1
|
Staedtler ES, Sapio MR, King DM, Maric D, Ghetti A, Mannes AJ, Iadarola MJ. The μ-opioid receptor differentiates two distinct human nociceptive populations relevant to clinical pain. Cell Rep Med 2024; 5:101788. [PMID: 39413733 PMCID: PMC11513826 DOI: 10.1016/j.xcrm.2024.101788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 06/26/2024] [Accepted: 09/19/2024] [Indexed: 10/18/2024]
Abstract
The shortfall in new analgesic agents is a major impediment to reducing reliance on opioid medications for control of severe pain. In both animals and man, attenuating nociceptive transmission from primary afferent neurons with a μ-opioid receptor agonist yields highly effective analgesia. Consequently, deeper molecular characterization of human nociceptive afferents expressing OPRM1, the μ-opioid receptor gene, is a key component for advancing analgesic drug discovery and understanding clinical pain control. A co-expression matrix for the μ-opioid receptor and a variety of nociceptive channels as well as δ- and κ-opioid receptors is established by multiplex in situ hybridization. Our results indicate an OPRM1-positive population with strong molecular resemblance to rodent peptidergic C-nociceptors associated with tissue damage pain and an OPRM1-negative population sharing molecular characteristics of murine non-peptidergic C-nociceptors. The empirical identification of two distinct human nociceptive populations that differ profoundly in their presumed responsiveness to opioids provides an actionable translational framework for human pain control.
Collapse
Affiliation(s)
- Ellen S Staedtler
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Matthew R Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Diana M King
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dragan Maric
- National Institute of Neurological Disorders and Stroke, Flow and Imaging Cytometry Core Facility, Bethesda, MD 20892, USA
| | | | - Andrew J Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
2
|
Abstract
Neuropathic pain is a debilitating form of pain arising from injury or disease of the nervous system that affects millions of people worldwide. Despite its prevalence, the underlying mechanisms of neuropathic pain are still not fully understood. Dendritic spines are small protrusions on the surface of neurons that play an important role in synaptic transmission. Recent studies have shown that dendritic spines reorganize in the superficial and deeper laminae of the spinal cord dorsal horn with the development of neuropathic pain in multiple models of disease or injury. Given the importance of dendritic spines in synaptic transmission, it is possible that studying dendritic spines could lead to new therapeutic approaches for managing intractable pain. In this review article, we highlight the emergent role of dendritic spines in neuropathic pain, as well as discuss the potential for studying dendritic spines for the development of new therapeutics.
Collapse
Affiliation(s)
- Curtis A Benson
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Jared F King
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Marike L Reimer
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Sierra D Kauer
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Andrew M Tan
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
3
|
Zhang LG, Cheng J, An MQ, Li CJ, Dong LG, Wang JM, Liu CF, Wang F, Mao CJ. Safinamide alleviates hyperalgesia via inhibiting hyperexcitability of DRG neurons in a mouse model of Parkinson's disease. Behav Brain Res 2024; 459:114787. [PMID: 38042302 DOI: 10.1016/j.bbr.2023.114787] [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: 08/14/2023] [Revised: 11/17/2023] [Accepted: 11/25/2023] [Indexed: 12/04/2023]
Abstract
Pain is a widespread non-motor symptom that presents significant treatment challenges in patients with Parkinson's disease (PD). Safinamide, a new drug recently introduced for PD treatment, has demonstrated analgesic effects on pain in PD patients, though the underlying mechanisms remain unclear. To investigate the analgesic and anti-PD effect of safinamide, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model was used, and rasagiline as positive control on motor symptoms. Notably, only safinamide alleviated hyperalgesia in MPTP mice. Whole-cell patch-clamp recordings of dorsal root ganglion (DRG) neurons revealed hyperexcitability in MPTP mice, which safinamide counteracted in a concentration-dependent manner. The voltage clamp further demonstrated that sodium current in DRG neurons of MPTP mice was enhanced and safinamide reduced sodium current density. RT-qPCR identified upregulated Nav1.7 and Nav1.8 transcripts (Scn9a and Scn10a) in DRG neurons of MPTP mice. Our results suggest that safinamide could relieve hyperalgesia by inhibiting DRG neuron hyperexcitability in MPTP mice.
Collapse
Affiliation(s)
- Li-Ge Zhang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Jing Cheng
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Meng-Qi An
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Cheng-Jie Li
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Li-Guo Dong
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian-Min Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.
| | - Cheng-Jie Mao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
4
|
Amaya-Rodriguez CA, Carvajal-Zamorano K, Bustos D, Alegría-Arcos M, Castillo K. A journey from molecule to physiology and in silico tools for drug discovery targeting the transient receptor potential vanilloid type 1 (TRPV1) channel. Front Pharmacol 2024; 14:1251061. [PMID: 38328578 PMCID: PMC10847257 DOI: 10.3389/fphar.2023.1251061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/14/2023] [Indexed: 02/09/2024] Open
Abstract
The heat and capsaicin receptor TRPV1 channel is widely expressed in nerve terminals of dorsal root ganglia (DRGs) and trigeminal ganglia innervating the body and face, respectively, as well as in other tissues and organs including central nervous system. The TRPV1 channel is a versatile receptor that detects harmful heat, pain, and various internal and external ligands. Hence, it operates as a polymodal sensory channel. Many pathological conditions including neuroinflammation, cancer, psychiatric disorders, and pathological pain, are linked to the abnormal functioning of the TRPV1 in peripheral tissues. Intense biomedical research is underway to discover compounds that can modulate the channel and provide pain relief. The molecular mechanisms underlying temperature sensing remain largely unknown, although they are closely linked to pain transduction. Prolonged exposure to capsaicin generates analgesia, hence numerous capsaicin analogs have been developed to discover efficient analgesics for pain relief. The emergence of in silico tools offered significant techniques for molecular modeling and machine learning algorithms to indentify druggable sites in the channel and for repositioning of current drugs aimed at TRPV1. Here we recapitulate the physiological and pathophysiological functions of the TRPV1 channel, including structural models obtained through cryo-EM, pharmacological compounds tested on TRPV1, and the in silico tools for drug discovery and repositioning.
Collapse
Affiliation(s)
- Cesar A. Amaya-Rodriguez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Departamento de Fisiología y Comportamiento Animal, Facultad de Ciencias Naturales, Exactas y Tecnología, Universidad de Panamá, Ciudad de Panamá, Panamá
| | - Karina Carvajal-Zamorano
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Daniel Bustos
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado Universidad Católica del Maule, Talca, Chile
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca, Chile
| | - Melissa Alegría-Arcos
- Núcleo de Investigación en Data Science, Facultad de Ingeniería y Negocios, Universidad de las Américas, Santiago, Chile
| | - Karen Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado Universidad Católica del Maule, Talca, Chile
| |
Collapse
|
5
|
Abstract
Efforts to design devices emulating complex cognitive abilities and response processes of biological systems have long been a coveted goal. Recent advancements in flexible electronics, mirroring human tissue's mechanical properties, hold significant promise. Artificial neuron devices, hinging on flexible artificial synapses, bioinspired sensors, and actuators, are meticulously engineered to mimic the biological systems. However, this field is in its infancy, requiring substantial groundwork to achieve autonomous systems with intelligent feedback, adaptability, and tangible problem-solving capabilities. This review provides a comprehensive overview of recent advancements in artificial neuron devices. It starts with fundamental principles of artificial synaptic devices and explores artificial sensory systems, integrating artificial synapses and bioinspired sensors to replicate all five human senses. A systematic presentation of artificial nervous systems follows, designed to emulate fundamental human nervous system functions. The review also discusses potential applications and outlines existing challenges, offering insights into future prospects. We aim for this review to illuminate the burgeoning field of artificial neuron devices, inspiring further innovation in this captivating area of research.
Collapse
Affiliation(s)
- Ke He
- Innovative Centre for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Cong Wang
- Innovative Centre for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Yongli He
- Innovative Centre for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Jiangtao Su
- Innovative Centre for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Xiaodong Chen
- Innovative Centre for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
- Institute for Digital Molecular Analytics and Science (IDMxS), Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
| |
Collapse
|
6
|
Zhang T, Zhang M, Cui S, Liang W, Jia Z, Guo F, Ou W, Wu Y, Zhang S. The core of maintaining neuropathic pain: Crosstalk between glial cells and neurons (neural cell crosstalk at spinal cord). Brain Behav 2023; 13:e2868. [PMID: 36602945 PMCID: PMC9927860 DOI: 10.1002/brb3.2868] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/29/2022] [Accepted: 12/06/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Neuropathic pain (NP) caused by the injury or dysfunction of the nervous system is a chronic pain state accompanied by hyperalgesia, and the available clinical treatment is relatively scarce. Hyperalgesia mediated by pro-inflammatory factors and chemokines plays an important role in the occurrence and maintenance of NP. DATA TREATMENT Therefore, we conducted a systematic literature review of experimental NP (PubMed Medline), in order to find the mechanism of inducing central sensitization and explore the intervention methods of hyperalgesia caused by real or simulated injury. RESULT In this review, we sorted out the activation pathways of microglia, astrocytes and neurons, and the process of crosstalk among them. It was found that in NP, the microglia P2X4 receptor is the key target, which can activate the mitogen-activated protein kinase pathway inward and then activate astrocytes and outwardly activate neuronal tropomyosin receptor kinase B receptor to activate neurons. At the same time, activated neurons continue to maintain the activation of astrocytes and microglia through chemokines on CXCL13/CXCR5 and CX3CL1/CX3CR1. This crosstalk process is the key to maintaining NP. CONCLUSION We summarize the further research on crosstalk among neurons, microglia, and astrocytes in the central nervous system, elaborate the ways and connections of relevant crosstalk, and find potential crosstalk targets, which provides a reference for drug development and preclinical research.
