1
|
The Effect of Ginger and Its Sub-Components on Pain. PLANTS 2022; 11:plants11172296. [PMID: 36079679 PMCID: PMC9460519 DOI: 10.3390/plants11172296] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/27/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022]
Abstract
Zingiber officinale Roscoe (ginger) has long been used as an herbal medicine to treat various diseases, and its main sub-components, [6]-gingerol and [6]-shogaol, were also reported to have anti-inflammatory, anti-oxidant, and anti-tumor effects. However, their effects on various types of pain and their underlying mechanisms of action have not been clearly analyzed and understood yet. Thus, in this review, by analyzing 16 studies that used Z. officinale, [6]-gingerol, and [6]-shogaol on mechanical, spontaneous and thermal pain, their effects and mechanisms of action have been analyzed. Pain was induced by either nerve injury or chemical injections in rodents. Nine studies analyzed the analgesic effect of Z. officinale, and four and three studies focused on [6]-gingerol and [6]-shogaol, respectively. Seven papers have demonstrated the underlying mechanism of action of their analgesic effects. Studies have focused on the spinal cord and one on the dorsal root ganglion (DRG) neurons. Involvement and change in the function of serotonergic receptors (5-HT1A, B, D, and 5A), transient receptor potential vanilloid 1 (TRPV1), N-methyl-D-aspartate (NMDA) receptors, phosphorylated extracellular signal-regulated kinase 1/2 (pERK1/2), histone deacetylase 1 (HDAC1), voltage-gated sodium channel 1.8 (Nav1.8), substance P (SP), and sciatic nerve’s morphology have been observed.
Collapse
|
2
|
Fouda MA, Ghovanloo MR, Ruben PC. Late sodium current: incomplete inactivation triggers seizures, myotonias, arrhythmias, and pain syndromes. J Physiol 2022; 600:2835-2851. [PMID: 35436004 DOI: 10.1113/jp282768] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/12/2022] [Indexed: 11/08/2022] Open
Abstract
Acquired and inherited dysfunction in voltage-gated sodium channels underlies a wide range of diseases. "In addition to the defects in trafficking and expression, sodium channelopathies are also caused by dysfunction in one or several gating properties, for instance activation or inactivation. Disruption of the channel inactivation leads to the increased late sodium current, which is a common defect in seizure disorders, cardiac arrhythmias skeletal muscle myotonia and pain. An increase in late sodium current leads to repetitive action potential in neurons and skeletal muscles, and prolonged action potential duration in the heart. In this topical review, we compare the effects of late sodium current in brain, heart, skeletal muscle, and peripheral nerves. Abstract figure legend Shows cartoon illustration of general Nav channel transitions between (1) resting, (2) open, and (3) fast inactivated states. Disruption of the inactivation process exacerbates (4) late sodium currents. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Mohamed A Fouda
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada.,Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt
| | | | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| |
Collapse
|
3
|
Ray N, Buchheit T. Improving Pain and Outcomes in the Perioperative Setting. Perioper Med (Lond) 2022. [DOI: 10.1016/b978-0-323-56724-4.00041-1] [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
|
4
|
Adams GL, Pall PS, Grauer SM, Zhou X, Ballard JE, Vavrek M, Kraus RL, Morissette P, Li N, Colarusso S, Bianchi E, Palani A, Klein R, John CT, Wang D, Tudor M, Nolting AF, Biba M, Nowak T, Makarov AA, Reibarkh M, Buevich AV, Zhong W, Regalado EL, Wang X, Gao Q, Shahripour A, Zhu Y, de Simone D, Frattarelli T, Pasquini NM, Magotti P, Iaccarino R, Li Y, Solly K, Lee KJ, Wang W, Chen F, Zeng H, Wang J, Regan H, Amin RP, Regan CP, Burgey CS, Henze DA, Sun C, Tellers DM. Development of ProTx-II Analogues as Highly Selective Peptide Blockers of Na v1.7 for the Treatment of Pain. J Med Chem 2021; 65:485-496. [PMID: 34931831 DOI: 10.1021/acs.jmedchem.1c01570] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Inhibitor cystine knot peptides, derived from venom, have evolved to block ion channel function but are often toxic when dosed at pharmacologically relevant levels in vivo. The article describes the design of analogues of ProTx-II that safely display systemic in vivo blocking of Nav1.7, resulting in a latency of response to thermal stimuli in rodents. The new designs achieve a better in vivo profile by improving ion channel selectivity and limiting the ability of the peptides to cause mast cell degranulation. The design rationale, structural modeling, in vitro profiles, and rat tail flick outcomes are disclosed and discussed.
Collapse
Affiliation(s)
- Gregory L Adams
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Parul S Pall
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Steven M Grauer
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Xiaoping Zhou
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | - Marissa Vavrek
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Richard L Kraus
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | - Nianyu Li
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Stefania Colarusso
- Peptides and Small Molecules R&D Department, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia (RM), Italy
| | - Elisabetta Bianchi
- Peptides and Small Molecules R&D Department, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia (RM), Italy
| | - Anandan Palani
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Rebecca Klein
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | - Deping Wang
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Matthew Tudor
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Andrew F Nolting
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Mirlinda Biba
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Timothy Nowak
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | | | | | | | - Wendy Zhong
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - Xiao Wang
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Qi Gao
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - Yuping Zhu
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Daniele de Simone
- Peptides and Small Molecules R&D Department, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia (RM), Italy
| | - Tommaso Frattarelli
- Peptides and Small Molecules R&D Department, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia (RM), Italy
| | - Nicolo' Maria Pasquini
- Peptides and Small Molecules R&D Department, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia (RM), Italy
| | - Paola Magotti
- Peptides and Small Molecules R&D Department, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia (RM), Italy
| | - Roberto Iaccarino
- Peptides and Small Molecules R&D Department, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia (RM), Italy
| | - Yuxing Li
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Kelli Solly
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Keun-Joong Lee
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Weixun Wang
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Feifei Chen
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Haoyu Zeng
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Jixin Wang
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Hilary Regan
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Rupesh P Amin
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | | | - Darrell A Henze
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Chengzao Sun
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - David M Tellers
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|
5
|
Han X, Shao J, Ren X, Li Y, Yu W, Lin C, Li L, Sun Y, Xu B, Luo H, Zhu C, Cao J, Li Z. The different mechanisms of peripheral and central TLR4 on chronic postsurgical pain in rats. J Anat 2021; 239:111-124. [PMID: 33730389 PMCID: PMC8197940 DOI: 10.1111/joa.13406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/30/2022] Open
Abstract
Chronic postsurgical pain (CPSP) is a common complication after surgery; however, the underlying mechanisms of CPSP are poorly understood. As one of the most important inflammatory pathways, the Toll-like receptor 4/nuclear factor-kappa B (TLR4/NF-κB) signaling pathway plays an important role in chronic pain. However, the precise role of the TLR4/NF-κB signaling pathway in CPSP remains unclear. In the present study, we established a rat model of CPSP induced by skin/muscle incision and retraction (SMIR) and verified the effects and mechanisms of central and peripheral TLR4 and NF-κB on hyperalgesia in SMIR rats. The results showed that TLR4 expression was increased in both the spinal dorsal horn and dorsal root ganglia (DRGs) of SMIR rats. However, the TLR4 expression pattern in the spinal cord was different from that in DRGs. In the spinal cord, TLR4 was expressed in both neurons and microglia, whereas it was expressed in neurons but not in satellite glial cells in DRGs. Further results demonstrate that the central and peripheral TLR4/NF-κB signaling pathway is involved in the SMIR-induced CPSP by different mechanisms. In the peripheral nervous system, we revealed that the TLR4/NF-κB signaling pathway induced upregulation of voltage-gated sodium channel 1.7 (Nav1.7) in DRGs, triggering peripheral hyperalgesia in SMIR-induced CPSP. In the central nervous system, the TLR4/NF-κB signaling pathway participated in SMIR-induced CPSP by activating microglia in the spinal cord. Ultimately, our findings demonstrated that activation of the peripheral and central TLR4/NF-κB signaling pathway involved in the development of SMIR-induced CPSP.
