repeated Administration is Associated with Down-Regulation of the α(2)δ-1 Subunit in Rats with Central Post-Stroke Pain Hypersensitivity. Neurosci Bull 2016; 32:41-50. [PMID: 26781878 DOI: 10.1007/s12264-015-0008-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/01/2015] [Indexed: 01/31/2023] Open
Abstract
The α2δ-1 subunit of the voltage-gated Ca(2+) channel (VGCC) is a molecular target of gabapentin (GBP), which has been used as a first-line drug for the relief of neuropathic pain. GBP exerts its anti-nociceptive effects by disrupting trafficking of the α2δ-1 subunit to the presynaptic membrane, resulting in decreased neurotransmitter release. We previously showed that GBP has an anti-allodynic effect in the first two weeks; but this is followed by insensitivity in the later stage after repeated administration in a rat model of central post-stroke pain (CPSP) hypersensitivity induced by intra-thalamic hemorrhage. To explore the mechanisms underlying GBP insensitivity, the cellular localization and time-course of expression of the α2δ-1 subunit in both the thalamus and spinal dorsal horn were studied in the same model. We found that the α2δ-1 subunit was mostly localized in neurons, but not astrocytes and microglia. The level of α2δ-1 protein increased in the first two weeks after injury but then decreased in the third week, when GBP insensitivity occurred. Furthermore, the α2δ-1 down-regulation was likely caused by later neuronal loss in the injured thalamus through a mechanism other than apoptosis. In summary, the present results suggest that the GBP receptor α2δ-1 is mainly expressed in thalamic neurons in which it is up-regulated in the early stage of CPSP but this is followed by dramatic down-regulation, which is likely associated with GBP insensitivity after long-term use.
Collapse
|
38
|
Yang F, Sun W, Yang Y, Wang Y, Li CL, Fu H, Wang XL, Yang F, He T, Chen J. SDF1-CXCR4 signaling contributes to persistent pain and hypersensitivity via regulating excitability of primary nociceptive neurons: involvement of ERK-dependent Nav1.8 up-regulation. J Neuroinflammation 2015; 12:219. [PMID: 26597700 PMCID: PMC4657286 DOI: 10.1186/s12974-015-0441-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 11/18/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Pain is one critical hallmark of inflammatory responses. A large number of studies have demonstrated that stromal cell-derived factor 1 (SDF1, also named as CXCL12) and its cognate receptor C-X-C chemokine receptor type 4 (CXCR4) play an important role in immune reaction and inflammatory processes. However, whether and how SDF1-CXCR4 signaling is involved in inflammatory pain remains unclear. METHODS Under the intraplantar (i.pl.) bee venom (BV) injection-induced persistent inflammatory pain state, the changes of SDF1 and CXCR4 expression and cellular localization in the rat dorsal root ganglion (DRG) were detected by immunofluorescent staining. The role of SDF1 and CXCR4 in the hyperexcitability of primary nociceptor neurons was assessed by electrophysiological recording. Western blot analysis was used to quantify the DRG Nav1.8 and phosphorylation of ERK (pERK) expression. Behavioral tests were conducted to evaluate the roles of CXCR4 as well as extracellular signal-regulated kinase (ERK) and Nav1.8 in the BV-induced persistent pain and hypersensitivity. RESULTS We showed that both SDF1 and CXCR4 were dramatically up-regulated in the DRG in i.pl. BV-induced inflammatory pain model. Double immunofluorescent staining showed that CXCR4 was localized in all sizes (large, medium, and small) of DRG neuronal soma, while SDF1 was exclusively expressed in satellite glial cells (SGCs). Electrophysiological recording showed that bath application with AMD3100, a potent and selective CXCR4 inhibitor, could reverse the hyperexcitability of medium- and small-sized DRG neurons harvested from rats following i.pl. BV injection. Furthermore, we demonstrated that the BV-induced ERK activation and Nav1.8 up-regulation in the DRG could be blocked by pre-antagonism against CXCR4 in the periphery with AMD3100 as well as by blockade of ERK activation by intrathecal (i.t.) or intraplantar (i.pl.) U0126. At behavioral level, the BV-induced persistent spontaneous pain as well as primary mechanical and thermal hypersensitivity could also be significantly suppressed by blocking CXCR4 and Nav1.8 in the periphery as well as by inhibition of ERK activation at the DRG level. CONCLUSIONS The present results suggest that peripheral inflammatory pain state can trigger over release of SDF1 from the activated SGCs in the DRG by which SGC-neuronal cross-talk is mediated by SDF1-CXCR4 coupling that result in subsequent ERK-dependent Nav1.8 up-regulation, leading to hyperexcitability of tonic type of the primary nociceptor cells and development and maintenance of persistent spontaneous pain and hypersensitivity.