Collapse
Affiliation(s)
- Tianrui Zhang
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Mingqian Zhang
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shuang Cui
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wulin Liang
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhanhong Jia
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Fanfan Guo
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wenjing Ou
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yonghong Wu
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shuofeng Zhang
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
7
|
Metea M, Palmero-Soler E, Crum L. Preclinical nerve conduction: Nerve battery options for primate studies. J Pharmacol Toxicol Methods 2022; 116:107187. [PMID: 35636693 DOI: 10.1016/j.vascn.2022.107187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/23/2022] [Accepted: 05/24/2022] [Indexed: 10/18/2022]
Abstract
Electrophysiological neurodiagnostic tests of nerve conduction (NC) are key assays included in preclinical safety and toxicology programs to assess the peripheral neuropathy (PN) liability of a new drug. Despite their increased use, standardization of nerve conduction studies (NCS) is lacking in the preclinical space, with limited regulatory guidelines stipulating type and number of nerves or minimum combinations appropriate for each stage of drug development or indication. Detection of subtle peripheral toxicities depends on choosing appropriate nerve targets for testing, especially when functional changes remain above the lower limit of normal values. To support robust preclinical toxicology study designs, the current short communication provides options and recommendations for selecting peripheral nerves for clinically translatable nerve conduction batteries applicable to toxicology and gene therapy, with a focus on clinically translatable primate models. A comprehensive compilation of accessible nerve locations is offered including lower and upper extremity motor nerves, and sensory nerves with origin at multiple DRG levels. Rankings of technique difficulty and repeatability across serial collections are presented for each assay informed by serial nerve conduction from 500 adult primates. The goal of this communication is to support the standardization and preclinical implementation of this important assay.
Collapse
Affiliation(s)
- Monica Metea
- Preclinical Electrophysiology Consulting, LLC, Mattapoisett, MA, USA.
| | | | - Lucas Crum
- Preclinical Electrophysiology Consulting, LLC, Mattapoisett, MA, USA
| |
Collapse
|
8
|
González-Cano R, Ruiz-Cantero MC, Santos-Caballero M, Gómez-Navas C, Tejada MÁ, Nieto FR. Tetrodotoxin, a Potential Drug for Neuropathic and Cancer Pain Relief? Toxins (Basel) 2021; 13:toxins13070483. [PMID: 34357955 PMCID: PMC8310002 DOI: 10.3390/toxins13070483] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/20/2022] Open
Abstract
Tetrodotoxin (TTX) is a potent neurotoxin found mainly in puffer fish and other marine and terrestrial animals. TTX blocks voltage-gated sodium channels (VGSCs) which are typically classified as TTX-sensitive or TTX-resistant channels. VGSCs play a key role in pain signaling and some TTX-sensitive VGSCs are highly expressed by adult primary sensory neurons. During pathological pain conditions, such as neuropathic pain, upregulation of some TTX-sensitive VGSCs, including the massive re-expression of the embryonic VGSC subtype NaV1.3 in adult primary sensory neurons, contribute to painful hypersensitization. In addition, people with loss-of-function mutations in the VGSC subtype NaV1.7 present congenital insensitive to pain. TTX displays a prominent analgesic effect in several models of neuropathic pain in rodents. According to this promising preclinical evidence, TTX is currently under clinical development for chemo-therapy-induced neuropathic pain and cancer-related pain. This review focuses primarily on the preclinical and clinical evidence that support a potential analgesic role for TTX in these pain states. In addition, we also analyze the main toxic effects that this neurotoxin produces when it is administered at therapeutic doses, and the therapeutic potential to alleviate neuropathic pain of other natural toxins that selectively block TTX-sensitive VGSCs.
Collapse
Affiliation(s)
- Rafael González-Cano
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, 18016 Granada, Spain; (R.G.-C.); (M.C.R.-C.); (M.S.-C.); (C.G.-N.)
- Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - M. Carmen Ruiz-Cantero
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, 18016 Granada, Spain; (R.G.-C.); (M.C.R.-C.); (M.S.-C.); (C.G.-N.)
- Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Miriam Santos-Caballero
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, 18016 Granada, Spain; (R.G.-C.); (M.C.R.-C.); (M.S.-C.); (C.G.-N.)
- Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Carlos Gómez-Navas
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, 18016 Granada, Spain; (R.G.-C.); (M.C.R.-C.); (M.S.-C.); (C.G.-N.)
| | | | - Francisco R. Nieto
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, 18016 Granada, Spain; (R.G.-C.); (M.C.R.-C.); (M.S.-C.); (C.G.-N.)
- Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
- Correspondence: ; Tel.: +34-958-242-056
| |
Collapse
|
9
|
Jergova S, Martinez H, Hernandez M, Schachner B, Gross S, Sagen J. Development of a Phantom Limb Pain Model in Rats: Behavioral and Histochemical Evaluation. FRONTIERS IN PAIN RESEARCH 2021; 2:675232. [PMID: 35295448 PMCID: PMC8915728 DOI: 10.3389/fpain.2021.675232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/18/2021] [Indexed: 11/13/2022] Open
Abstract
Therapeutic strategies targeting phantom limb pain (PLP) provide inadequate pain relief; therefore, a robust and clinically relevant animal model is necessary. Animal models of PLP are based on a deafferentation injury followed by autotomy behavior. Clinical studies have shown that the presence of pre-amputation pain increases the risk of developing PLP. In the current study, we used Sprague-Dawley male rats with formalin injections or constriction nerve injury at different sites or time points prior to axotomy to mimic clinical scenarios of pre-amputation inflammatory and neuropathic pain. Animals were scored daily for PLP autotomy behaviors, and several pain-related biomarkers were evaluated to discover possible underlying pathological changes. Majority displayed some degree of autotomy behavior following axotomy. Injury prior to axotomy led to more severe PLP behavior compared to animals without preceding injury. Autotomy behaviors were more directed toward the pretreatment insult origin, suggestive of pain memory. Increased levels of IL-1β in cerebrospinal fluid and enhanced microglial responses and the expression of NaV1.7 were observed in animals displaying more severe PLP outcomes. Decreased expression of GAD65/67 was consistent with greater PLP behavior. This study provides a preclinical basis for future understanding and treatment development in the management of PLP.
Collapse
|
10
|
Degradable polymeric vehicles for postoperative pain management. Nat Commun 2021; 12:1367. [PMID: 33649338 PMCID: PMC7921139 DOI: 10.1038/s41467-021-21438-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 01/20/2021] [Indexed: 01/31/2023] Open
Abstract
Effective control of pain management has the potential to significantly decrease the need for prescription opioids following a surgical procedure. While extended release products for pain management are available commercially, the implementation of a device that safely and reliably provides extended analgesia and is sufficiently flexible to facilitate a diverse array of release profiles would serve to advance patient comfort, quality of care and compliance following surgical procedures. Herein, we review current polymeric systems that could be utilized in new, controlled post-operative pain management devices and highlight where opportunities for improvement exist.
Collapse
|
11
|
L-bupivacaine Inhibition of Nociceptive Transmission in Rat Peripheral and Dorsal Horn Neurons. Anesthesiology 2021; 134:88-102. [PMID: 33166389 DOI: 10.1097/aln.0000000000003596] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Although the widely used single L-enantiomers of local anesthetics have less toxic effects on the cardiovascular and central nervous systems, the mechanisms mediating their antinociceptive actions are not well understood. The authors hypothesized that significant differences in the ion channel blocking abilities of the enantiomers of bupivacaine would be identified. METHODS The authors performed electrophysiologic analysis on rat dorsal root ganglion neurons in vitro and on spinal transmissions in vivo. RESULTS In the dorsal root ganglion, these anesthetics decreased the amplitudes of action potentials. The half-maximum inhibitory concentrations of D-enantiomer D-bupivacaine were almost equal for Aβ (29.5 μM), Aδ (29.7μM), and C (29.8 μM) neurons. However, the half-maximum inhibitory concentrations of L-bupivacaine was lower for Aδ (19.35 μM) and C (19.5 μM) neurons than for A β (79.4 μM) neurons. Moreover, D-bupivacaine almost equally inhibited tetrodotoxin-resistant (mean ± SD: 15.8 ± 10.9% of the control, n = 14, P < 0.001) and tetrodotoxin-sensitive (15.4 ± 15.6% of the control, n = 11, P = 0.004) sodium currents. In contrast, L-bupivacaine suppressed tetrodotoxin-resistant sodium currents (26.1 ± 19.5% of the control, n = 18, P < 0.001) but not tetrodotoxin-sensitive sodium currents (74.5 ± 18.2% of the control, n = 11, P = 0.477). In the spinal dorsal horn, L-bupivacaine decreased the area of pinch-evoked excitatory postsynaptic currents (39.4 ± 11.3% of the control, n = 7, P < 0.001) but not touch-evoked responses (84.2 ± 14.5% of the control, n = 6, P = 0.826). In contrast, D-bupivacaine equally decreased pinch- and touch-evoked responses (38.8 ± 9.5% of the control, n = 6, P = 0.001, 42.9 ± 11.8% of the control, n = 6, P = 0.013, respectively). CONCLUSIONS These results suggest that the L-enantiomer of bupivacaine (L-bupivacaine) effectively inhibits noxious transmission to the spinal dorsal horn by blocking action potential conduction through C and Aδ afferent fibers. EDITOR’S PERSPECTIVE
Collapse
|
12
|
Cho K, Heo J, Han J, Hong HD, Jeon H, Hwang HJ, Hong CY, Kim D, Han JW, Baek K. Industrial Applications of Dinoflagellate Phycotoxins Based on Their Modes of Action: A Review. Toxins (Basel) 2020; 12:E805. [PMID: 33353166 PMCID: PMC7766252 DOI: 10.3390/toxins12120805] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 11/24/2022] Open
Abstract
Dinoflagellates are an important group of phytoplanktons, characterized by two dissimilar flagella and distinctive features of both plants and animals. Dinoflagellate-generated harmful algal blooms (HABs) and associated damage frequently occur in coastal areas, which are concomitant with increasing eutrophication and climate change derived from anthropogenic waste and atmospheric carbon dioxide, respectively. The severe damage and harmful effects of dinoflagellate phycotoxins in the fishing industry have been recognized over the past few decades, and the management and monitoring of HABs have attracted much attention, leaving aside the industrial application of their valuable toxins. Specific modes of action of the organisms' toxins can effectively be utilized for producing beneficial materials, such as Botox and other therapeutic agents. This review aims to explore the potential industrial applications of marine dinoflagellate phycotoxins; furthermore, this review focuses on their modes of action and summarizes the available knowledge on them.