Collapse
Affiliation(s)
- Xuemin Han
- The Second Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Children’s Hospital of Soochow UniversitySoochowChina
| | - Jinping Shao
- Department of Human AnatomySchool of Basic MedicineZhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| | - Xiuhua Ren
- Department of Human AnatomySchool of Basic MedicineZhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| | - Yaru Li
- The Second Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Wenli Yu
- Department of Human AnatomySchool of Basic MedicineZhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| | - Caihong Lin
- Department of Human AnatomySchool of Basic MedicineZhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| | - Lei Li
- Department of Human AnatomySchool of Basic MedicineZhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| | - Yanyan Sun
- Department of Human AnatomySchool of Basic MedicineZhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| | - Bo Xu
- Department of AnesthesiologyGeneral Hospital of Southern Theatre Command of PLAGuangzhouChina
| | - Huan Luo
- Klinik für AugenheilkundeCharité–Universitätsmedizin Berlin, corporate member of Freie Universität BerlinHumboldt‐Universität zu Berlin, and Berlin Institute of HealthGermany
| | - Changlian Zhu
- Center for Brain Repair and RehabilitationInstitute of Neuroscience and PhysiologyGothenburg UniversityGothenburgSweden
| | - Jing Cao
- Department of Human AnatomySchool of Basic MedicineZhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| | - Zhisong Li
- The Second Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
6
|
A Narrative Review on Perioperative Pain Management Strategies in Enhanced Recovery Pathways-The Past, Present and Future. J Clin Med 2021; 10:jcm10122568. [PMID: 34200695 PMCID: PMC8229260 DOI: 10.3390/jcm10122568] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/05/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022] Open
Abstract
Effective pain management is a key component in the continuum of perioperative care to ensure optimal outcomes for surgical patients. The overutilization of opioids in the past few decades for postoperative pain control has been a major contributor to the current opioid epidemic. Multimodal analgesia (MMA) and enhanced recovery after surgery (ERAS) pathways have been repeatedly shown to significantly improve postoperative outcomes such as pain, function and satisfaction. The current review aims to examine the history of perioperative MMA strategies in ERAS and provide an update with recent evidence. Furthermore, this review details recent advancements in personalized pain medicine. We speculate that the next important step for improving perioperative pain management could be through incorporating these personalized metrics, such as clinical pharmacogenomic testing and patient-reported outcome measurements, into ERAS program.
Collapse
|
7
|
McArthur JR, Munasinghe NR, Finol-Urdaneta RK, Adams DJ, Christie MJ. Spider Venom Peptide Pn3a Inhibition of Primary Afferent High Voltage-Activated Calcium Channels. Front Pharmacol 2021; 11:633679. [PMID: 33584315 PMCID: PMC7875911 DOI: 10.3389/fphar.2020.633679] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/30/2020] [Indexed: 11/19/2022] Open
Abstract
Despite potently inhibiting the nociceptive voltage-gated sodium (Nav) channel, Nav1.7, µ-theraphotoxin Pn3a is antinociceptive only upon co-administration with sub-therapeutic opioid agonists, or by itself at doses >3,000-fold greater than its Nav1.7 IC50 by a yet undefined mechanism. Nav channels are structurally related to voltage-gated calcium (Cav) channels, Cav1 and Cav2. These channels mediate the high voltage-activated (HVA) calcium currents (ICa) that orchestrate synaptic transmission in nociceptive dorsal root ganglion (DRG) neurons and are fine-tuned by opioid receptor (OR) activity. Using whole-cell patch clamp recording, we found that Pn3a (10 µM) inhibits ∼55% of rat DRG neuron HVA-ICa and 60–80% of Cav1.2, Cav1.3, Cav2.1, and Cav2.2 mediated currents in HEK293 cells, with no inhibition of Cav2.3. As a major DRG ICa component, Cav2.2 inhibition by Pn3a (IC50 = 3.71 ± 0.21 µM) arises from an 18 mV hyperpolarizing shift in the voltage dependence of inactivation. We observed that co-application of Pn3a and µ-OR agonist DAMGO results in enhanced HVA-ICa inhibition in DRG neurons whereas co-application of Pn3a with the OR antagonist naloxone does not, underscoring HVA channels as shared targets of Pn3a and opioids. We provide evidence that Pn3a inhibits native and recombinant HVA Cavs at previously reportedly antinociceptive concentrations in animal pain models. We show additive modulation of DRG HVA-ICa by sequential application of low Pn3a doses and sub-therapeutic opioids ligands. We propose Pn3a's antinociceptive effects result, at least in part, from direct inhibition of HVA-ICa at high Pn3a doses, or through additive inhibition by low Pn3a and mild OR activation.
Collapse
Affiliation(s)
- Jeffrey R McArthur
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| | - Nehan R Munasinghe
- Discipline of Pharmacology, University of Sydney, Sydney, NSW, Australia
| | - Rocio K Finol-Urdaneta
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia.,Electrophysiology Facility for Cell Phenotyping and Drug Discovery, IHMRI, Wollongong, NSW, Australia
| | - David J Adams
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| | | |
Collapse
|
8
|
Jiang Y, Idikuda V, Chowdhury S, Chanda B. Activation of the archaeal ion channel MthK is exquisitely regulated by temperature. eLife 2020; 9:e59055. [PMID: 33274718 PMCID: PMC7717905 DOI: 10.7554/elife.59055] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 11/17/2020] [Indexed: 11/13/2022] Open
Abstract
Physiological response to thermal stimuli in mammals is mediated by a structurally diverse class of ion channels, many of which exhibit polymodal behavior. To probe the diversity of biophysical mechanisms of temperature-sensitivity, we characterized the temperature-dependent activation of MthK, a two transmembrane calcium-activated potassium channel from thermophilic archaebacteria. Our functional complementation studies show that these channels are more efficient at rescuing K+ transport at 37°C than at 24°C. Electrophysiological activity of the purified MthK is extremely sensitive (Q10 >100) to heating particularly at low-calcium concentrations whereas channels lacking the calcium-sensing RCK domain are practically insensitive. By analyzing single-channel activities at limiting calcium concentrations, we find that temperature alters the coupling between the cytoplasmic RCK domains and the pore domain. These findings reveal a hitherto unexplored mechanism of temperature-dependent regulation of ion channel gating and shed light on ancient origins of temperature-sensitivity.
Collapse
Affiliation(s)
- Yihao Jiang
- Department of Anesthesiology, Washington University School of MedicineSt. LouisUnited States
- Center for the Investigation of Membrane Excitability Diseases (CIMED)St. LouisUnited States
| | - Vinay Idikuda
- Department of Anesthesiology, Washington University School of MedicineSt. LouisUnited States
- Center for the Investigation of Membrane Excitability Diseases (CIMED)St. LouisUnited States
| | - Sandipan Chowdhury
- Department of Anesthesiology, Washington University School of MedicineSt. LouisUnited States
- Center for the Investigation of Membrane Excitability Diseases (CIMED)St. LouisUnited States
| | - Baron Chanda
- Department of Anesthesiology, Washington University School of MedicineSt. LouisUnited States
- Center for the Investigation of Membrane Excitability Diseases (CIMED)St. LouisUnited States
| |
Collapse
|
9
|
Newly Discovered Action of HpTx3 from Venom of Heteropoda venatoria on Na v1.7 and Its Pharmacological Implications in Analgesia. Toxins (Basel) 2019; 11:toxins11120680. [PMID: 31757020 PMCID: PMC6950750 DOI: 10.3390/toxins11120680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023] Open
Abstract
It has been reported that Heteropodatoxin3 (HpTx3), a peptidic neurotoxin purified from the venom of the spider species Heteropoda venatoria, could inhibit Kv4.2 channels. Our present study newly found that HpTx3 also has potent and selective inhibitory action on Nav1.7, with an IC50 of 135.61 ± 12.98 nM. Without effect on the current–voltage (I-V) relationship of Nav1.7, HpTx3 made minor alternation in the voltage-dependence of activation and steady-state inactivation of Nav1.7 (4.15 mV and 7.29 mV, respectively) by interacting with the extracellular S3–S4 loop (S3b–S4 sequence) in domain II and the domain IV of the Nav channel subtype, showing the characteristics of both pore blocker and gate modifier toxin. During the interaction of HpTx3 with the S3b–S4 sequence of Nav1.7, the amino acid residue D in the sequence played a key role. When administered intraperitoneally or intramuscularly, HpTx3 displayed potent analgesic activity in a dose-dependent manner in different mouse pain models induced by formalin, acetic acid, complete Freund’s adjuvant, hot plate, or spared nerve injury, demonstrating that acute, inflammatory, and neuropathic pains were all effectively inhibited by the toxin. In most cases HpTx3 at doses of ≥ 1mg/kg could produce the analgesic effect comparable to that of 1 mg/kg morphine. These results suggest that HpTx3 not only can be used as a molecular probe to investigate ion channel function and pain mechanism, but also has potential in the development of the drugs that treat the Nav1.7 channel-related pain.