Collapse
Affiliation(s)
- Fei Yang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, #569 Xinsi Road, Baqiao, Xi'an, 710038, People's Republic of China
| | - Wei Sun
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, #569 Xinsi Road, Baqiao, Xi'an, 710038, People's Republic of China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, People's Republic of China
| | - Yan Yang
- Beijing Institute for Brain Disorders, Beijing, 100069, People's Republic of China
| | - Yan Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, #569 Xinsi Road, Baqiao, Xi'an, 710038, People's Republic of China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, People's Republic of China
| | - Chun-Li Li
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, #569 Xinsi Road, Baqiao, Xi'an, 710038, People's Republic of China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, People's Republic of China
| | - Han Fu
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, #569 Xinsi Road, Baqiao, Xi'an, 710038, People's Republic of China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, People's Republic of China
| | - Xiao-Liang Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, #569 Xinsi Road, Baqiao, Xi'an, 710038, People's Republic of China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, People's Republic of China
| | - Fan Yang
- Beijing Institute for Brain Disorders, Beijing, 100069, People's Republic of China
| | - Ting He
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, #569 Xinsi Road, Baqiao, Xi'an, 710038, People's Republic of China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, People's Republic of China
| | - Jun Chen
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, #569 Xinsi Road, Baqiao, Xi'an, 710038, People's Republic of China. .,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, People's Republic of China. .,Beijing Institute for Brain Disorders, Beijing, 100069, People's Republic of China.
| |
Collapse
|
39
|
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: 5.6] [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
|
40
|
LI GUIXIA, LIU XIFANG, DU JINGNAN, CHEN JIANZHAO, SHE FENGLIN, WU CHUNFU, LI CHUNLI. Positive shift of Nav1.8 current inactivation curve in injured neurons causes neuropathic pain following chronic constriction injury. Mol Med Rep 2015; 12:3583-3590. [DOI: 10.3892/mmr.2015.3839] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 04/24/2015] [Indexed: 11/05/2022] Open
|
41
|
Su J, Gao T, Shi T, Xiang Q, Xu X, Wiesenfeld-Hallin Z, Hökfelt T, Svensson CI. Phenotypic changes in dorsal root ganglion and spinal cord in the collagen antibody-induced arthritis mouse model. J Comp Neurol 2015; 523:1505-28. [DOI: 10.1002/cne.23749] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 12/26/2014] [Accepted: 01/24/2015] [Indexed: 12/31/2022]
Affiliation(s)
- Jie Su
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm 171 77 Sweden
| | - Tianle Gao
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm 171 77 Sweden
| | - Tiejun Shi
- Department of Neuroscience; Karolinska Institutet; Stockholm 171 77 Sweden
| | - Qiong Xiang
- Department of Neuroscience; Karolinska Institutet; Stockholm 171 77 Sweden
| | - Xiaojun Xu
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm 171 77 Sweden
| | | | - Tomas Hökfelt
- Department of Neuroscience; Karolinska Institutet; Stockholm 171 77 Sweden
| | - Camilla I. Svensson
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm 171 77 Sweden
| |
Collapse
|
42
|
Persistent pain after spinal cord injury is maintained by primary afferent activity. J Neurosci 2014; 34:10765-9. [PMID: 25100607 DOI: 10.1523/jneurosci.5316-13.2014] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Chronic pain caused by insults to the CNS (central neuropathic pain) is widely assumed to be maintained exclusively by central mechanisms. However, chronic hyperexcitablility occurs in primary nociceptors after spinal cord injury (SCI), suggesting that SCI pain also depends upon continuing activity of peripheral sensory neurons. The present study in rats (Rattus norvegicus) found persistent upregulation after SCI of protein, but not mRNA, for a voltage-gated Na(+) channel, Nav1.8, that is expressed almost exclusively in primary afferent neurons. Selectively knocking down Nav1.8 after SCI suppressed spontaneous activity in dissociated dorsal root ganglion neurons, reversed hypersensitivity of hindlimb withdrawal reflexes, and reduced ongoing pain assessed by a conditioned place preference test. These results show that activity in primary afferent neurons contributes to ongoing SCI pain.