Collapse
Affiliation(s)
- Kichul Cho
- Department of Applied Marine Bioresource Science, National Marine Biodiversity Institute of Korea (MABIK), Seocheon-gun, Chungchungnam-do 33662, Korea; (K.C.); (J.H.); (H.D.H.); (H.J.); (H.-J.H.); (K.B.)
| | - Jina Heo
- Growth Engine Research Department, Chungbuk Research Institute (CRI), Chungju, Chungchungbuk-do 28517, Korea;
| | - Jinwook Han
- Department of Applied Marine Bioresource Science, National Marine Biodiversity Institute of Korea (MABIK), Seocheon-gun, Chungchungnam-do 33662, Korea; (K.C.); (J.H.); (H.D.H.); (H.J.); (H.-J.H.); (K.B.)
| | - Hyun Dae Hong
- Department of Applied Marine Bioresource Science, National Marine Biodiversity Institute of Korea (MABIK), Seocheon-gun, Chungchungnam-do 33662, Korea; (K.C.); (J.H.); (H.D.H.); (H.J.); (H.-J.H.); (K.B.)
| | - Hancheol Jeon
- Department of Applied Marine Bioresource Science, National Marine Biodiversity Institute of Korea (MABIK), Seocheon-gun, Chungchungnam-do 33662, Korea; (K.C.); (J.H.); (H.D.H.); (H.J.); (H.-J.H.); (K.B.)
| | - Hyun-Ju Hwang
- Department of Applied Marine Bioresource Science, National Marine Biodiversity Institute of Korea (MABIK), Seocheon-gun, Chungchungnam-do 33662, Korea; (K.C.); (J.H.); (H.D.H.); (H.J.); (H.-J.H.); (K.B.)
| | - Chang-Yu Hong
- Department of Environmental and Urban Research, Jeju Research Institute, Jeju-si, Jeju-do 63147, Korea;
| | - Daekyung Kim
- Daegu Center, Korea Basic Science Institute (KBSI), Daegu, Gyeongsangbuk-do 41566, Korea
| | - Jong Won Han
- Department of Applied Marine Bioresource Science, National Marine Biodiversity Institute of Korea (MABIK), Seocheon-gun, Chungchungnam-do 33662, Korea; (K.C.); (J.H.); (H.D.H.); (H.J.); (H.-J.H.); (K.B.)
| | - Kyunghwa Baek
- Department of Applied Marine Bioresource Science, National Marine Biodiversity Institute of Korea (MABIK), Seocheon-gun, Chungchungnam-do 33662, Korea; (K.C.); (J.H.); (H.D.H.); (H.J.); (H.-J.H.); (K.B.)
| |
Collapse
|
13
|
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: 18.8] [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
|
14
|
Resilience to Stress and Resilience to Pain: Lessons from Molecular Neurobiology and Genetics. Trends Mol Med 2020; 26:924-935. [PMID: 32976800 DOI: 10.1016/j.molmed.2020.03.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/18/2020] [Accepted: 03/25/2020] [Indexed: 12/12/2022]
Abstract
What biological factors account for resilience to pain or to behavioral stress? Here, we discuss examples of cellular and molecular mechanisms within disparate parts of the nervous system as contributors to such resilience. In some especially well-studied humans, it is possible to identify particular neuronal cell types in the peripheral nervous system (PNS) and pinpoint specific genes that are major contributors to pain resilience. We also discuss more complex factors that operate within the central nervous system (CNS) to confer resilience to behavioral stress. We propose that genetic and neurobiological substrates for resilience are discoverable and suggest more generally that neurology and psychiatry hold lessons for each other as investigators search for actionable, biological underpinnings of disease.
Collapse
|
15
|
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: 5.5] [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
|
16
|
Viswanath O, Urits I, Burns J, Charipova K, Gress K, McNally A, Urman RD, Welschmeyer A, Berger AA, Kassem H, Sanchez MG, Kaye AD, Eubanks TN, Cornett EM, Ngo AL. Central Neuropathic Mechanisms in Pain Signaling Pathways: Current Evidence and Recommendations. Adv Ther 2020; 37:1946-1959. [PMID: 32291648 PMCID: PMC7467462 DOI: 10.1007/s12325-020-01334-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Indexed: 12/17/2022]
Abstract
Purpose This is a comprehensive review of the current literature on central neuropathic pain mechanisms that is secondary to spinal cord injury. It reviews recent and seminal findings on the pathophysiology, diagnosis, and treatment and compares treatment options and recommendations. Recent Findings Neuropathic pain (NP) is a common complication of spinal cord injury (SCI). Chronicity of NP is attributed to increased abundance of inflammatory mediators and ion channel dysfunction leading to afferent nerve sensitization; nerve damage and nerve–glia cross talk have also been implicated. Conventional treatment is medical and has had limited success. Recent studies have made headway in identifying novel biomarkers, including microRNA and psychosocial attributes that can predict progress from SCI to chronic NP (CNP). Recent advances have provided evidence of efficacy for two promising drugs. Baclofen was able to provide good, long-lasting pain relief. Ziconotide, a voltage-gated calcium channel blocker, was studied in a small trial and was able to provide good analgesia in most participants. However, several participants had to be withdrawn because of worrisome creatine phosphokinase (CPK) elevations, and further studies are required to define its safety profile. Non-medical interventions include brain sensitization and biofeedback techniques. These methods have recently had encouraging results, albeit preliminary. Case reports of non-conventional techniques, such as hypnosis, were also reported. Summary CNP is a common complication of SCI and is a prevalent disorder with significant morbidity and disability. Conventional medical treatment is limited in efficacy. Recent studies identified baclofen and ziconotide as possible new therapies, alongside non-medical interventions. Further research into the pathophysiology is required to identify further therapy candidates. A multidisciplinary approach, including psychosocial support, medical and non-medical interventions, is likely needed to achieve therapeutic effects in this difficult to treat syndrome.
Collapse
Affiliation(s)
- Omar Viswanath
- Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ, USA
- Department of Anesthesiology, University of Arizona College of Medicine Phoenix, Phoenix, AZ, USA
- Department of Anesthesiology, Creighton University School of Medicine, Omaha, NE, USA
| | - Ivan Urits
- Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Harvard Medical School, Boston, MA, USA.
| | - James Burns
- Georgetown University School of Medicine, Washington, DC, USA
| | | | - Kyle Gress
- Georgetown University School of Medicine, Washington, DC, USA
| | | | - Richard D Urman
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Ali Welschmeyer
- Georgetown University School of Medicine, Washington, DC, USA
| | - Amnon A Berger
- Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Harvard Medical School, Boston, MA, USA
| | - Hisham Kassem
- Department of Anesthesiology, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Manuel G Sanchez
- Department of Pain Medicine, Pain Specialty Group, Newington, NH, USA
| | - Alan D Kaye
- Department of Anesthesiology, Louisiana State University Health Shreveport, Shreveport, LA, USA
| | - Treniece N Eubanks
- Department of Anesthesiology, Louisiana State University Health Shreveport, Shreveport, LA, USA
| | - Elyse M Cornett
- Department of Anesthesiology, Louisiana State University Health Shreveport, Shreveport, LA, USA
| | - Anh L Ngo
- Department of Pain Medicine, Pain Specialty Group, Newington, NH, USA
- Harvard Medical School, Boston, USA
| |
Collapse
|
17
|
Role of Potassium Ions Quantum Tunneling in the Pathophysiology of Phantom Limb Pain. Brain Sci 2020; 10:brainsci10040241. [PMID: 32325702 PMCID: PMC7226264 DOI: 10.3390/brainsci10040241] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 12/19/2022] Open
Abstract
(1) Background: multiple theories were proposed to explain the phenomenon of phantom limb pain (PLP). Nevertheless, the phenomenon is still shrouded in mystery. The aim of this study is to explore the phenomenon from a new perspective, where quantum tunneling of ions, a promising field in medical practice, might play a major role. (2) Methods: investigators designed a quantum mathematical model based on the Schrödinger equation to examine the probability of potassium ions quantum tunneling through closed membrane potassium channels to the inside of phantom axons, leading to the generation of action potential. (3) Results: the model suggests that the probability of action potential induction at a certain region of the membrane of phantom neurons, when a neuron of the stump area is stimulated over 1 mm2 surface area of the membrane available for tunneling is 1.04 × 10−2. Furthermore, upon considering two probabilities of potassium channelopathies, one that decreased the energy of the barrier by 25% and another one by 50%, the tunneling probability became 1.22 × 10−8 and 3.86 × 10−4, respectively. (4) Conclusion: quantum models of potassium ions can provide a reliable theoretical hypothesis to unveil part of the ambiguity behind PLP.