Collapse
|
10
|
François-Moutal L, Dustrude ET, Wang Y, Brustovetsky T, Dorame A, Ju W, Moutal A, Perez-Miller S, Brustovetsky N, Gokhale V, Khanna M, Khanna R. Inhibition of the Ubc9 E2 SUMO-conjugating enzyme-CRMP2 interaction decreases NaV1.7 currents and reverses experimental neuropathic pain. Pain 2018; 159:2115-2127. [PMID: 29847471 PMCID: PMC6150792 DOI: 10.1097/j.pain.0000000000001294] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We previously reported that destruction of the small ubiquitin-like modifier (SUMO) modification site in the axonal collapsin response mediator protein 2 (CRMP2) was sufficient to selectively decrease trafficking of the voltage-gated sodium channel NaV1.7 and reverse neuropathic pain. Here, we further interrogate the biophysical nature of the interaction between CRMP2 and the SUMOylation machinery, and test the hypothesis that a rationally designed CRMP2 SUMOylation motif (CSM) peptide can interrupt E2 SUMO-conjugating enzyme Ubc9-dependent modification of CRMP2 leading to a similar suppression of NaV1.7 currents. Microscale thermophoresis and amplified luminescent proximity homogeneous alpha assay revealed a low micromolar binding affinity between CRMP2 and Ubc9. A heptamer peptide harboring CRMP2's SUMO motif, also bound with similar affinity to Ubc9, disrupted the CRMP2-Ubc9 interaction in a concentration-dependent manner. Importantly, incubation of a tat-conjugated cell-penetrating peptide (t-CSM) decreased sodium currents, predominantly NaV1.7, in a model neuronal cell line. Dialysis of t-CSM peptide reduced CRMP2 SUMOylation and blocked surface trafficking of NaV1.7 in rat sensory neurons. Fluorescence dye-based imaging in rat sensory neurons demonstrated inhibition of sodium influx in the presence of t-CSM peptide; by contrast, calcium influx was unaffected. Finally, t-CSM effectively reversed persistent mechanical and thermal hypersensitivity induced by a spinal nerve injury, a model of neuropathic pain. Structural modeling has now identified a pocket-harboring CRMP2's SUMOylation motif that, when targeted through computational screening of ligands/molecules, is expected to identify small molecules that will biochemically and functionally target CRMP2's SUMOylation to reduce NaV1.7 currents and reverse neuropathic pain.
Collapse
Affiliation(s)
- Liberty François-Moutal
- Department of Pharmacology, The University of Arizona Health Sciences, Tucson, Arizona 85724
| | - Erik T. Dustrude
- Department of Pharmacology, The University of Arizona Health Sciences, Tucson, Arizona 85724
| | - Yue Wang
- Department of Pharmacology, The University of Arizona Health Sciences, Tucson, Arizona 85724
| | - Tatiana Brustovetsky
- Department of Pharmacology and Toxicology, and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Angie Dorame
- Department of Pharmacology, The University of Arizona Health Sciences, Tucson, Arizona 85724
| | - Weina Ju
- Department of Neurology, First Hospital of Jilin University, Jilin University, 71 Xinmin Street, Changchun, 130021, Jilin Province, China
- Department of Pharmacology, First Hospital of Jilin University, Jilin University, 71 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Aubin Moutal
- Department of Pharmacology, The University of Arizona Health Sciences, Tucson, Arizona 85724
| | - Samantha Perez-Miller
- Department of Pharmacology, The University of Arizona Health Sciences, Tucson, Arizona 85724
| | - Nickolay Brustovetsky
- Department of Pharmacology and Toxicology, and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Vijay Gokhale
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona Health Sciences, Tucson, Arizona 85724
| | - May Khanna
- Department of Pharmacology, The University of Arizona Health Sciences, Tucson, Arizona 85724
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona 85724
| | - Rajesh Khanna
- Department of Pharmacology, The University of Arizona Health Sciences, Tucson, Arizona 85724
- Neuroscience Graduate Interdisciplinary Program, College of Medicine, The University of Arizona Health Sciences, Tucson, Arizona 85724
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona 85724
| |
Collapse
|
11
|
Chen C, Xu B, Shi X, Zhang M, Zhang Q, Zhang T, Zhao W, Zhang R, Wang Z, Li N, Fang Q. GpTx-1 and [Ala 5 , Phe 6 , Leu 26 , Arg 28 ]GpTx-1, two peptide Na V 1.7 inhibitors: analgesic and tolerance properties at the spinal level. Br J Pharmacol 2018; 175:3911-3927. [PMID: 30076786 DOI: 10.1111/bph.14461] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 07/10/2018] [Accepted: 07/13/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE The voltage-gated sodium channel NaV 1.7 is considered a therapeutic target for pain treatment based on human genetic evidence. GpTx-1 and its potent analogue [Ala5 , Phe6 , Leu26 , Arg28 ]GpTx-1 (GpTx-1-71) were recently characterized as NaV 1.7 inhibitors in vitro. Furthermore, the present work was conducted to investigate the analgesic properties of these two peptides in different pain models after spinal administration. EXPERIMENTAL APPROACH The antinociceptive activities of both GpTx-1 and GpTx-1-71 were investigated in mouse models of acute, visceral, inflammatory and neuropathic pain. Furthermore, the side effects of GpTx-1 and GpTx-1-71 were evaluated in rotarod, antinociceptive tolerance, acute hyperlocomotion and gastrointestinal transit tests. KEY RESULTS The i.t. administration of both GpTx-1 and GpTx-1-71 dose-dependently produced powerful antinociception in the different pain models. This effect was attenuated by the opioid receptor antagonist naloxone, suggesting the involvement of the opioid system. In this study, repeated administration of these two_peptides produced spinal analgesia without a loss of potency over 8 days in mouse models of acute, inflammatory and neuropathic pain. Moreover, spinal administration of GpTx-1 and GpTx-1-71 did not induce significant effects on motor coordination, evoke acute hyperlocomotion or increase gastrointestinal transit time. CONCLUSIONS AND IMPLICATIONS Our data indicate that the NaV 1.7 peptide inhibitors GpTx-1 and GpTx-1-71 produce powerful, nontolerance-forming analgesia in preclinical pain models, which might be dependent on the endogenous opioid system. In addition, at the spinal level, the limited side effects imply that these NaV 1.7 peptide inhibitors could be potentially developed as GpTx-1-based drugs for pain relief.
Collapse
Affiliation(s)
- Chao Chen
- Guangdong Provincial Key Laboratory of Nano-Micro Materials Research, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Biao Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xuerui Shi
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Mengna Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Qinqin Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Ting Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Weidong Zhao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Run Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Zilong Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Ning Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Quan Fang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
12
|
Tham SW, Giles M. Current pain management strategies for patients with erythromelalgia: a critical review. J Pain Res 2018; 11:1689-1698. [PMID: 30214279 PMCID: PMC6121769 DOI: 10.2147/jpr.s154462] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Erythromelalgia (EM) is a rare disorder characterized by erythematous, warm, painful extremities, which is often precipitated by cold conditions. The pathophysiology of EM is incompletely understood. Recent investigations have identified sodium channelopathy as a genetic cause for this pain condition, classified as primary inherited EM. Other subtypes are idiopathic EM and secondary EM. The management of pain in EM is challenging as no single therapy has been found to be effective. There is varying response to pharmacotherapy and significant variability within this clinical population, resulting in a stepwise trial and error approach. Consequently, EM is often associated with poorer health-related quality of life with higher morbidity. There is currently no consensus or guidelines on management of pain in EM. This is a review of the literature on management of pain using pharmacologic, procedural intervention and nonpharmacologic treatment in children and adults with EM.
Collapse
Affiliation(s)
- See Wan Tham
- Seattle Children's Research Institute, Seattle, WA, USA,
- Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, WA, USA,
| | - Marian Giles
- Seattle Children's Research Institute, Seattle, WA, USA,
- Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, WA, USA,
| |
Collapse
|
13
|
Godazgar M, Zhang Q, Chibalina MV, Rorsman P. Biphasic voltage-dependent inactivation of human Na V 1.3, 1.6 and 1.7 Na + channels expressed in rodent insulin-secreting cells. J Physiol 2018; 596:1601-1626. [PMID: 29441586 PMCID: PMC5924821 DOI: 10.1113/jp275587] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/06/2018] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS Na+ current inactivation is biphasic in insulin-secreting cells, proceeding with two voltage dependences that are half-maximal at ∼-100 mV and -60 mV. Inactivation of voltage-gated Na+ (NaV ) channels occurs at ∼30 mV more negative voltages in insulin-secreting Ins1 and primary β-cells than in HEK, CHO or glucagon-secreting αTC1-6 cells. The difference in inactivation between Ins1 and non-β-cells persists in the inside-out patch configuration, discounting an involvement of a diffusible factor. In Ins1 cells and primary β-cells, but not in HEK cells, inactivation of a single NaV subtype is biphasic and follows two voltage dependences separated by 30-40 mV. We propose that NaV channels adopt different inactivation behaviours depending on the local membrane environment. ABSTRACT Pancreatic β-cells are equipped with voltage-gated Na+ channels that undergo biphasic voltage-dependent steady-state inactivation. A small Na+ current component (10-15%) inactivates over physiological membrane potentials and contributes to action potential firing. However, the major Na+ channel component is completely inactivated at -90 to -80 mV and is therefore inactive in the β-cell. It has been proposed that the biphasic inactivation reflects the contribution of different NaV α-subunits. We tested this possibility by expression of TTX-resistant variants of the NaV subunits found in β-cells (NaV 1.3, NaV 1.6 and NaV 1.7) in insulin-secreting Ins1 cells and in non-β-cells (including HEK and CHO cells). We found that all NaV subunits inactivated at 20-30 mV more negative membrane potentials in Ins1 cells than in HEK or CHO cells. The more negative inactivation in Ins1 cells does not involve a diffusible intracellular factor because the difference between Ins1 and CHO persisted after excision of the membrane. NaV 1.7 inactivated at 15--20 mV more negative membrane potentials than NaV 1.3 and NaV 1.6 in Ins1 cells but this small difference is insufficient to solely explain the biphasic inactivation in Ins1 cells. In Ins1 cells, but never in the other cell types, widely different components of NaV inactivation (separated by 30 mV) were also observed following expression of a single type of NaV α-subunit. The more positive component exhibited a voltage dependence of inactivation similar to that found in HEK and CHO cells. We propose that biphasic NaV inactivation in insulin-secreting cells reflects insertion of channels in membrane domains that differ with regard to lipid and/or membrane protein composition.