Collapse
|
43
|
Yu YQ, Chen XF, Yang Y, Yang F, Chen J. Electrophysiological identification of tonic and phasic neurons in sensory dorsal root ganglion and their distinct implications in inflammatory pain. Physiol Res 2014; 63:793-9. [PMID: 25157654 DOI: 10.33549/physiolres.932708] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
In the mammalian autonomic nervous system, tonic and phasic neurons can be differentiated on firing patterns in response to long depolarizing current pulse. However, the similar firing patterns in the somatic primary sensory neurons and their functional significance are not well investigated. Here, we identified two types of neurons innervating somatic sensory in rat dorsal root ganglia (DRG). Tonic neurons fire action potentials (APs) in an intensity-dependent manner, whereas phasic neurons typically generate only one AP firing at the onset of stimulation regardless of intensity. Combining retrograde labeling of somatic DRG neurons with fluorescent tracer DiI, we further find that these neurons demonstrate distinct changes under inflammatory pain states induced by complete Freund's adjuvant (CFA) or bee venom toxin melittin. In tonic neurons, CFA and melittin treatments significantly decrease rheobase and AP durations (depolarization and repolarization), enhance amplitudes of overshoot and afterhyperpolarization (AHP), and increase the number of evoked action potentials. In phasic neurons, however, the same inflammation treatments cause fewer changes in these electrophysiological parameters except for the increased overshoot and decreased AP durations. In the present study, we find that tonic neurons are more hyperexcitable than phasic neurons after peripheral noxious inflammatory stimulation. The results indicate the distinct contributions of two types of DRG neurons in inflammatory pain.
Collapse
Affiliation(s)
- Y-Q Yu
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Fourth Military Medical University, Xi'an, PR China.
| | | | | | | | | |
Collapse
|
44
|
Liu X, Li C, Chen J, Du J, Zhang J, Li G, Jin X, Wu C. AGAP, a new recombinant neurotoxic polypeptide, targets the voltage-gated calcium channels in rat small diameter DRG neurons. Biochem Biophys Res Commun 2014; 452:60-5. [PMID: 25148943 DOI: 10.1016/j.bbrc.2014.08.051] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 08/11/2014] [Indexed: 10/24/2022]
Abstract
A previous study showed that antitumor-analgesic peptide (AGAP), a novel recombinant polypeptide, which had been expressed in Escherichia coli, exhibits analgesic and antitumor effects in mice. In the present study, we investigated the underlying analgesic mechanism of AGAP. The effect of AGAP on voltage-gated calcium channels (VGCCs) was assessed in acutely isolated rat dorsal root ganglia (DRG) neurons using the whole-cell patch clamp technique. The results showed that AGAP potently inhibited VGCCs, especially high-voltage activated (HVA) calcium channels. AGAP inhibited HVA and T-type calcium currents in a dose-dependent manner, but had no significant effect on their dynamic functions in rat small-diameter DRG neurons. AGAP inhibited N- and L-type calcium currents at 78.2% and 57.3%, respectively. Thus, the present study demonstrates that AGAP affects calcium currents through the inhibition of N-, L- and T-type channels in DRG neurons, explaining the potential mechanisms of antinociception.