Collapse
|
18
|
Dehghan Z, Ayat H, Mohammad Ahadi A. Expression, Purification and Docking Studies on IMe-AGAP, the First Antitumor-analgesic Like Peptide from Iranian Scorpion Mesobuthus eupeus. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 19:206-216. [PMID: 33680023 PMCID: PMC7757975 DOI: 10.22037/ijpr.2019.15339.13028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Scorpion venom contains different toxins with multiple biological functions. IMe-AGAP is the first Analgesic-Antitumor like Peptide (AGAP) isolated from Iranian scorpion Mesobuthus eupeus. This peptide is similar to AGAP toxin with high analgesic activity, extracted from Chinese scorpion and inhibits NaV1.8 and NaV1.9 voltage-gated sodium channels involved in the pain pathway. In this study, IMe-AGAP was cloned in a prokaryotic expression vector; expression of toxin in Escherichia coli (E. coli) was assayed and then purified. In in-silico studies, peptide sequence was compared with other scorpion analgesic toxins. The structures of IMe-AGAP and sodium channels were modeled using homology modeling. Structural evaluation and stereo-chemical analysis of modeled structures were performed using RAMPAGE web server Ramachandran plots. Hex Server was used to investigate the interactions between IMe-AGAP and S3-S4 and also S5-S6 segments of NaV1.8 and NaV1.9. Binding energies calculation was used for evaluation of protein docking. Soluble expression of IMe-AGAP in bacteria was investigated by SDS-PAGE analysis. Pure recombinant protein was obtained by Ni-NTA affinity chromatography. The results of three-dimensional structure prediction showed βαββ topology for the toxin that is similar to the conserved structure of α-toxins. Comparison analysis between IMe-AGAP and AGAP toxins exhibited high similarity in homology modeling. Docking analysis demonstrated that IMe-AGAP can interact with NaV1.8 and NaV1.9 domains involved in pain. According to the results of homology studies and docking, IMe-AGAP might be a novel potential drug for pain treatment.
Collapse
|
19
|
Yasko JR, Moss IL, Mains RE. Transcriptional Profiling of Non-injured Nociceptors After Spinal Cord Injury Reveals Diverse Molecular Changes. Front Mol Neurosci 2019; 12:284. [PMID: 32038157 PMCID: PMC6988781 DOI: 10.3389/fnmol.2019.00284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/08/2019] [Indexed: 01/01/2023] Open
Abstract
Traumatic spinal cord injury (SCI) has devastating implications for patients, including a high predisposition for developing chronic pain distal to the site of injury. Chronic pain develops weeks to months after injury, consequently, patients are treated after irreparable changes have occurred. Nociceptors are central to chronic pain; however, the diversity of this cellular population presents challenges to understanding mechanisms and attributing pain modalities to specific cell types. To begin to address how peripheral sensory neurons below the injury level may contribute to the below-level pain reported by SCI patients, we examined SCI-induced changes in gene expression in lumbar dorsal root ganglia (DRG) below the site of injury. SCI was performed at the T10 vertebral level, with injury produced by a vessel clip with a closing pressure of 15 g for 1 min. Alterations in gene expression produce long-term sensory changes, therefore, we were interested in studying SCI-induced transcripts before the onset of chronic pain, which may trigger changes in downstream signaling pathways and ultimately facilitate the transmission of pain. To examine changes in the nociceptor subpopulation in DRG distal to the site of injury, we retrograde labeled sensory neurons projecting to the hairy hindpaw skin with fluorescent dye and collected the corresponding lumbar (L2–L6) DRG 4 days post-injury. Following dissociation, labeled neurons were purified by fluorescence-activated cell sorting (FACS). RNA was extracted from sorted sensory neurons of naïve, sham, or SCI mice and sequenced. Transcript abundances validated that the desired population of nociceptors were isolated. Cross-comparisons to data sets from similar studies confirmed, we were able to isolate our cells of interest and identify a unique pattern of gene expression within a subpopulation of neurons projecting to the hairy hindpaw skin. Differential gene expression analysis showed high expression levels and significant transcript changes 4 days post-injury in SCI cell populations relevant to the onset of chronic pain. Regulatory interrelationships predicted by pathway analysis implicated changes within the synaptogenesis signaling pathway as well as networks related to inflammatory signaling mechanisms, suggesting a role for synaptic plasticity and a correlation with pro-inflammatory signaling in the transition from acute to chronic pain.
Collapse
Affiliation(s)
- Jessica R Yasko
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, United States
| | - Isaac L Moss
- Department of Orthopedic Surgery and the Comprehensive Spine Center, University of Connecticut Health Center, Farmington, CT, United States
| | - Richard E Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, United States
| |
Collapse
|
20
|
Rong G, Tuttle EE, Neal Reilly A, Clark HA. Recent Developments in Nanosensors for Imaging Applications in Biological Systems. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2019; 12:109-128. [PMID: 30857408 PMCID: PMC6958676 DOI: 10.1146/annurev-anchem-061417-125747] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Sensors are key tools for monitoring the dynamic changes of biomolecules and biofunctions that encode valuable information that helps us understand underlying biological processes of fundamental importance. Because of their distinctive size-dependent physicochemical properties, materials with nanometer scales have recently emerged as promising candidates for biological sensing applications by offering unique insights into real-time changes of key physiological parameters. This review focuses on recent advances in imaging-based nanosensor developments and applications categorized by their signal transduction mechanisms, namely, fluorescence, plasmonics, MRI, and photoacoustics. We further discuss the synergy created by multimodal nanosensors in which sensor components work based on two or more signal transduction mechanisms.
Collapse
Affiliation(s)
- Guoxin Rong
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, USA;
| | - Erin E Tuttle
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, USA
| | - Ashlyn Neal Reilly
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, USA;
| | - Heather A Clark
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, USA;
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, USA
| |
Collapse
|
21
|
Matsuda M, Huh Y, Ji RR. Roles of inflammation, neurogenic inflammation, and neuroinflammation in pain. J Anesth 2019; 33:131-139. [PMID: 30448975 PMCID: PMC6813778 DOI: 10.1007/s00540-018-2579-4] [Citation(s) in RCA: 269] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/26/2018] [Indexed: 12/15/2022]
Abstract
Inflammation is the body's response to injury and infection, involving a complex biological response of the somatosensory, immune, autonomic, and vascular systems. Inflammatory mediators such as prostaglandin, proinflammatory cytokines, and chemokines induce pain via direct activation of nociceptors, the primary sensory neurons that detect noxious stimuli. Neurogenic inflammation is triggered by nerve activation and results in neuropeptide release and rapid plasma extravasation and edema, contributing to pain conditions such as headache. Neuroinflammation is a localized inflammation in the peripheral nervous system (PNS) and central nervous system (CNS). A characteristic feature of neuroinflammation is the activation of glial cells in dorsal root ganglia, spinal cord, and brain which leads to the production of proinflammatory cytokines and chemokines in the PNS and CNS that drives peripheral sensitization and central sensitization. Here, we discuss the distinct roles of inflammation, neurogenic inflammation, and neuroinflammation in the regulation of different types of pain conditions, with a special focus on neuroinflammation in postoperative pain and opioid-induced hyperalgesia.
Collapse
Affiliation(s)
- Megumi Matsuda
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, 3 Genome CT, MSRB3 Room 6148, Durham, NC, 27710, USA.
- Research Unit for the Neurobiology of Pain, Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Yul Huh
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, 3 Genome CT, MSRB3 Room 6148, Durham, NC, 27710, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, 3 Genome CT, MSRB3 Room 6148, Durham, NC, 27710, USA.
| |
Collapse
|
22
|
Rong G, Kim EH, Qiang Y, Di W, Zhong Y, Zhao X, Fang H, Clark HA. Imaging Sodium Flux during Action Potentials in Neurons with Fluorescent Nanosensors and Transparent Microelectrodes. ACS Sens 2018; 3:2499-2505. [PMID: 30358986 DOI: 10.1021/acssensors.8b00903] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Sodium flux plays a pivotal role in neurobiological processes including initiation of action potentials and regulation of neuronal cell excitability. However, unlike the wide range of fluorescent calcium indicators used extensively for cellular studies, the choice of sodium probes remains limited. We have previously demonstrated optode-based nanosensors (OBNs) for detecting sodium ions with advantageous modular properties such as tunable physiological sensing range, full reversibility, and superb selectivity against key physiological interfering ion potassium. (1) Motivated by bridging the gap between the great interest in sodium imaging of neuronal cell activity as an alternative to patch clamp and limited choices of optical sodium indicators, in this Letter we report the application of nanosensors capable of detecting intracellular sodium flux in isolated rat dorsal root ganglion neurons during electrical stimulation using transparent microelectrodes. Taking advantage of the ratiometric detection scheme offered by this fluorescent modular sensing platform, we performed dual color imaging of the sensor to monitor the intracellular sodium currents underlying trains of action potentials in real time. The combination of nanosensors and microelectrodes for monitoring neuronal sodium dynamics is a novel tool for investigating the regulatory role of sodium ions involved during neural activities.
Collapse
|
23
|
Petersen BA, Nanivadekar AC, Chandrasekaran S, Fisher LE. Phantom limb pain: peripheral neuromodulatory and neuroprosthetic approaches to treatment. Muscle Nerve 2018; 59:154-167. [DOI: 10.1002/mus.26294] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Bailey A. Petersen
- Department of Bioengineering; University of Pittsburgh; 3520 Fifth Avenue, Pittsburgh Pennsylvania 15213 USA
| | - Ameya C. Nanivadekar
- Department of Bioengineering; University of Pittsburgh; 3520 Fifth Avenue, Pittsburgh Pennsylvania 15213 USA
| | - Santosh Chandrasekaran
- Department of Physical Medicine and Rehabilitation; University of Pittsburgh; Pittsburgh Pennsylvania USA
| | - Lee E. Fisher
- Department of Bioengineering; University of Pittsburgh; 3520 Fifth Avenue, Pittsburgh Pennsylvania 15213 USA
- Department of Physical Medicine and Rehabilitation; University of Pittsburgh; Pittsburgh Pennsylvania USA
| |
Collapse
|
24
|
Tu H, Zhang D, Li YL. Cellular and Molecular Mechanisms Underlying Arterial Baroreceptor Remodeling in Cardiovascular Diseases and Diabetes. Neurosci Bull 2018; 35:98-112. [PMID: 30146675 DOI: 10.1007/s12264-018-0274-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 05/31/2018] [Indexed: 01/23/2023] Open
Abstract
Clinical trials and animal experimental studies have demonstrated an association of arterial baroreflex impairment with the prognosis and mortality of cardiovascular diseases and diabetes. As a primary part of the arterial baroreflex arc, the pressure sensitivity of arterial baroreceptors is blunted and involved in arterial baroreflex dysfunction in cardiovascular diseases and diabetes. Changes in the arterial vascular walls, mechanosensitive ion channels, and voltage-gated ion channels contribute to the attenuation of arterial baroreceptor sensitivity. Some endogenous substances (such as angiotensin II and superoxide anion) can modulate these morphological and functional alterations through intracellular signaling pathways in impaired arterial baroreceptors. Arterial baroreceptors can be considered as a potential therapeutic target to improve the prognosis of patients with cardiovascular diseases and diabetes.