Collapse
Affiliation(s)
- Mahdieh Godazgar
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of MedicineUniversity of OxfordChurchill HospitalOxfordOX3 7LEUK
| | - Quan Zhang
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of MedicineUniversity of OxfordChurchill HospitalOxfordOX3 7LEUK
| | - Margarita V. Chibalina
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of MedicineUniversity of OxfordChurchill HospitalOxfordOX3 7LEUK
| | - Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of MedicineUniversity of OxfordChurchill HospitalOxfordOX3 7LEUK
- Metabolic PhysiologyDepartment of Neuroscience and PhysiologyMedicinaregatan 11GothenburgS‐413 09Sweden
| |
Collapse
|
14
|
Ferrante M, Tahvildari B, Duque A, Hadzipasic M, Salkoff D, Zagha EW, Hasselmo ME, McCormick DA. Distinct Functional Groups Emerge from the Intrinsic Properties of Molecularly Identified Entorhinal Interneurons and Principal Cells. Cereb Cortex 2018; 27:3186-3207. [PMID: 27269961 DOI: 10.1093/cercor/bhw143] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Inhibitory interneurons are an important source of synaptic inputs that may contribute to network mechanisms for coding of spatial location by entorhinal cortex (EC). The intrinsic properties of inhibitory interneurons in the EC of the mouse are mostly undescribed. Intrinsic properties were recorded from known cell types, such as, stellate and pyramidal cells and 6 classes of molecularly identified interneurons (regulator of calcineurin 2, somatostatin, serotonin receptor 3a, neuropeptide Y neurogliaform (NGF), neuropeptide Y non-NGF, and vasoactive intestinal protein) in acute brain slices. We report a broad physiological diversity between and within cell classes. We also found differences in the ability to produce postinhibitory rebound spikes and in the frequency and amplitude of incoming EPSPs. To understand the source of this intrinsic variability we applied hierarchical cluster analysis to functionally classify neurons. These analyses revealed physiologically derived cell types in EC that mostly corresponded to the lines identified by biomarkers with a few unexpected and important differences. Finally, we reduced the complex multidimensional space of intrinsic properties to the most salient five that predicted the cellular biomolecular identity with 81.4% accuracy. These results provide a framework for the classification of functional subtypes of cortical neurons by their intrinsic membrane properties.
Collapse
Affiliation(s)
- Michele Ferrante
- Center for Memory and Brain.,Center for Systems Neuroscience.,Department of Psychological and Brain Sciences, Boston University, Boston, MA 02215, USA
| | - Babak Tahvildari
- Department of Neurobiology.,Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| | - Alvaro Duque
- Department of Neurobiology.,Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| | - Muhamed Hadzipasic
- Interdepartmental Program in Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| | - David Salkoff
- Department of Neurobiology.,Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| | - Edward William Zagha
- Department of Neurobiology.,Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| | - Michael E Hasselmo
- Center for Memory and Brain.,Center for Systems Neuroscience.,Department of Psychological and Brain Sciences, Boston University, Boston, MA 02215, USA
| | - David A McCormick
- Department of Neurobiology.,Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| |
Collapse
|
15
|
Haskins W, Benitez S, Mercado JM, Acosta CG. Cutaneous inflammation regulates THIK1 expression in small C-like nociceptor dorsal root ganglion neurons. Mol Cell Neurosci 2017; 83:13-26. [DOI: 10.1016/j.mcn.2017.06.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 06/29/2017] [Accepted: 06/30/2017] [Indexed: 10/19/2022] Open
|
16
|
Vetter I, Deuis JR, Mueller A, Israel MR, Starobova H, Zhang A, Rash LD, Mobli M. NaV1.7 as a pain target – From gene to pharmacology. Pharmacol Ther 2017; 172:73-100. [DOI: 10.1016/j.pharmthera.2016.11.015] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
17
|
Estacion M, Waxman SG. Nonlinear effects of hyperpolarizing shifts in activation of mutant Na v1.7 channels on resting membrane potential. J Neurophysiol 2017; 117:1702-1712. [PMID: 28148645 DOI: 10.1152/jn.00898.2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/11/2017] [Accepted: 01/26/2017] [Indexed: 01/16/2023] Open
Abstract
The Nav1.7 sodium channel is preferentially expressed within dorsal root ganglion (DRG) and sympathetic ganglion neurons. Gain-of-function mutations that cause the painful disorder inherited erythromelalgia (IEM) shift channel activation in a hyperpolarizing direction. When expressed within DRG neurons, these mutations produce a depolarization of resting membrane potential (RMP). The biophysical basis for the depolarized RMP has to date not been established. To explore the effect on RMP of the shift in activation associated with a prototypical IEM mutation (L858H), we used dynamic-clamp models that represent graded shifts that fractionate the effect of the mutation on activation voltage dependence. Dynamic-clamp recording from DRG neurons using a before-and-after protocol for each cell made it possible, even in the presence of cell-to-cell variation in starting RMP, to assess the effects of these graded mutant models. Our results demonstrate a nonlinear, progressively larger effect on RMP as the shift in activation voltage dependence becomes more hyperpolarized. The observed differences in RMP were predicted by the "late" current of each mutant model. Since the depolarization of RMP imposed by IEM mutant channels is known, in itself, to produce hyperexcitability of DRG neurons, the development of pharmacological agents that normalize or partially normalize activation voltage dependence of IEM mutant channels merits further study.NEW & NOTEWORTHY Inherited erythromelalgia (IEM), the first human pain disorder linked to a sodium channel, is widely regarded as a genetic model of neuropathic pain. IEM is produced by Nav1.7 mutations that hyperpolarize activation. These mutations produce a depolarization of resting membrane potential (RMP) in dorsal root ganglion neurons. Using dynamic clamp to explore the effect on RMP of the shift in activation, we demonstrate a nonlinear effect on RMP as the shift in activation voltage dependence becomes more hyperpolarized.
Collapse
Affiliation(s)
- Mark Estacion
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and.,Rehabilitation Research Center, Department of Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and .,Rehabilitation Research Center, Department of Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
18
|
Asiedu MN, Han C, Dib-Hajj SD, Waxman SG, Price TJ, Dussor G. The AMPK Activator A769662 Blocks Voltage-Gated Sodium Channels: Discovery of a Novel Pharmacophore with Potential Utility for Analgesic Development. PLoS One 2017; 12:e0169882. [PMID: 28118359 PMCID: PMC5261566 DOI: 10.1371/journal.pone.0169882] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 12/23/2016] [Indexed: 12/12/2022] Open
Abstract
Voltage-gated sodium channels (VGSC) regulate neuronal excitability by governing action potential (AP) generation and propagation. Recent studies have revealed that AMP-activated protein kinase (AMPK) activators decrease sensory neuron excitability, potentially by preventing sodium (Na+) channel phosphorylation by kinases such as ERK or via modulation of translation regulation pathways. The direct positive allosteric modulator A769662 displays substantially greater efficacy than other AMPK activators in decreasing sensory neuron excitability suggesting additional mechanisms of action. Here, we show that A769662 acutely inhibits AP firing stimulated by ramp current injection in rat trigeminal ganglion (TG) neurons. PT1, a structurally dissimilar AMPK activator that reduces nerve growth factor (NGF) -induced hyperexcitability, has no influence on AP firing in TG neurons upon acute application. In voltage-clamp recordings, application of A769662 reduces VGSC current amplitudes. These findings, based on acute A769662 application, suggest a direct channel blocking effect. Indeed, A769662 dose-dependently blocks VGSC in rat TG neurons and in Nav1.7-transfected cells with an IC50 of ~ 10 μM. A769662 neither displayed use-dependent inhibition nor interacted with the local anesthetic (LA) binding site. Popliteal fossa administration of A769662 decreased noxious thermal responses with a peak effect at 5 mins demonstrating an analgesic effect. These data indicate that in addition to AMPK activation, A769662 acts as a direct blocker/modulator of VGSCs, a potential mechanism enhancing the analgesic property of this compound.