Collapse
Affiliation(s)
- Xifang Liu
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Chunli Li
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China.
| | - Jianzhao Chen
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Jingnan Du
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Jinghai Zhang
- Department of Biochemistry, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Guixia Li
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Xiaoquan Jin
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Chunfu Wu
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
45
|
Tetrodotoxin-resistant sodium channels in sensory neurons generate slow resurgent currents that are enhanced by inflammatory mediators. J Neurosci 2014; 34:7190-7. [PMID: 24849353 DOI: 10.1523/jneurosci.5011-13.2014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Resurgent sodium currents contribute to the regeneration of action potentials and enhanced neuronal excitability. Tetrodotoxin-sensitive (TTX-S) resurgent currents have been described in many different neuron populations, including cerebellar and dorsal root ganglia (DRG) neurons. In most cases, sodium channel Nav1.6 is the major contributor to these TTX-S resurgent currents. Here we report a novel TTX-resistant (TTX-R) resurgent current recorded from rat DRG neurons. The TTX-R resurgent currents are similar to classic TTX-S resurgent currents in many respects, but not all. As with TTX-S resurgent currents, they are activated by membrane repolarization, inhibited by lidocaine, and enhanced by a peptide-mimetic of the β4 sodium channel subunit intracellular domain. However, the TTX-R resurgent currents exhibit much slower kinetics, occur at more depolarized voltages, and are sensitive to the Nav1.8 blocker A803467. Moreover, coimmunoprecipitation experiments from rat DRG lysates indicate the endogenous sodium channel β4 subunits associate with Nav1.8 in DRG neurons. These results suggest that slow TTX-R resurgent currents in DRG neurons are mediated by Nav1.8 and are generated by the same mechanism underlying TTX-S resurgent currents. We also show that both TTX-S and TTX-R resurgent currents in DRG neurons are enhanced by inflammatory mediators. Furthermore, the β4 peptide increased excitability of small DRG neurons in the presence of TTX. We propose that these slow TTX-R resurgent currents contribute to the membrane excitability of nociceptive DRG neurons under normal conditions and that enhancement of both types of resurgent currents by inflammatory mediators could contribute to sensory neuronal hyperexcitability associated with inflammatory pain.
Collapse
|
46
|
Hockley JRF, Boundouki G, Cibert-Goton V, McGuire C, Yip PK, Chan C, Tranter M, Wood JN, Nassar MA, Blackshaw LA, Aziz Q, Michael GJ, Baker MD, Winchester WJ, Knowles CH, Bulmer DC. Multiple roles for NaV1.9 in the activation of visceral afferents by noxious inflammatory, mechanical, and human disease-derived stimuli. Pain 2014; 155:1962-75. [PMID: 24972070 PMCID: PMC4220011 DOI: 10.1016/j.pain.2014.06.015] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/28/2014] [Accepted: 06/19/2014] [Indexed: 01/17/2023]
Abstract
Chronic visceral pain affects millions of individuals worldwide and remains poorly understood, with current therapeutic options constrained by gastrointestinal adverse effects. Visceral pain is strongly associated with inflammation and distension of the gut. Here we report that the voltage-gated sodium channel subtype NaV1.9 is expressed in half of gut-projecting rodent dorsal root ganglia sensory neurons. We show that NaV1.9 is required for normal mechanosensation, for direct excitation and for sensitization of mouse colonic afferents by mediators from inflammatory bowel disease tissues, and by noxious inflammatory mediators individually. Excitatory responses to ATP or PGE2 were substantially reduced in NaV1.9(-/-) mice. Deletion of NaV1.9 substantially attenuates excitation and subsequent mechanical hypersensitivity after application of inflammatory soup (IS) (bradykinin, ATP, histamine, PGE2, and 5HT) to visceral nociceptors located in the serosa and mesentery. Responses to mechanical stimulation of mesenteric afferents were also reduced by loss of NaV1.9, and there was a rightward shift in stimulus-response function to ramp colonic distension. By contrast, responses to rapid, high-intensity phasic distension of the colon are initially unaffected; however, run-down of responses to repeat phasic distension were exacerbated in NaV1.9(-/-) afferents. Finally colonic afferent activation by supernatants derived from inflamed human tissue was greatly reduced in NaV1.9(-/-) mice. These results demonstrate that NaV1.9 is required for persistence of responses to intense mechanical stimulation, contributes to inflammatory mechanical hypersensitivity, and is essential for activation by noxious inflammatory mediators, including those from diseased human bowel. These observations indicate that NaV1.9 represents a high-value target for development of visceral analgesics.