Collapse
Affiliation(s)
- Huiyin Tu
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Dongze Zhang
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
25
|
Collins KL, Russell HG, Schumacher PJ, Robinson-Freeman KE, O'Conor EC, Gibney KD, Yambem O, Dykes RW, Waters RS, Tsao JW. A review of current theories and treatments for phantom limb pain. J Clin Invest 2018; 128:2168-2176. [PMID: 29856366 DOI: 10.1172/jci94003] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Following amputation, most amputees still report feeling the missing limb and often describe these feelings as excruciatingly painful. Phantom limb sensations (PLS) are useful while controlling a prosthesis; however, phantom limb pain (PLP) is a debilitating condition that drastically hinders quality of life. Although such experiences have been reported since the early 16th century, the etiology remains unknown. Debate continues regarding the roles of the central and peripheral nervous systems. Currently, the most posited mechanistic theories rely on neuronal network reorganization; however, greater consideration should be given to the role of the dorsal root ganglion within the peripheral nervous system. This Review provides an overview of the proposed mechanistic theories as well as an overview of various treatments for PLP.
Collapse
Affiliation(s)
| | - Hannah G Russell
- Department of Neurology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Patrick J Schumacher
- Department of Neurology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | | | - Ellen C O'Conor
- Department of Neurology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Kyla D Gibney
- Department of Neurology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Olivia Yambem
- Department of Neurology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Robert W Dykes
- School of Physical and Occupational Therapy, McGill University, Montreal, Quebec, Canada
| | | | - Jack W Tsao
- Department of Neurology, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Neurology, Memphis Veterans Affairs Medical Center, Memphis, Tennessee, USA.,Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, Tennessee, USA
| |
Collapse
|
26
|
Buyan A, Sun D, Corry B. Protonation state of inhibitors determines interaction sites within voltage-gated sodium channels. Proc Natl Acad Sci U S A 2018; 115:E3135-E3144. [PMID: 29467289 PMCID: PMC5889629 DOI: 10.1073/pnas.1714131115] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Voltage-gated sodium channels are essential for carrying electrical signals throughout the body, and mutations in these proteins are responsible for a variety of disorders, including epilepsy and pain syndromes. As such, they are the target of a number of drugs used for reducing pain or combatting arrhythmias and seizures. However, these drugs affect all sodium channel subtypes found in the body. Designing compounds to target select sodium channel subtypes will provide a new therapeutic pathway and would maximize treatment efficacy while minimizing side effects. Here, we examine the binding preferences of nine compounds known to be sodium channel pore blockers in molecular dynamics simulations. We use the approach of replica exchange solute tempering (REST) to gain a more complete understanding of the inhibitors' behavior inside the pore of NavMs, a bacterial sodium channel, and NavPas, a eukaryotic sodium channel. Using these simulations, we are able to show that both charged and neutral compounds partition into the bilayer, but neutral forms more readily cross it. We show that there are two possible binding sites for the compounds: (i) a site on helix 6, which has been previously determined by many experimental and computational studies, and (ii) an additional site, occupied by protonated compounds in which the positively charged part of the drug is attracted into the selectivity filter. Distinguishing distinct binding poses for neutral and charged compounds is essential for understanding the nature of pore block and will aid the design of subtype-selective sodium channel inhibitors.
Collapse
Affiliation(s)
- Amanda Buyan
- Research School of Biology, Australian National University, Acton, ACT 2601, Australia
| | - Delin Sun
- Research School of Biology, Australian National University, Acton, ACT 2601, Australia
| | - Ben Corry
- Research School of Biology, Australian National University, Acton, ACT 2601, Australia
| |
Collapse
|
27
|
|
28
|
Chen L, Huang J, Zhao P, Persson AK, Dib-Hajj FB, Cheng X, Tan A, Waxman SG, Dib-Hajj SD. Conditional knockout of Na V1.6 in adult mice ameliorates neuropathic pain. Sci Rep 2018; 8:3845. [PMID: 29497094 PMCID: PMC5832877 DOI: 10.1038/s41598-018-22216-w] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/19/2018] [Indexed: 01/09/2023] Open
Abstract
Voltage-gated sodium channels NaV1.7, NaV1.8 and NaV1.9 have been the focus for pain studies because their mutations are associated with human pain disorders, but the role of NaV1.6 in pain is less understood. In this study, we selectively knocked out NaV1.6 in dorsal root ganglion (DRG) neurons, using NaV1.8-Cre directed or adeno-associated virus (AAV)-Cre mediated approaches, and examined the specific contribution of NaV1.6 to the tetrodotoxin-sensitive (TTX-S) current in these neurons and its role in neuropathic pain. We report here that NaV1.6 contributes up to 60% of the TTX-S current in large, and 34% in small DRG neurons. We also show NaV1.6 accumulates at nodes of Ranvier within the neuroma following spared nerve injury (SNI). Although NaV1.8-Cre driven NaV1.6 knockout does not alter acute, inflammatory or neuropathic pain behaviors, AAV-Cre mediated NaV1.6 knockout in adult mice partially attenuates SNI-induced mechanical allodynia. Additionally, AAV-Cre mediated NaV1.6 knockout, mostly in large DRG neurons, significantly attenuates excitability of these neurons after SNI and reduces NaV1.6 accumulation at nodes of Ranvier at the neuroma. Together, NaV1.6 in NaV1.8-positive neurons does not influence pain thresholds under normal or pathological conditions, but NaV1.6 in large NaV1.8-negative DRG neurons plays an important role in neuropathic pain.
Collapse
Affiliation(s)
- Lubin Chen
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Jianying Huang
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Anna-Karin Persson
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Fadia B Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Xiaoyang Cheng
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Andrew Tan
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA. .,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA. .,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA.
| |
Collapse
|
29
|
Wu Y, Ma H, Zhang F, Zhang C, Zou X, Cao Z. Selective Voltage-Gated Sodium Channel Peptide Toxins from Animal Venom: Pharmacological Probes and Analgesic Drug Development. ACS Chem Neurosci 2018; 9:187-197. [PMID: 29161016 DOI: 10.1021/acschemneuro.7b00406] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (Navs) play critical roles in action potential generation and propagation. Nav channelopathy as well as pathological sensitization contribute to allodynia and hyperalgesia. Recent evidence has demonstrated the significant roles of Nav subtypes (Nav1.3, 1.7, 1.8, and 1.9) in nociceptive transduction, and therefore these Navs may represent attractive targets for analgesic drug discovery. Animal toxins are structurally diverse peptides that are highly potent yet selective on ion channel subtypes and therefore represent valuable probes to elucidate the structures, gating properties, and cellular functions of ion channels. In this review, we summarize recent advances on peptide toxins from animal venom that selectively target Nav1.3, 1.7, 1.8, and 1.9, along with their potential in analgesic drug discovery.
Collapse
Affiliation(s)
- Ying Wu
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Hui Ma
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Fan Zhang
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Chunlei Zhang
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaohan Zou
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Zhengyu Cao
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
30
|
Bang S, Yoo J, Gong X, Liu D, Han Q, Luo X, Chang W, Chen G, Im ST, Kim YH, Strong JA, Zhang MZ, Zhang JM, Lee SY, Ji RR. Differential Inhibition of Nav1.7 and Neuropathic Pain by Hybridoma-Produced and Recombinant Monoclonal Antibodies that Target Nav1.7 : Differential activities of Nav1.7-targeting monoclonal antibodies. Neurosci Bull 2018; 34:22-41. [PMID: 29333591 PMCID: PMC5799132 DOI: 10.1007/s12264-018-0203-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 12/18/2017] [Indexed: 12/21/2022] Open
Abstract
The voltage-gated Na+ channel subtype Nav1.7 is important for pain and itch in rodents and humans. We previously showed that a Nav1.7-targeting monoclonal antibody (SVmab) reduces Na+ currents and pain and itch responses in mice. Here, we investigated whether recombinant SVmab (rSVmab) binds to and blocks Nav1.7 similar to SVmab. ELISA tests revealed that SVmab was capable of binding to Nav1.7-expressing HEK293 cells, mouse DRG neurons, human nerve tissue, and the voltage-sensor domain II of Nav1.7. In contrast, rSVmab showed no or weak binding to Nav1.7 in these tests. Patch-clamp recordings showed that SVmab, but not rSVmab, markedly inhibited Na+ currents in Nav1.7-expressing HEK293 cells. Notably, electrical field stimulation increased the blocking activity of SVmab and rSVmab in Nav1.7-expressing HEK293 cells. SVmab was more effective than rSVmab in inhibiting paclitaxel-induced mechanical allodynia. SVmab also bound to human DRG neurons and inhibited their Na+ currents. Finally, potential reasons for the differential efficacy of SVmab and rSVmab and future directions are discussed.