Collapse
Affiliation(s)
- Marina N. Asiedu
- University of Arizona, Department of Pharmacology, Tucson, Arizona, United States of America
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Richardson, Texas, United States of America
| | - Chongyang Han
- Yale School of Medicine, Department of Neurology, Center for Neuroscience and Regeneration Research, and Veterans Administration Connecticut Healthcare System, Rehabilitation Research Center, West Haven, Connecticut, United States of America
| | - Sulayman D. Dib-Hajj
- Yale School of Medicine, Department of Neurology, Center for Neuroscience and Regeneration Research, and Veterans Administration Connecticut Healthcare System, Rehabilitation Research Center, West Haven, Connecticut, United States of America
| | - Stephen G. Waxman
- Yale School of Medicine, Department of Neurology, Center for Neuroscience and Regeneration Research, and Veterans Administration Connecticut Healthcare System, Rehabilitation Research Center, West Haven, Connecticut, United States of America
| | - Theodore J. Price
- University of Arizona, Department of Pharmacology, Tucson, Arizona, United States of America
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Richardson, Texas, United States of America
| | - Gregory Dussor
- University of Arizona, Department of Pharmacology, Tucson, Arizona, United States of America
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Richardson, Texas, United States of America
- * E-mail:
| |
Collapse
|
19
|
Hierarchical CRMP2 posttranslational modifications control NaV1.7 function. Proc Natl Acad Sci U S A 2016; 113:E8443-E8452. [PMID: 27940916 DOI: 10.1073/pnas.1610531113] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Voltage-gated sodium channels are crucial determinants of neuronal excitability and signaling. Trafficking of the voltage-gated sodium channel NaV1.7 is dysregulated in neuropathic pain. We identify a trafficking program for NaV1.7 driven by hierarchical interactions with posttranslationally modified versions of the binding partner collapsin response mediator protein 2 (CRMP2). The binding described between CRMP2 and NaV1.7 was enhanced by conjugation of CRMP2 with small ubiquitin-like modifier (SUMO) and further controlled by the phosphorylation status of CRMP2. We determined that CRMP2 SUMOylation is enhanced by prior phosphorylation by cyclin-dependent kinase 5 and antagonized by Fyn phosphorylation. As a consequence of CRMP2 loss of SUMOylation and binding to NaV1.7, the channel displays decreased membrane localization and current density, and reduces neuronal excitability. Preventing CRMP2 SUMOylation with a SUMO-impaired CRMP2-K374A mutant triggered NaV1.7 internalization in a clathrin-dependent manner involving the E3 ubiquitin ligase Nedd4-2 (neural precursor cell expressed developmentally down-regulated protein 4) and endocytosis adaptor proteins Numb and epidermal growth factor receptor pathway substrate 15. Collectively, our work shows that diverse modifications of CRMP2 cross-talk to control NaV1.7 activity and illustrate a general principle for regulation of NaV1.7.
Collapse
|
20
|
Shao J, Cao J, Wang J, Ren X, Su S, Li M, Li Z, Zhao Q, Zang W. MicroRNA-30b regulates expression of the sodium channel Nav1.7 in nerve injury-induced neuropathic pain in the rat. Mol Pain 2016; 12:12/0/1744806916671523. [PMID: 27765894 PMCID: PMC5081156 DOI: 10.1177/1744806916671523] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 08/03/2016] [Indexed: 01/08/2023] Open
Abstract
Voltage-gated sodium channels, which are involved in pain pathways, have emerged as major targets for therapeutic intervention in pain disorders. Nav1.7, the tetrodotoxin-sensitive voltage-gated sodium channel isoform encoded by SCN9A and predominantly expressed in pain-sensing neurons in the dorsal root ganglion, plays a crucial role in nociception. MicroRNAs are highly conserved, small non-coding RNAs. Through binding to the 3′ untranslated region of their target mRNAs, microRNAs induce the cleavage and/or inhibition of protein translation. Based on bioinformatics analysis using TargetScan software, we determined that miR-30b directly targets SCN9A. To investigate the roles of Nav1.7 and miR-30b in neuropathic pain, we examined changes in the expression of Nav1.7 in the dorsal root ganglion by miR-30b over-expression or knockdown in rats with spared nerve injury. Our results demonstrated that the expression of miR-30b and Nav1.7 was down-regulated and up-regulated, respectively, in the dorsal root ganglion of spared nerve injury rats. MiR-30b over-expression in spared nerve injury rats inhibited SCN9A transcription, resulting in pain relief. In addition, miR-30b knockdown significantly increased hypersensitivity to pain in naive rats. We also observed that miR-30b decreased Nav1.7 expression in PC12 cells. Taken together, our results suggest that miR-30b plays an important role in neuropathic pain by regulating Nav1.7 expression. Therefore, miR-30b may be a promising target for the treatment of chronic neuropathic pain.
Collapse
Affiliation(s)
- Jinping Shao
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Jing Cao
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Jiannan Wang
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Xiuhua Ren
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Songxue Su
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Ming Li
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Zhihua Li
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Qingzan Zhao
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Weidong Zang
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
21
|
Theile JW, Fuller MD, Chapman ML. The Selective Nav1.7 Inhibitor, PF-05089771, Interacts Equivalently with Fast and Slow Inactivated Nav1.7 Channels. Mol Pharmacol 2016; 90:540-548. [PMID: 27587537 DOI: 10.1124/mol.116.105437] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/31/2016] [Indexed: 12/18/2022] Open
Abstract
Voltage-gated sodium (Nav) channel inhibitors are used clinically as analgesics and local anesthetics. However, the absence of Nav channel isoform selectivity of current treatment options can result in adverse cardiac and central nervous system side effects, limiting their therapeutic utility. Human hereditary gain- or loss-of-pain disorders have demonstrated an essential role of Nav1.7 sodium channels in the sensation of pain, thus making this channel an attractive target for new pain therapies. We previously identified a novel, state-dependent human Nav1.7 selective inhibitor (PF-05089771, IC50 = 11 nM) that interacts with the voltage-sensor domain (VSD) of domain IV. We further characterized the state-dependent interaction of PF-05089771 by systematically varying the voltage, frequency, and duration of conditioning prepulses to provide access to closed, open, and fast- or slow-inactivated states. The current study demonstrates that PF-05089771 exhibits a slow onset of block that is depolarization and concentration dependent, with a similarly slow recovery from block. Furthermore, the onset of block by PF-05089771 develops with similar rates using protocols that bias channels into predominantly fast- or slow-inactivated states, suggesting that channel inhibition is less dependent on the availability of a particular inactivated state than the relative time that the channel is depolarized. Taken together, the inhibitory profile of PF-05089771 suggests that a conformational change in the domain IV VSD after depolarization is necessary and sufficient to reveal a high-affinity binding site with which PF-05089771 interacts, stabilizing the channel in a nonconducting conformation from which recovery is slow.
Collapse
Affiliation(s)
- Jonathan W Theile
- Neusentis US, Pfizer Global R&D, (currently Icagen, Inc.), Durham, North Carolina
| | - Matthew D Fuller
- Neusentis US, Pfizer Global R&D, (currently Icagen, Inc.), Durham, North Carolina
| | - Mark L Chapman
- Neusentis US, Pfizer Global R&D, (currently Icagen, Inc.), Durham, North Carolina
| |
Collapse
|
22
|
Ikegami D, Igarashi K, Otsuka M, Kuzumaki N, Narita M. [The importance of epigenetic analysis in pain]. Nihon Yakurigaku Zasshi 2016; 147:225-229. [PMID: 27063906 DOI: 10.1254/fpj.147.225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
|
23
|
Abstract
SUMMARY The experience of pain is a subjective one and more than a simple sensation. Pain is commonly defined as an unpleasant sensory and emotional experience due to actual or potential tissue damage or described in such terms. Pain may be broadly classified into physiological and pathological pain. Nociceptive and inflammatory pains are physiological pain states, as they are protective and adaptive, whereas pathological pain is nonprotective and maladaptive. Nociception is the result of suprathreshold stimulation of peripheral nociceptors. Inflammatory pain follows release of various chemical mediators after tissue injury including surgery leading to peripheral sensitization. Nociceptive input is then transmitted to the spinal cord via primary afferents. Modulation of the nociceptive input occurs in the dorsal horn of the spinal cord, influenced by descending inhibitory systems. Central sensitization is a neuromodulatory change that results in the development of secondary hyperalgesia. The modulated nociceptive input then travels up the ascending tracts, mainly via the spinothalamic tract to the thalamus and subsequently to the higher centers of the brain. Pathological pain such as neuropathic pain and central nervous system dysfunctional pain are the result of neuroplasticity of the peripheral and central nervous system. Abnormal ectopic firing of neurons in the absence of a stimulus, increased neuronal hypersensitivity, changes within ion channels, and even alteration in gene expression and changes in the cortical representation are involved in the pathogenesis of these pain states. The development of persistent postsurgical pain is an example for this complex process.