Collapse
Affiliation(s)
- James R F Hockley
- Wingate Institute of Neurogastroenterology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AJ, UK; National Centre for Bowel Research and Surgical Innovation, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - George Boundouki
- Wingate Institute of Neurogastroenterology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AJ, UK; National Centre for Bowel Research and Surgical Innovation, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Vincent Cibert-Goton
- National Centre for Bowel Research and Surgical Innovation, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Cian McGuire
- National Centre for Bowel Research and Surgical Innovation, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Ping K Yip
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Christopher Chan
- National Centre for Bowel Research and Surgical Innovation, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Michael Tranter
- National Centre for Bowel Research and Surgical Innovation, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Mohammed A Nassar
- Department of Biomedical Science, The University of Sheffield, Sheffield S10 2TN, UK
| | - L Ashley Blackshaw
- Wingate Institute of Neurogastroenterology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AJ, UK; National Centre for Bowel Research and Surgical Innovation, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Qasim Aziz
- Wingate Institute of Neurogastroenterology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AJ, UK
| | - Gregory J Michael
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Mark D Baker
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Wendy J Winchester
- Neusentis (Pfizer Ltd), The Portway Building, Granta Science Park, Cambridge CB21 6GS, UK
| | - Charles H Knowles
- National Centre for Bowel Research and Surgical Innovation, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - David C Bulmer
- Wingate Institute of Neurogastroenterology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AJ, UK; National Centre for Bowel Research and Surgical Innovation, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK.
| |
Collapse
|
47
|
Belkouch M, Dansereau MA, Tétreault P, Biet M, Beaudet N, Dumaine R, Chraibi A, Mélik-Parsadaniantz S, Sarret P. Functional up-regulation of Nav1.8 sodium channel in Aβ afferent fibers subjected to chronic peripheral inflammation. J Neuroinflammation 2014; 11:45. [PMID: 24606981 PMCID: PMC4007624 DOI: 10.1186/1742-2094-11-45] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 02/21/2014] [Indexed: 02/05/2023] Open
Abstract
Background Functional alterations in the properties of Aβ afferent fibers may account for the increased pain sensitivity observed under peripheral chronic inflammation. Among the voltage-gated sodium channels involved in the pathophysiology of pain, Nav1.8 has been shown to participate in the peripheral sensitization of nociceptors. However, to date, there is no evidence for a role of Nav1.8 in controlling Aβ-fiber excitability following persistent inflammation. Methods Distribution and expression of Nav1.8 in dorsal root ganglia and sciatic nerves were qualitatively or quantitatively assessed by immunohistochemical staining and by real time-polymerase chain reaction at different time points following complete Freund’s adjuvant (CFA) administration. Using a whole-cell patch-clamp configuration, we further determined both total INa and TTX-R Nav1.8 currents in large-soma dorsal root ganglia (DRG) neurons isolated from sham or CFA-treated rats. Finally, we analyzed the effects of ambroxol, a Nav1.8-preferring blocker on the electrophysiological properties of Nav1.8 currents and on the mechanical sensitivity and inflammation of the hind paw in CFA-treated rats. Results Our findings revealed that Nav1.8 is up-regulated in NF200-positive large sensory neurons and is subsequently anterogradely transported from the DRG cell bodies along the axons toward the periphery after CFA-induced inflammation. We also demonstrated that both total INa and Nav1.8 peak current densities are enhanced in inflamed large myelinated Aβ-fiber neurons. Persistent inflammation leading to nociception also induced time-dependent changes in Aβ-fiber neuron excitability by shifting the voltage-dependent activation of Nav1.8 in the hyperpolarizing direction, thus decreasing the current threshold for triggering action potentials. Finally, we found that ambroxol significantly reduces the potentiation of Nav1.8 currents in Aβ-fiber neurons observed following intraplantar CFA injection and concomitantly blocks CFA-induced mechanical allodynia, suggesting that Nav1.8 regulation in Aβ-fibers contributes to inflammatory pain. Conclusions Collectively, these findings support a key role for Nav1.8 in controlling the excitability of Aβ-fibers and its potential contribution to the development of mechanical allodynia under persistent inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Philippe Sarret
- Department of Physiology and Biophysics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12th Avenue North, Sherbrooke, Quebec J1H 5N4, Canada.