Collapse
Affiliation(s)
- Sangsu Bang
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Jiho Yoo
- Department of Biochemistry, Duke University Medical Center, 303 Research Drive, Durham, NC, 27710, USA
| | - Xingrui Gong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0531, USA
- Department of Anesthesiology, Shanghai Children's Medical Center, Shanghai, 200127, China
| | - Di Liu
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Qingjian Han
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Xin Luo
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Wonseok Chang
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
- Department of Physiology and Biophysics, College of Medicine, Eulji University, 143-5 Yongdu-Dong, Jung-Gu, Daejeon, 34824, Korea
| | - Gang Chen
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Sang-Taek Im
- Department of Physiology, College of Medicine, Gachon University, Incheon, 21999, Korea
| | - Yong Ho Kim
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
- Department of Physiology, College of Medicine, Gachon University, Incheon, 21999, Korea
| | - Judith A Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0531, USA
| | - Ma-Zhong Zhang
- Department of Anesthesiology, Shanghai Children's Medical Center, Shanghai, 200127, China
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0531, USA.
| | - Seok-Yong Lee
- Department of Biochemistry, Duke University Medical Center, 303 Research Drive, Durham, NC, 27710, USA.
| | - Ru-Rong Ji
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA.
| |
Collapse
|
31
|
Physical basis of specificity and delayed binding of a subtype selective sodium channel inhibitor. Sci Rep 2018; 8:1356. [PMID: 29358762 PMCID: PMC5778059 DOI: 10.1038/s41598-018-19850-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 01/09/2018] [Indexed: 12/19/2022] Open
Abstract
Nerve and muscle signalling is controlled by voltage-gated sodium (Nav) channels which are the targets of local anesthetics, anti-epileptics and anti-arrythmics. Current medications do not selectively target specific types of Nav found in the body, but compounds that do so have the potential to be breakthrough treatments for chronic pain, epilepsy and other neuronal disorders. We use long computer simulations totaling more than 26 μs to show how a promising lead compound can target one Nav implicated in pain perception and specific channels found in bacteria, and accurately predict the affinity of the compound to different channel types. Most importantly, we provide two explanations for the slow kinetics of this class of compound that limits their therapeutic utility. Firstly, the negative charge on the compound is essential for high affinity binding but is also responsible for energetic barriers that slow binding. Secondly, the compound has to undergo a conformational reorientation during the binding process. This knowledge aids the design of compounds affecting specific eukaryotic and bacterial channels and suggests routes for future drug development.
Collapse
|
32
|
Abstract
Acutely ill patients are challenging to frontline nurses because they frequently also have multiple chronic conditions. This article empowers all nurses to develop a foundational understanding of the physiology of acute and chronic pain. The skills, knowledge, and attitude to care for patients experiencing pain are a legal and ethical responsibility of all nurses. This article discusses the physiology of pain to include the neuronal receptors that respond to various painful stimuli, substances that stimulate nociceptors, the nerve pathways, modulation of the perception of pain, and acute verses chronic physiologic changes.
Collapse
|
33
|
Effect of an Acid-sensing Ion Channels Inhibitor on Pain-related Behavior by Nucleus Pulposus Applied on the Nerve Root in Rats. Spine (Phila Pa 1976) 2017; 42:E633-E641. [PMID: 27879566 DOI: 10.1097/brs.0000000000001918] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Controlled, interventional animal study. OBJECTIVE To examine the effect of an inhibitor of acid-sensing ion channel 3 (ASIC3) on pain-related behavior induced by application of the nucleus pulposus (NP) onto the dorsal root ganglion (DRG) in rats. SUMMARY OF BACKGROUND DATA ASIC3 is associated with acidosis pain in inflamed or ischemic tissues and is expressed in sensory neurons and NP cells. The ASIC3 inhibitor, APETx2, increases the mechanical threshold of pain in models of knee osteoarthritis or postoperative pain. However, the efficacy of APETx2 for pain relief in the NP application model remains unknown. METHODS Autologous NP was applied to the left L5 nerve root of 183 adult female Sprague-Dawley rats. The DRGs were treated with NP plus one of the following four treatments: saline solution (SM), low (0.01 μg: LD), medium (0.1 μg: MD), or high dose (1.0 μg: HD) of APETx2. Behavioral testing was performed to investigate the mechanical withdrawal threshold using von Frey hairs. Expression of nerve growth factor, hypoxia-inducible factor-1α (HIF1α), activating transcription factor-3, and ionized calcium-binding adaptor molecule-1 was evaluated using immunohistochemistry. Statistical differences among multiple groups were assessed using the Steel test, the Tukey-Kramer test, and the Dunnett test. P < 0.05 were considered significant. RESULTS The thresholds in the HD group were higher than those in the SM group at Days 14 and 21 (P < 0.05). In the MD group, the threshold was higher than in the SM group at Day 14 (P < 0.05). High doses of APETx2 reduced the expression of HIF1α after Day 14 compared with the SM group (P < 0.05). CONCLUSION APETx2 significantly improved pain-related behavior in a dose-dependent manner. APETx2 may inhibit ASIC3 and partly inhibit Nav1.8 channels. This ASIC3 channel inhibitor may be a potential therapeutic agent in early-stage lumbar disc herniation. LEVEL OF EVIDENCE N/A.
Collapse
|
34
|
Expression and Role of Voltage-Gated Sodium Channels in Human Dorsal Root Ganglion Neurons with Special Focus on Nav1.7, Species Differences, and Regulation by Paclitaxel. Neurosci Bull 2017; 34:4-12. [PMID: 28424991 PMCID: PMC5648619 DOI: 10.1007/s12264-017-0132-3] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/08/2017] [Indexed: 12/20/2022] Open
Abstract
Voltage-gated sodium channels (Navs) play an important role in human pain sensation. However, the expression and role of Nav subtypes in native human sensory neurons are unclear. To address this issue, we obtained human dorsal root ganglion (hDRG) tissues from healthy donors. PCR analysis of seven DRG-expressed Nav subtypes revealed that the hDRG has higher expression of Nav1.7 (~50% of total Nav expression) and lower expression of Nav1.8 (~12%), whereas the mouse DRG has higher expression of Nav1.8 (~45%) and lower expression of Nav1.7 (~18%). To mimic Nav regulation in chronic pain, we treated hDRG neurons in primary cultures with paclitaxel (0.1–1 μmol/L) for 24 h. Paclitaxel increased the Nav1.7 but not Nav1.8 expression and also increased the transient Na+ currents and action potential firing frequency in small-diameter (<50 μm) hDRG neurons. Thus, the hDRG provides a translational model in which to study “human pain in a dish” and test new pain therapeutics.
Collapse
|
35
|
Role of the Excitability Brake Potassium Current I KD in Cold Allodynia Induced by Chronic Peripheral Nerve Injury. J Neurosci 2017; 37:3109-3126. [PMID: 28179555 DOI: 10.1523/jneurosci.3553-16.2017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/24/2017] [Accepted: 01/26/2017] [Indexed: 11/21/2022] Open
Abstract
Cold allodynia is a common symptom of neuropathic and inflammatory pain following peripheral nerve injury. The mechanisms underlying this disabling sensory alteration are not entirely understood. In primary somatosensory neurons, cold sensitivity is mainly determined by a functional counterbalance between cold-activated TRPM8 channels and Shaker-like Kv1.1-1.2 channels underlying the excitability brake current IKD Here we studied the role of IKD in damage-triggered painful hypersensitivity to innocuous cold. We found that cold allodynia induced by chronic constriction injury (CCI) of the sciatic nerve in mice, was related to both an increase in the proportion of cold-sensitive neurons (CSNs) in DRGs contributing to the sciatic nerve, and a decrease in their cold temperature threshold. IKD density was reduced in high-threshold CSNs from CCI mice compared with sham animals, with no differences in cold-induced TRPM8-dependent current density. The electrophysiological properties and neurochemical profile of CSNs revealed an increase of nociceptive-like phenotype among neurons from CCI animals compared with sham mice. These results were validated using a mathematical model of CSNs, including IKD and TRPM8, showing that a reduction in IKD current density shifts the thermal threshold to higher temperatures and that the reduction of this current induces cold sensitivity in former cold-insensitive neurons expressing low levels of TRPM8-like current. Together, our results suggest that cold allodynia is largely due to a functional downregulation of IKD in both high-threshold CSNs and in a subpopulation of polymodal nociceptors expressing TRPM8, providing a general molecular and neural mechanism for this sensory alteration.SIGNIFICANCE STATEMENT This paper unveils the critical role of the brake potassium current IKD in damage-triggered cold allodynia. Using a well-known form of nerve injury and combining behavioral analysis, calcium imaging, patch clamping, and pharmacological tools, validated by mathematical modeling, we determined that the functional expression of IKD is reduced in sensory neurons in response to peripheral nerve damage. This downregulation not only enhances cold sensitivity of high-threshold cold thermoreceptors signaling cold discomfort, but it also transforms a subpopulation of polymodal nociceptors signaling pain into neurons activated by mild temperature drops. Our results suggest that cold allodynia is linked to a reduction of IKD in both high-threshold cold thermoreceptors and nociceptors expressing TRPM8, providing a general model for this form of cold-induced pain.
Collapse
|
36
|
|
37
|
Bothriurus bonariensis scorpion venom activates voltage-dependent sodium channels in insect and mammalian nervous systems. Chem Biol Interact 2016; 258:1-9. [DOI: 10.1016/j.cbi.2016.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 08/05/2016] [Accepted: 08/15/2016] [Indexed: 01/12/2023]
|
38
|
Smith NE, Corry B. Mutant bacterial sodium channels as models for local anesthetic block of eukaryotic proteins. Channels (Austin) 2016; 10:225-37. [PMID: 26852716 DOI: 10.1080/19336950.2016.1148224] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Voltage gated sodium channels are the target of a range of local anesthetic, anti-epileptic and anti-arrhythmic compounds. But, gaining a molecular level understanding of their mode of action is difficult as we only have atomic resolution structures of bacterial sodium channels not their eukaryotic counterparts. In this study we used molecular dynamics simulations to demonstrate that the binding sites of both the local anesthetic benzocaine and the anti-epileptic phenytoin to the bacterial sodium channel NavAb can be altered significantly by the introduction of point mutations. Free energy techniques were applied to show that increased aromaticity in the pore of the channel, used to emulate the aromatic residues observed in eukaryotic Nav1.2, led to changes in the location of binding and dissociation constants of each drug relative to wild type NavAb. Further, binding locations and dissociation constants obtained for both benzocaine (660 μM) and phenytoin (1 μM) in the mutant channels were within the range expected from experimental values obtained from drug binding to eukaryotic sodium channels, indicating that these mutant NavAb may be a better model for drug binding to eukaryotic channels than the wild type.