Collapse
|
24
|
Zhang Q, Chibalina MV, Bengtsson M, Groschner LN, Ramracheya R, Rorsman NJG, Leiss V, Nassar MA, Welling A, Gribble FM, Reimann F, Hofmann F, Wood JN, Ashcroft FM, Rorsman P. Na+ current properties in islet α- and β-cells reflect cell-specific Scn3a and Scn9a expression. J Physiol 2014; 592:4677-96. [PMID: 25172946 PMCID: PMC4253470 DOI: 10.1113/jphysiol.2014.274209] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Mouse pancreatic β- and α-cells are equipped with voltage-gated Na+ currents that inactivate over widely different membrane potentials (half-maximal inactivation (V0.5) at −100 mV and −50 mV in β- and α-cells, respectively). Single-cell PCR analyses show that both α- and β-cells have Nav1.3 (Scn3) and Nav1.7 (Scn9a) α subunits, but their relative proportions differ: β-cells principally express Nav1.7 and α-cells Nav1.3. In α-cells, genetically ablating Scn3a reduces the Na+ current by 80%. In β-cells, knockout of Scn9a lowers the Na+ current by >85%, unveiling a small Scn3a-dependent component. Glucagon and insulin secretion are inhibited in Scn3a−/− islets but unaffected in Scn9a-deficient islets. Thus, Nav1.3 is the functionally important Na+ channel α subunit in both α- and β-cells because Nav1.7 is largely inactive at physiological membrane potentials due to its unusually negative voltage dependence of inactivation. Interestingly, the Nav1.7 sequence in brain and islets is identical and yet the V0.5 for inactivation is >30 mV more negative in β-cells. This may indicate the presence of an intracellular factor that modulates the voltage dependence of inactivation.
Collapse
Affiliation(s)
- Quan Zhang
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, UK
| | - Margarita V Chibalina
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, UK
| | - Martin Bengtsson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, UK
| | - Lukas N Groschner
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, UK
| | - Reshma Ramracheya
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, UK
| | - Nils J G Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, UK
| | - Veronika Leiss
- Institut für Pharmakologie und Toxikologie, TU München, Biedersteiner Str. 29, 80802, München, Germany Institut für Pharmakologie und Toxikologie, Eberhard-Karls Universität, Wilhelmstr. 56, 72074, Tübingen, Germany
| | - Mohammed A Nassar
- Molecular Nociception Group, University College London (UCL), Gower Street, London, WC1E 6BT, UK Biomedical Science, University of Sheffield, Sheffield, S10 2TN, UK
| | - Andrea Welling
- Institut für Pharmakologie und Toxikologie, TU München, Biedersteiner Str. 29, 80802, München, Germany
| | - Fiona M Gribble
- Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, UK
| | - Frank Reimann
- Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, UK
| | - Franz Hofmann
- Institut für Pharmakologie und Toxikologie, TU München, Biedersteiner Str. 29, 80802, München, Germany FOR 923, Institut für Pharmakologie und Toxikologie, TU München, Biedersteiner Str. 29, 80802, München, Germany
| | - John N Wood
- Molecular Nociception Group, University College London (UCL), Gower Street, London, WC1E 6BT, UK
| | - Frances M Ashcroft
- Henry Wellcome Centre for Gene Function, Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| | - Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, UK Department of Neuroscience and Physiology, Sahlgren's Academy, University of Göteborg, Box 430, SE40530, Göteborg, Sweden
| |
Collapse
|
25
|
Chowdhury S, Jarecki BW, Chanda B. A molecular framework for temperature-dependent gating of ion channels. Cell 2014; 158:1148-1158. [PMID: 25156949 DOI: 10.1016/j.cell.2014.07.026] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 07/02/2014] [Accepted: 07/18/2014] [Indexed: 12/13/2022]
Abstract
Perception of heat or cold in higher organisms is mediated by specialized ion channels whose gating is exquisitely sensitive to temperature. The physicochemical underpinnings of this temperature-sensitive gating have proven difficult to parse. Here, we took a bottom-up protein design approach and rationally engineered ion channels to activate in response to thermal stimuli. By varying amino acid polarities at sites undergoing state-dependent changes in solvation, we were able to systematically confer temperature sensitivity to a canonical voltage-gated ion channel. Our results imply that the specific heat capacity change during channel gating is a major determinant of thermosensitive gating. We also show that reduction of gating charges amplifies temperature sensitivity of designer channels, which accounts for low-voltage sensitivity in all known temperature-gated ion channels. These emerging principles suggest a plausible molecular mechanism for temperature-dependent gating that reconcile how ion channels with an overall conserved transmembrane architecture may exhibit a wide range of temperature-sensing phenotypes. :
Collapse
Affiliation(s)
- Sandipan Chowdhury
- Graduate Program in Biophysics, 1111 Highland Ave, School of Medicine and Public Health, University of Wisconsin, Madison, Madison, WI 53705, USA; Department of Neuroscience, 1111 Highland Ave, School of Medicine and Public Health, University of Wisconsin, Madison, Madison, WI 53705, USA
| | - Brian W Jarecki
- Department of Neuroscience, 1111 Highland Ave, School of Medicine and Public Health, University of Wisconsin, Madison, Madison, WI 53705, USA
| | - Baron Chanda
- Graduate Program in Biophysics, 1111 Highland Ave, School of Medicine and Public Health, University of Wisconsin, Madison, Madison, WI 53705, USA; Department of Neuroscience, 1111 Highland Ave, School of Medicine and Public Health, University of Wisconsin, Madison, Madison, WI 53705, USA.
| |
Collapse
|
26
|
Langeslag M, Malsch P, Welling A, Kress M. Reduced excitability of gp130-deficient nociceptors is associated with increased voltage-gated potassium currents and Kcna4 channel upregulation. Pflugers Arch 2014; 466:2153-65. [DOI: 10.1007/s00424-014-1443-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 12/18/2013] [Accepted: 01/04/2014] [Indexed: 12/20/2022]
|
27
|
Cheng Q, Zhou Y. Novel role of KT5720 on regulating hyperpolarization-activated cyclic nucleotide-gated channel activity and dorsal root ganglion neuron excitability. DNA Cell Biol 2013; 32:320-8. [PMID: 23713946 DOI: 10.1089/dna.2013.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are expressed in dorsal root ganglion (DRG) neurons, which are involved in diverse mechanisms that regulate DRG functions. Protein kinase A (PKA) is an essential kinase that plays a key role in almost all types of cells; it regulates the ion channel activity, the intracellular Ca(2+) concentration, as well as modulates cellular signals transduction. Nevertheless, the effect of PKA inhibition on the HCN channel activity in DRG neuron remains to be elucidated. Here we investigated the impact of PKA inhibition on the HCN channel activity and DRG neurons excitability. Our patch-clamp experiments both under whole-cell and single-channel conditions demonstrated that PKA inhibition with KT5720, a cell membrane permeable PKA-specific inhibitor, significantly attenuated HCN channel currents. Current clamp recording on freshly isolated DRG neurons showed KT5720 reduced overshoot amplitude and enhanced the threshold of the action potential. Moreover, our live-cell Ca(2+) imaging experiments illustrated KT5720 markedly reduced the intracellular Ca(2+) level. Collectively, this is the first report that addresses KT5720 attenuates the HCN channel activity and intracellular Ca(2+), thus reducing DRG neurons excitability. Therefore, our data strongly suggest that PKA is a potential target for curing HCN and DRG neuron relevant diseases.
Collapse
Affiliation(s)
- Qiuping Cheng
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | | |
Collapse
|
28
|
Ossipov MH. The perception and endogenous modulation of pain. SCIENTIFICA 2012; 2012:561761. [PMID: 24278716 PMCID: PMC3820628 DOI: 10.6064/2012/561761] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 11/19/2012] [Indexed: 06/02/2023]
Abstract
Pain is often perceived an unpleasant experience that includes sensory and emotional/motivational responses. Accordingly, pain serves as a powerful teaching signal enabling an organism to avoid injury, and is critical to survival. However, maladaptive pain, such as neuropathic or idiopathic pain, serves no survival function. Genomic studies of individuals with congenital insensitivity to pain or paroxysmal pain syndromes considerable increased our understanding of the function of peripheral nociceptors, and especially of the roles of voltage-gated sodium channels and of nerve growth factor (NGF)/TrkA receptors in nociceptive transduction and transmission. Brain imaging studies revealed a "pain matrix," consisting of cortical and subcortical regions that respond to noxious inputs and can positively or negatively modulate pain through activation of descending pain modulatory systems. Projections from the periaqueductal grey (PAG) and the rostroventromedial medulla (RVM) to the trigeminal and spinal dorsal horns can inhibit or promote further nociceptive inputs. The "pain matrix" can explain such varied phenomena as stress-induced analgesia, placebo effect and the role of expectation on pain perception. Disruptions in these systems may account for the existence idiopathic pan states such as fibromyalgia. Increased understanding of pain modulatory systems will lead to development of more effective therapeutics for chronic pain.