| |
Collapse
|
48
|
Baroni D, Picco C, Barbieri R, Moran O. Antisense-mediated post-transcriptional silencing of SCN1B gene modulates sodium channel functional expression. Biol Cell 2013; 106:13-29. [PMID: 24138709 DOI: 10.1111/boc.201300040] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 10/11/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND INFORMATION Voltage-dependent sodium channels are membrane proteins essential for cell excitability. They are composed by a pore-forming α-subunit and one or more β subunits. Nine α subunit and five β subunit isoforms have been identified in mammals: β1, its splice variant β1B, β2, β3 and β4. Although they do not form the ion channel pore, β subunits modulate both function as well as expression of sodium channels on cell membrane. RESULTS To investigate the role of β1 subunit on the modulation of sodium channel expression, we silenced this auxiliary subunit with specific antisense oligonucleotides (ASONs) in two rat cell lines, the GH3 and the H9C2, from neuro-ectoderm and cardiac myocyte origin, respectively. Treatment of cells with ASONs determined a reduction of about 50% of β1 subunit mRNA and protein expression in both cell lines. We found that this level of β1 subunit silencing resulted in an overall decrease of α subunit mRNA, protein expression and a decrease of sodium current density, without altering significantly the voltage-dependent and kinetic properties of the currents. In GH3 cells, the β1 subunit silencing reduced the expression of Nav1.1, Nav1.3 and Nav1.6 isoforms, whereas the Nav 1.2 isoform expression remained unaltered. The expression of the only α subunit present in H9C2 cells, the Nav1.5, was also reduced by β1 subunit silencing. CONCLUSIONS These results indicate that the β1 subunit may exert an isoform-specific fine-tuned modulation of sodium channel expression.
Collapse
|
49
|
Blockade of Nav1.8 Currents in Nociceptive Trigeminal Neurons Contributes to Anti-trigeminovascular Nociceptive Effect of Amitriptyline. Neuromolecular Med 2013; 16:308-21. [DOI: 10.1007/s12017-013-8282-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 11/08/2013] [Indexed: 01/22/2023]
|
50
|
Yu J, Fang Q, Lou GD, Shou WT, Yue JX, Tang YY, Hou WW, Xu TL, Ohtsu H, Zhang SH, Chen Z. Histamine modulation of acute nociception involves regulation of Nav 1.8 in primary afferent neurons in mice. CNS Neurosci Ther 2013; 19:649-58. [PMID: 23773488 DOI: 10.1111/cns.12134] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 05/06/2013] [Accepted: 05/09/2013] [Indexed: 11/27/2022] Open
Abstract
AIMS To explore the role of histamine in acute pain perception and its possible mechanisms. METHODS Pain-like behaviors induced by four types of noxious stimuli (hot-plate, tail-pressure, acetic acid, and formalin) were accessed in mice. Nav 1.8 expression and functions in primary afferent neurons were compared between histidine decarboxylase knockout (HDC(-/-) ) mice and their wild-types. RESULTS HDC(-/-) mice, lacking in endogenous histamine, showed elevated sensitivity to all these noxious stimuli, as compared with the wild-types. In addition, a depletion of endogenous histamine with α-fluoromethylhistidine (α-FMH), a specific HDC inhibitor, or feeding mice a low-histamine diet also enhanced nociception in the wild-types. Nav 1.8 expression in primary afferent neurons was increased both in HDC(-/-) and in α-FMH-treated wild-type mice. A higher Nav 1.8 current density, a lower action potential (AP) threshold, and a higher firing rate in response to suprathreshold stimulation were observed in nociception-related small DRG neurons of HDC(-/-) mice. Nav 1.8 inhibitor A-803467, but not TTX, diminished the hyperexcitability and blocked repetitive AP firing of these neurons. CONCLUSION Our results indicate that histamine participates in acute pain modulation in a dose-related manner. The regulation of Nav 1.8 expression and the excitability of nociceptive primary afferent neurons may be involved in the underlying mechanisms.
Collapse
Affiliation(s)
- Jie Yu
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|