Collapse
Affiliation(s)
- Natalie E Smith
- a Research School of Biology, Australian National University , Canberra , ACT , Australia
| | - Ben Corry
- a Research School of Biology, Australian National University , Canberra , ACT , Australia
| |
Collapse
|
39
|
Barbosa C, Cummins TR. Unusual Voltage-Gated Sodium Currents as Targets for Pain. CURRENT TOPICS IN MEMBRANES 2016; 78:599-638. [PMID: 27586296 DOI: 10.1016/bs.ctm.2015.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pain is a serious health problem that impacts the lives of many individuals. Hyperexcitability of peripheral sensory neurons contributes to both acute and chronic pain syndromes. Because voltage-gated sodium currents are crucial to the transmission of electrical signals in peripheral sensory neurons, the channels that underlie these currents are attractive targets for pain therapeutics. Sodium currents and channels in peripheral sensory neurons are complex. Multiple-channel isoforms contribute to the macroscopic currents in nociceptive sensory neurons. These different isoforms exhibit substantial variations in their kinetics and pharmacology. Furthermore, sodium current complexity is enhanced by an array of interacting proteins that can substantially modify the properties of voltage-gated sodium channels. Resurgent sodium currents, atypical currents that can enhance recovery from inactivation and neuronal firing, are increasingly being recognized as playing potentially important roles in sensory neuron hyperexcitability and pain sensations. Here we discuss unusual sodium channels and currents that have been identified in nociceptive sensory neurons, describe what is known about the molecular determinants of the complex sodium currents in these neurons. Finally, we provide an overview of therapeutic strategies to target voltage-gated sodium currents in nociceptive neurons.
Collapse
Affiliation(s)
- C Barbosa
- Indiana University School of Medicine, Indianapolis, IN, United States
| | - T R Cummins
- Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
40
|
Chuquilin M, Alghalith Y, Fernandez KH. Neurocutaneous disease. J Am Acad Dermatol 2016; 74:197-212. [DOI: 10.1016/j.jaad.2015.04.060] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 04/06/2015] [Accepted: 04/22/2015] [Indexed: 12/14/2022]
|
41
|
Zamponi GW, Han C, Waxman SG. Voltage-Gated Ion Channels as Molecular Targets for Pain. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
42
|
Li N, Liu Z, Wang G, Wang S. Downregulation of the sodium channel Nav1.6 by potential transcriptomic deregulation may explain sensory deficits in critical illness neuropathy. Life Sci 2015; 143:231-6. [PMID: 26562765 DOI: 10.1016/j.lfs.2015.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 10/08/2015] [Accepted: 11/07/2015] [Indexed: 11/24/2022]
Abstract
AIMS Sepsis patients and other patients in the critical care settings are at very high risk of mortality due to the primary illness. However, a fraction of patients, even after showing initial clinical improvement, deteriorates relentlessly at later stages. Increasingly, it is being identified that this is mostly due to dysfunction of the neurological system. MAIN METHODS We obtained peripheral nerve biopsies from the sural nerve from ICU patients. Nav1.6 expression was significantly diminished. The expression of cellular membrane anchoring protein for Nav1.6, ankyrin, remained unaffected, suggesting that genomic repression may be responsible for the diminished expression of the sodium channels. We examined the expression of two regulatory transcription factors: (a) a positive regulator YY1 that binds to the promoter region of sodium channels and (b) an upstream negative neuronal regulator REST. KEY FINDINGS REST expression was significantly elevated, while YY1 expression was diminished. Finally, we also observed that the cholinergic synthetic enzyme acyltransferase was also significantly diminished in sensory nerve lysates. Finally, circulating antibodies was detected in the peripheral blood against all the major sodium channels Nav1.6, 1.8 and 1.9, which contribute to the development and propagation of action potentials. SIGNIFICANCE This may potentially explain why its dysfunction affects neurological functions across all systems of the body during critical illness. The underlying mechanism of why the expression of the REST transcriptional factor is affected in critical illnesses remains our future goals of investigation.
Collapse
Affiliation(s)
- Nan Li
- Department of Intensive Care Unit, The First Hospital of Jilin University, Jilin 130021, China
| | - Zhongmin Liu
- Department of Intensive Care Unit, The First Hospital of Jilin University, Jilin 130021, China
| | - Guang Wang
- Department of Intensive Care Unit, The First Hospital of Jilin University, Jilin 130021, China
| | - Shiji Wang
- Department of Intensive Care Unit, The First Hospital of Jilin University, Jilin 130021, China.
| |
Collapse
|
43
|
Zhang D, Muelleman RL, Li YL. Angiotensin II-superoxide-NFκB signaling and aortic baroreceptor dysfunction in chronic heart failure. Front Neurosci 2015; 9:382. [PMID: 26528122 PMCID: PMC4607814 DOI: 10.3389/fnins.2015.00382] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 10/02/2015] [Indexed: 11/13/2022] Open
Abstract
Chronic heart failure (CHF) affects approximately 5.7 million people in the United States. Increasing evidence from both clinical and experimental studies indicates that the sensitivity of arterial baroreflex is blunted in the CHF state, which is a predictive risk factor for sudden cardiac death. Normally, the arterial baroreflex regulates blood pressure and heart rate through sensing mechanical alteration of arterial vascular walls by baroreceptor terminals in the aortic arch and carotid sinus. There are aortic baroreceptor neurons in the nodose ganglion (NG), which serve as the main afferent component of the arterial baroreflex. Functional changes of baroreceptor neurons are involved in the arterial baroreflex dysfunction in CHF. In the CHF state, circulating angiotensin II (Ang II) and local Ang II concentration in the NG are elevated, and AT1R mRNA and protein are overexpressed in the NG. Additionally, Ang II-superoxide-NFκB signaling pathway regulates the neuronal excitability of aortic baroreceptors through influencing the expression and activation of Nav channels in aortic baroreceptors, and subsequently causes the impairment of the arterial baroreflex in CHF. These new findings provide a basis for potential pharmacological interventions for the improvement of the arterial baroreflex sensitivity in the CHF state. This review summarizes the mechanisms responsible for the arterial baroreflex dysfunction in CHF.
Collapse
Affiliation(s)
- Dongze Zhang
- Department of Emergency Medicine, University of Nebraska Medical Center Omaha, NE, USA
| | - Robert L Muelleman
- Department of Emergency Medicine, University of Nebraska Medical Center Omaha, NE, USA
| | - Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center Omaha, NE, USA
| |
Collapse
|
44
|
Kharatmal SB, Singh JN, Sharma SS. Calpain inhibitor, MDL 28170 confer electrophysiological, nociceptive and biochemical improvement in diabetic neuropathy. Neuropharmacology 2015; 97:113-21. [DOI: 10.1016/j.neuropharm.2015.05.040] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 05/14/2015] [Accepted: 05/27/2015] [Indexed: 10/23/2022]
|
45
|
Barbosa C, Tan ZY, Wang R, Xie W, Strong JA, Patel RR, Vasko MR, Zhang JM, Cummins TR. Navβ4 regulates fast resurgent sodium currents and excitability in sensory neurons. Mol Pain 2015; 11:60. [PMID: 26408173 PMCID: PMC4582632 DOI: 10.1186/s12990-015-0063-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 09/10/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Increased electrical activity in peripheral sensory neurons including dorsal root ganglia (DRG) and trigeminal ganglia neurons is an important mechanism underlying pain. Voltage gated sodium channels (VGSC) contribute to the excitability of sensory neurons and are essential for the upstroke of action potentials. A unique type of VGSC current, resurgent current (INaR), generates an inward current at repolarizing voltages through an alternate mechanism of inactivation referred to as open-channel block. INaRs are proposed to enable high frequency firing and increased INaRs in sensory neurons are associated with pain pathologies. While Nav1.6 has been identified as the main carrier of fast INaR, our understanding of the mechanisms that contribute to INaR generation is limited. Specifically, the open-channel blocker in sensory neurons has not been identified. Previous studies suggest Navβ4 subunit mediates INaR in central nervous system neurons. The goal of this study was to determine whether Navβ4 regulates INaR in DRG sensory neurons. RESULTS Our immunocytochemistry studies show that Navβ4 expression is highly correlated with Nav1.6 expression predominantly in medium-large diameter rat DRG neurons. Navβ4 knockdown decreased endogenous fast INaR in medium-large diameter neurons as measured with whole-cell voltage clamp. Using a reduced expression system in DRG neurons, we isolated recombinant human Nav1.6 sodium currents in rat DRG neurons and found that overexpression of Navβ4 enhanced Nav1.6 INaR generation. By contrast neither overexpression of Navβ2 nor overexpression of a Navβ4-mutant, predicted to be an inactive form of Navβ4, enhanced Nav1.6 INaR generation. DRG neurons transfected with wild-type Navβ4 exhibited increased excitability with increases in both spontaneous activity and evoked activity. Thus, Navβ4 overexpression enhanced INaR and excitability, whereas knockdown or expression of mutant Navβ4 decreased INaR generation. CONCLUSION INaRs are associated with inherited and acquired pain disorders. However, our ability to selectively target and study this current has been hindered due to limited understanding of how it is generated in sensory neurons. This study identified Navβ4 as an important regulator of INaR and excitability in sensory neurons. As such, Navβ4 is a potential target for the manipulation of pain sensations.