Collapse
Affiliation(s)
- Michael H. Ossipov
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
29
|
Sodium channel Na(v)1.7 is essential for lowering heat pain threshold after burn injury. J Neurosci 2012; 32:10819-32. [PMID: 22875917 DOI: 10.1523/jneurosci.0304-12.2012] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Marked hypersensitivity to heat and mechanical (pressure) stimuli develop after a burn injury, but the neural mechanisms underlying these effects are poorly understood. In this study, we establish a new mouse model of focal second-degree burn injury to investigate the molecular and cellular basis for burn injury-induced pain. This model features robust injury-induced behavioral effects and tissue-specific altered cytokine profile, but absence of glial activation in spinal dorsal horn. Three voltage-gated sodium channels, Na(v)1.7, Na(v)1.8, and Na(v)1.9, are preferentially expressed in peripheral somatosensory neurons of the dorsal root ganglia (DRGs) and have been implicated in injury-induced neuronal hyperexcitability. Using knock-out mice, we provide evidence that Na(v)1.7 selectively contributes to burn-induced hypersensitivity to heat, but not mechanical, stimuli. After burn model injury, wild-type mice display increased sensitivity to heat stimuli, and a normally non-noxious warm stimulus induces activity-dependent Fos expression in spinal dorsal horn neurons. Strikingly, both effects are absent in Na(v)1.7 conditional knock-out (cKO) mice. Furthermore, burn injury increases density and shifts activation of tetrodotoxin-sensitive currents in a hyperpolarized direction, both pro-excitatory properties, in DRG neurons from wild-type but not Na(v)1.7 cKO mice. We propose that, in sensory neurons damaged by burn injury to the hindpaw, Na(v)1.7 currents contribute to the hyperexcitability of sensory neurons, their communication with postsynaptic spinal pain pathways, and behavioral thresholds to heat stimuli. Our results offer insights into the molecular and cellular mechanisms of modality-specific pain signaling, and suggest Na(v)1.7-blocking drugs may be effective in burn patients.
Collapse
|
30
|
Buchheit T, Van de Ven T, Shaw A. Epigenetics and the transition from acute to chronic pain. PAIN MEDICINE 2012; 13:1474-90. [PMID: 22978429 DOI: 10.1111/j.1526-4637.2012.01488.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE The objective of this study was to review the epigenetic modifications involved in the transition from acute to chronic pain and to identify potential targets for the development of novel, individualized pain therapeutics. BACKGROUND Epigenetics is the study of heritable modifications in gene expression and phenotype that do not require a change in genetic sequence to manifest their effects. Environmental toxins, medications, diet, and psychological stresses can alter epigenetic processes such as DNA methylation, histone acetylation, and RNA interference. As epigenetic modifications potentially play an important role in inflammatory cytokine metabolism, steroid responsiveness, and opioid sensitivity, they are likely key factors in the development of chronic pain. Although our knowledge of the human genetic code and disease-associated polymorphisms has grown significantly in the past decade, we have not yet been able to elucidate the mechanisms that lead to the development of persistent pain after nerve injury or surgery. DESIGN This is a focused literature review of epigenetic science and its relationship to chronic pain. RESULTS Significant laboratory and clinical data support the notion that epigenetic modifications are affected by the environment and lead to differential gene expression. Similar to mechanisms involved in the development of cancer, neurodegenerative disease, and inflammatory disorders, the literature endorses an important potential role for epigenetics in chronic pain. CONCLUSIONS Epigenetic analysis may identify mechanisms critical to the development of chronic pain after injury, and may provide new pathways and target mechanisms for future drug development and individualized medicine.
Collapse
Affiliation(s)
- Thomas Buchheit
- Department of Anesthesiology, Duke University Medical Center, Durham VA Medical Center, Durham, NC 27710, USA.
| | | | | |
Collapse
|
31
|
Farmer C, Cox JJ, Fletcher EV, Woods CG, Wood JN, Schorge S. Splice variants of Na(V)1.7 sodium channels have distinct β subunit-dependent biophysical properties. PLoS One 2012; 7:e41750. [PMID: 22911851 PMCID: PMC3404004 DOI: 10.1371/journal.pone.0041750] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 06/25/2012] [Indexed: 12/13/2022] Open
Abstract
Genes encoding the α subunits of neuronal sodium channels have evolutionarily conserved sites of alternative splicing but no functional differences have been attributed to the splice variants. Here, using NaV1.7 as an exemplar, we show that the sodium channel isoforms are functionally distinct when co-expressed with β subunits. The gene, SCN9A, encodes the α subunit of the NaV1.7 channel, and contains both sites of alternative splicing that are highly conserved. In conditions where the intrinsic properties of the NaV1.7 splice variants were similar when expressed alone, co-expression of β1 subunits had different effects on channel availability that were determined by splicing at either site in the α subunit. While the identity of exon 5 determined the degree to which β1 subunits altered voltage-dependence of activation (P = 0.027), the length of exon 11 regulated how far β1 subunits depolarised voltage-dependence of inactivation (P = 0.00012). The results could have a significant impact on channel availability, for example with the long version of exon 11, the co-expression of β1 subunits could lead to nearly twice as large an increase in channel availability compared to channels containing the short version. Our data suggest that splicing can change the way that NaV channels interact with β subunits. Because splicing is conserved, its unexpected role in regulating the functional impact of β subunits may apply to multiple voltage-gated sodium channels, and the full repertoire of β subunit function may depend on splicing in α subunits.
Collapse
Affiliation(s)
- Clare Farmer
- UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - James J. Cox
- UCL Wolfson Institute of Biomedical Research, London, United Kingdom
| | - E. V. Fletcher
- UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - C. Geoffrey Woods
- Department of Medical Genetics, Cambridge Institute for Medical Research, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - John N. Wood
- UCL Wolfson Institute of Biomedical Research, London, United Kingdom
| | - Stephanie Schorge
- UCL Institute of Neurology, Queen Square, London, United Kingdom
- * E-mail:
| |
Collapse
|
32
|
Wang GK, Strichartz GR. State-Dependent Inhibition of Sodium Channels by Local Anesthetics: A 40-Year Evolution. BIOCHEMISTRY MOSCOW SUPPLEMENT SERIES A-MEMBRANE AND CELL BIOLOGY 2012; 6:120-127. [PMID: 23710324 DOI: 10.1134/s1990747812010151] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Knowledge about the mechanism of impulse blockade by local anesthetics has evolved over the past four decades, from the realization that Na+ channels were inhibited to affect the impulse blockade to an identification of the amino acid residues within the Na+ channel that bind the local anesthetic molecule. Within this period appreciation has grown of the state-dependent nature of channel inhibition, with rapid binding and unbinding at relatively high affinity to the open state, and weaker binding to the closed resting state. Slow binding of high affinity for the inactivated state accounts for the salutary therapeutic as well as the toxic actions of diverse class I anti-arrhythmic agents, but may have little importance for impulse blockade, which requires concentrations high enough to block the resting state. At the molecular level, residues on the S6 transmembrane segments in three of the homologous domains of the channel appear to contribute to the binding of local anesthetics, with some contribution also from parts of the selectivity filter. Binding to the inactivated state, and perhaps the open state, involves some residues that are not identical to those that bind these drugs in the resting state, suggesting spatial flexibility in the "binding site". Questions remaining include the mechanism that links local anesthetic binding with the inhibition of gating charge movements, and the molecular nature of the theoretical "hydrophobic pathway" that may be critical for determining the recovery rates from blockade of closed channels, and thus account for both therapeutic and cardiotoxic actions.
Collapse
Affiliation(s)
- G-K Wang
- Pain Research Center, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston MA 02115, USA
| | | |
Collapse
|
33
|
Distinct Nav1.7-dependent pain sensations require different sets of sensory and sympathetic neurons. Nat Commun 2012; 3:791. [PMID: 22531176 PMCID: PMC3337979 DOI: 10.1038/ncomms1795] [Citation(s) in RCA: 205] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 03/19/2012] [Indexed: 11/29/2022] Open
Abstract
Human acute and inflammatory pain requires the expression of voltage-gated sodium channel Nav1.7 but its significance for neuropathic pain is unknown. Here we show that Nav1.7 expression in different sets of mouse sensory and sympathetic neurons underlies distinct types of pain sensation. Ablating Nav1.7 gene (SCN9A) expression in all sensory neurons using Advillin-Cre abolishes mechanical pain, inflammatory pain and reflex withdrawal responses to heat. In contrast, heat-evoked pain is retained when SCN9A is deleted only in Nav1.8-positive nociceptors. Surprisingly, responses to the hotplate test, as well as neuropathic pain, are unaffected when SCN9A is deleted in all sensory neurons. However, deleting SCN9A in both sensory and sympathetic neurons abolishes these pain sensations and recapitulates the pain-free phenotype seen in humans with SCN9A loss-of-function mutations. These observations demonstrate an important role for Nav1.7 in sympathetic neurons in neuropathic pain, and provide possible insights into the mechanisms that underlie gain-of-function Nav1.7-dependent pain conditions. Sodium channel Nav1.7 is essential for acute human pain but its role in chronic neuropathic pain is unclear. Minett and colleagues show that Nav1.7 expression specifically in sympathetic neurons, rather than sensory neurons, is required for the development of chronic neuropathic pain after injury.