Collapse
Affiliation(s)
- Cindy Barbosa
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, USA. .,Department of Pharmacology and Toxicology, Stark Neurosciences Research Institute, 320 West 25th Street, NB-414F, Indianapolis, IN, 46202-2266, USA.
| | - Zhi-Yong Tan
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, USA. .,Department of Pharmacology and Toxicology, Stark Neurosciences Research Institute, 320 West 25th Street, NB-414F, Indianapolis, IN, 46202-2266, USA.
| | - Ruizhong Wang
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, USA.
| | - Wenrui Xie
- Department of Anesthesiology, University of Cincinnati, Cincinnati, OH, USA.
| | - Judith A Strong
- Department of Anesthesiology, University of Cincinnati, Cincinnati, OH, USA.
| | - Reesha R Patel
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, USA. .,Department of Pharmacology and Toxicology, Stark Neurosciences Research Institute, 320 West 25th Street, NB-414F, Indianapolis, IN, 46202-2266, USA.
| | - Michael R Vasko
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, USA. .,Department of Pharmacology and Toxicology, Stark Neurosciences Research Institute, 320 West 25th Street, NB-414F, Indianapolis, IN, 46202-2266, USA.
| | - Jun-Ming Zhang
- Department of Anesthesiology, University of Cincinnati, Cincinnati, OH, USA.
| | - Theodore R Cummins
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, USA. .,Department of Pharmacology and Toxicology, Stark Neurosciences Research Institute, 320 West 25th Street, NB-414F, Indianapolis, IN, 46202-2266, USA.
| |
Collapse
|
46
|
Li XY, Toyoda H. Role of leak potassium channels in pain signaling. Brain Res Bull 2015; 119:73-9. [PMID: 26321392 DOI: 10.1016/j.brainresbull.2015.08.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 08/04/2015] [Accepted: 08/24/2015] [Indexed: 01/05/2023]
Abstract
Potassium (K(+)) channels are membrane proteins that allow rapid and selective flow of K(+) ions across the cell membrane, generating electrical signals in neurons. Thus, K(+) channels play a critical role in determining the neuronal excitability. Two-pore domain (K2P) "leak" K(+) channels give rise to leak K(+) currents that are responsible for the resting membrane potential and input resistance. The wide expression of leak K(+) channels in the central and peripheral nervous system suggests that these channels are critically involved in pain signaling and behavior. Indeed, it has become apparent in the past decade that the leak K(+) channels play essential roles in the development of pain. In this review, we describe evidence for the roles of TASK1, TASK3, TREK1, TREK2, TRAAK and TRESK channels in pain signaling and behavior. Furthermore, we describe the possible involvement of TASK2 and TWIK1 channels in pain.
Collapse
Affiliation(s)
- Xiang-Yao Li
- Institute of Neuroscience, School of Medicine, Zhejiang University, Zhejiang, China
| | - Hiroki Toyoda
- Department of Neuroscience and Oral Physiology, Osaka University Graduate School of Dentistry, 1-8, Yamadaoka, Suita, Japan.
| |
Collapse
|
47
|
Stemkowski PL, Noh MC, Chen Y, Smith PA. Increased excitability of medium-sized dorsal root ganglion neurons by prolonged interleukin-1β exposure is K(+) channel dependent and reversible. J Physiol 2015; 593:3739-55. [PMID: 26110238 PMCID: PMC4560594 DOI: 10.1113/jp270905] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/17/2015] [Indexed: 01/27/2023] Open
Abstract
KEY POINTS Neuropathic pain resulting from peripheral nerve injury is initiated and maintained by persistent ectopic activity in primary afferent neurons. Sciatic nerve injury increases the excitability of medium-sized dorsal root ganglion (DRG) neurons. Levels of the inflammatory cytokine interleukin 1β (IL-1β) increase and peak after 7 days. Five to six days of exposure of medium sized DRG neurons to 100 pm IL-1β promotes persistent increases in excitability which abate within 3-4 days of cytokine removal. This is associated with a profound attenuation of K(+) channel currents but only modest increases in function of cyclic nucleotide-sensitive hyperpolarization-activated channels (HCNs) and of voltage-gated Na(+) and Ca(2+) channel currents. It is unlikely, therefore, that direct interaction of IL-1β with DRG neurons is capable of initiating an enduring phenotypic shift in their electrophysiological properties that follows sciatic nerve injury. The findings also underline the importance of K(+) channel modulation in the actions of inflammatory mediators on peripheral neurons. ABSTRACT Chronic constriction injury of rat sciatic nerve promotes signs of neuropathic pain. This is associated with an increase in the level of interleukin 1β (IL-1β) in primary afferents that peaks at 7 days. This initial cytokine exposure has been proposed to trigger an enduring alteration in neuronal phenotype that underlies chronic hyper-excitability in sensory nerves, which initiates and maintains chronic neuropathic pain. We have shown previously that 5-6 days of exposure of rat dorsal root ganglia (DRGs) to 100 pm IL-1β increases the excitability of medium-sized neurons. We have now found using whole-cell recording that this increased excitability reverts to control levels within 3-4 days of cytokine removal. The effects of IL-1β were dominated by changes in K(+) currents. Thus, the amplitudes of A-current, delayed rectifier and Ca(2+) -sensitive K(+) currents were reduced by ∼68%, ∼64% and ∼36%, respectively. Effects of IL-1β on other cation currents were modest by comparison. There was thus a slight decrease in availability of high voltage-activated Ca(2+) channel current, a small increase in rates of activation of hyperpolarization-activated cyclic nucleotide-gated channel current (IH ), and a shift in the voltage dependence of activation of tetrodotoxin-sensitive sodium current (TTX-S INa ) to more negative potentials. It is unlikely, therefore, that direct interaction of IL-1β with DRG neurons initiates an enduring phenotypic shift in their electrophysiological properties following sciatic nerve injury. Persistent increases in primary afferent excitability following nerve injury may instead depend on altered K(+) channel function and on the continued presence of slightly elevated levels IL-1β and other cytokines.
Collapse
Affiliation(s)
- Patrick L Stemkowski
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada, T2N 4N1
| | - Myung-Chul Noh
- Centre for Neuroscience and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| | - Yishen Chen
- Centre for Neuroscience and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| | - Peter A Smith
- Centre for Neuroscience and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| |
Collapse
|
48
|
Structure and function of μ-conotoxins, peptide-based sodium channel blockers with analgesic activity. Future Med Chem 2015; 6:1677-98. [PMID: 25406007 DOI: 10.4155/fmc.14.107] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
μ-Conotoxins block voltage-gated sodium channels (VGSCs) and compete with tetrodotoxin for binding to the sodium conductance pore. Early efforts identified µ-conotoxins that preferentially blocked the skeletal muscle subtype (NaV1.4). However, the last decade witnessed a significant increase in the number of µ-conotoxins and the range of VGSC subtypes inhibited (NaV1.2, NaV1.3 or NaV1.7). Twenty µ-conotoxin sequences have been identified to date and structure-activity relationship studies of several of these identified key residues responsible for interactions with VGSC subtypes. Efforts to engineer-in subtype specificity are driven by in vivo analgesic and neuromuscular blocking activities. This review summarizes structural and pharmacological studies of µ-conotoxins, which show promise for development of selective blockers of NaV1.2, and perhaps also NaV1.1,1.3 or 1.7.
Collapse
|
49
|
Nazemi S, Manaheji H, Noorbakhsh SM, Zaringhalam J, Sadeghi M, Mohammad-Zadeh M, Haghparast A. Inhibition of microglial activity alters spinal wide dynamic range neuron discharge and reduces microglial Toll-like receptor 4 expression in neuropathic rats. Clin Exp Pharmacol Physiol 2015; 42:772-9. [DOI: 10.1111/1440-1681.12414] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 04/15/2015] [Accepted: 04/18/2015] [Indexed: 11/28/2022]
Affiliation(s)
- Samad Nazemi
- Department of Physiology; Sabzevar University of Medical Sciences; Sabzevar Iran
- Department of Neurophysiology; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Homa Manaheji
- Department of Neurophysiology; Shahid Beheshti University of Medical Sciences; Tehran Iran
- Neuroscience Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | | | - Jalal Zaringhalam
- Department of Neurophysiology; Shahid Beheshti University of Medical Sciences; Tehran Iran
- Neuroscience Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Mehdi Sadeghi
- Department of Physiology; Bushehr University of Medical Sciences; Bushehr Iran
| | | | - Abbas Haghparast
- Neuroscience Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
| |
Collapse
|
50
|
Wang ZJ, Tabakoff B, Levinson SR, Heinbockel T. Inhibition of Nav1.7 channels by methyl eugenol as a mechanism underlying its antinociceptive and anesthetic actions. Acta Pharmacol Sin 2015; 36:791-9. [PMID: 26051112 DOI: 10.1038/aps.2015.26] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 02/05/2015] [Indexed: 11/09/2022] Open
Abstract
AIM Methyl eugenol is a major active component extracted from the Chinese herb Asari Radix et Rhizoma, which has been used to treat toothache and other pain. Previous in vivo studies have shown that methyl eugenol has anesthetic and antinociceptive effects. The aim of this study was to determine the possible mechanism underlying its effect on nervous system disorders. METHODS The direct interaction of methyl eugenol with Na(+) channels was explored and characterized using electrophysiological recordings from Nav1.7-transfected CHO cells. RESULTS In whole-cell patch clamp mode, methyl eugenol tonically inhibited peripheral nerve Nav1.7 currents in a concentration- and voltage-dependent manner, with an IC50 of 295 μmol/L at a -100 mV holding potential. Functionally, methyl eugenol preferentially bound to Nav1.7 channels in the inactivated and/or open state, with weaker binding to channels in the resting state. Thus, in the presence of methyl eugenol, Nav1.7 channels exhibited reduced availability for activation in a steady-state inactivation protocol, strong use-dependent inhibition, enhanced binding kinetics, and slow recovery from inactivation compared to untreated channels. An estimation of the affinity of methyl eugenol for the resting and inactivated states of the channel also demonstrated that methyl eugenol preferentially binds to inactivated channels, with a 6.4 times greater affinity compared to channels in the resting state. The failure of inactivated channels to completely recover to control levels at higher concentrations of methyl eugenol implies that the drug may drive more drug-bound, fast-inactivated channels into drug-bound, slow-inactivated channels. CONCLUSION Methyl eugenol is a potential candidate as an effective local anesthetic and analgesic. The antinociceptive and anesthetic effects of methyl eugenol result from the inhibitory action of methyl eugenol on peripheral Na(+) channels.
Collapse
|