Collapse
|
34
|
Abstract
An estimated 15-50% of the population experiences pain at any given time, at great personal and societal cost. Pain is the most common reason patients seek medical attention, and there is a high degree of individual variability in reporting the incidence and severity of symptoms. Research suggests that pain sensitivity and risk for chronic pain are complex heritable traits of polygenic origin. Animal studies and candidate gene testing in humans have provided some progress in understanding the heritability of pain, but the application of the genome-wide association methodology offers a new tool for further elucidating the genetic contributions to normal pain responding and pain in clinical populations. Although the determination of the genetics of pain is still in its infancy, it is clear that a number of genes play a critical role in determining pain sensitivity or susceptibility to chronic pain. This review presents an update of the most recent findings that associate genetic variation with variability in pain and an overview of the candidate genes with the highest translational potential.
Collapse
Affiliation(s)
- Erin E Young
- Department of Anesthesiology, Molecular Epidemiology of Pain Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | | | | |
Collapse
|
35
|
Hartemann A, Attal N, Bouhassira D, Dumont I, Gin H, Jeanne S, Said G, Richard JL. Painful diabetic neuropathy: Diagnosis and management. DIABETES & METABOLISM 2011; 37:377-88. [DOI: 10.1016/j.diabet.2011.06.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 06/13/2011] [Indexed: 01/01/2023]
|
36
|
Theile JW, Jarecki BW, Piekarz AD, Cummins TR. Nav1.7 mutations associated with paroxysmal extreme pain disorder, but not erythromelalgia, enhance Navbeta4 peptide-mediated resurgent sodium currents. J Physiol 2010; 589:597-608. [PMID: 21115638 DOI: 10.1113/jphysiol.2010.200915] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Abnormal pain sensitivity associated with inherited and acquired pain disorders occurs through increased excitability of peripheral sensory neurons in part due to changes in the properties of voltage-gated sodium channels (Navs). Resurgent sodium currents (I(NaR)) are atypical currents believed to be associated with increased excitability of neurons and may have implications in pain. Mutations in Nav1.7 (peripheral Nav isoform) associated with two genetic pain disorders, inherited erythromelalgia (IEM) and paroxysmal extreme pain disorder (PEPD), enhance Nav1.7 function via distinct mechanisms. We show that changes in Nav1.7 function due to mutations associated with PEPD, but not IEM, are important in I(NaR) generation, suggesting that I(NaR) may play a role in pain associated with PEPD. This knowledge provides us with a better understanding of the mechanism of I(NaR) generation and may lead to the development of specialized treatment for pain disorders associated with I(NaR).
Collapse
Affiliation(s)
- Jonathan W Theile
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | |
Collapse
|
37
|
Kim DH, Schwartz CE. The genetics of pain: implications for evaluation and treatment of spinal disease. Spine J 2010; 10:827-40. [PMID: 20615760 DOI: 10.1016/j.spinee.2010.05.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 03/07/2010] [Accepted: 05/22/2010] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Variability in human pain experience appears to be at least partially determined by genetic inheritance. To the extent that awareness of individual pain sensitivity and the tendency to develop chronic pain after injury or surgery would be informative for clinical decision making, development and use of genetic testing for specific pain markers could contribute to improved outcomes in management of spinal disease. PURPOSE To review important and illustrative results from both classical and modern pain genetics studies and to introduce readers to critical definitions and concepts necessary to interpret the growing body of genetics literature relevant to spinal disease. STUDY DESIGN/SETTING Literature review and commentary. METHODS A review was performed of published English language studies in which genetic techniques were used to analyze the molecular basis of nociceptive signaling or processing with a particular emphasis on studies addressing genetic determinants of interindividual variability in pain sensitivity or predisposition to chronic pain. RESULTS There is compelling evidence indicating that interindividual differences in pain sensitivity and the risk of developing chronic pain syndromes are genetically determined. Despite a growing list of putative "pain genes," genetic association studies remain plagued with difficulty replicating initial findings in different cohorts. CONCLUSIONS Genome-wide association studies are potentially powerful means of identifying clinically relevant genetic markers predicting disease susceptibility, severity, and treatment response. However, accurate results require rigorous study design with use of large homogeneous populations and precise phenotypes.
Collapse
Affiliation(s)
- David H Kim
- Department of Orthopaedic Surgery, Tufts University Medical School, New England Baptist Hospital, Boston, MA 02120, USA.
| | | |
Collapse
|
38
|
Rusbridge C, Heath S, Gunn-Moore DA, Knowler SP, Johnston N, McFadyen AK. Feline orofacial pain syndrome (FOPS): a retrospective study of 113 cases. J Feline Med Surg 2010; 12:498-508. [PMID: 20451434 PMCID: PMC7128958 DOI: 10.1016/j.jfms.2010.03.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2010] [Indexed: 12/15/2022]
Abstract
Feline orofacial pain syndrome (FOPS) is a pain disorder of cats with behavioural signs of oral discomfort and tongue mutilation. This report describes the findings from a case series of 113 cats including 100 Burmese. FOPS is suspected to be a neuropathic pain disorder and the predominance within the Burmese cat breed suggests an inherited disorder, possibly involving central and/or ganglion processing of sensory trigeminal information. The disease is characterised by an episodic, typically unilateral, discomfort with pain-free intervals. The discomfort is triggered, in many cases, by mouth movements. The disease is often recurrent and with time may become unremitting - 12% of cases in this series were euthanased as a consequence of the condition. Sensitisation of trigeminal nerve endings as a consequence of oral disease or tooth eruption appears to be an important factor in the aetiology - 63% of cases had a history of oral lesions and at least 16% experienced their first sign of discomfort during eruption of permanent teeth. External factors can also influence the disease as FOPS events could be directly linked to a situation causing anxiety in 20% of cats. FOPS can be resistant to traditional analgesics and in some cases successful management required anti-convulsants with an analgesic effect.
Collapse
Affiliation(s)
- Clare Rusbridge
- Stone Lion Veterinary Centre, 41 High Street, Wimbledon SW19 5AU, UK.
| | | | | | | | | | | |
Collapse
|
39
|
Acute PAR2 activation reduces alpha, beta-MeATP sensitive currents in rat dorsal root ganglion neurons. Neuroreport 2010; 21:227-32. [PMID: 20118742 DOI: 10.1097/wnr.0b013e328336b6c8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
It has been reported that proteinase-activated receptor 2 (PAR2) receptor activation enhances the animal's pain response and PAR2 coexpresses with P2X3 in dorsal root ganglion neurons. However, whether PAR2 activation has a direct impact on P2X3 currents is still not clear. In this study, we performed the patch-clamp experiments in cultured dorsal root ganglion neurons and found that when incubated with trypsin or the PAR2 agonist SL-NH2 for a short time (3 min), instead of increasing, P2X3 currents amplitude decreased significantly. Meanwhile, the opening of P2X3 ion channel accelerated. Protein kinase A inhibitor H89 could not reverse above phenomenon, but played a synergistic effect on the contrary. These results suggest that the enhanced pain response caused by PAR2 activation is not through direct increase of the P2X3 current amplitude, and the acceleration of P2X3 opening may participate in the enhanced pain response in a long-time view. Moreover, protein kinase A does not participate in the inhibition of P2X3 currents caused by PAR2 activation.
Collapse
|
40
|
Lampert A, O'Reilly AO, Reeh P, Leffler A. Sodium channelopathies and pain. Pflugers Arch 2010; 460:249-63. [PMID: 20101409 DOI: 10.1007/s00424-009-0779-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Revised: 12/13/2009] [Accepted: 12/18/2009] [Indexed: 12/19/2022]
Abstract
Chronic pain often represents a severe, debilitating condition. Up to 10% of the worldwide population are affected, and many patients are poorly responsive to current treatment strategies. Nociceptors detect noxious conditions to produce the sensation of pain, and this signal is conveyed to the CNS by means of action potentials. The fast upstroke of action potentials is mediated by voltage-gated sodium channels, of which nine pore-forming alpha-subunits (Nav1.1-1.9) have been identified. Heterogeneous functional properties and distinct expression patterns denote specialized functions of each subunit. The Nav1.7 and Nav1.8 subunits have emerged as key molecules involved in peripheral pain processing and in the development of an increased pain sensitivity associated with inflammation and tissue injury. Several mutations in the SCN9A gene encoding for Nav1.7 have been identified as important cellular substrates for different heritable pain syndromes. This review aims to cover recent progress on our understanding of how biophysical properties of mutant Nav1.7 translate into an aberrant electrogenesis of nociceptors. We also recapitulate the role of Nav1.8 for peripheral pain processing and of additional sodium channelopathies which have been linked to disorders with pain as a significant component.
Collapse
Affiliation(s)
- Angelika Lampert
- Department of Physiology and Pathophysiology, Friedrich-Alexander University Erlangen-Nuremberg, Universitätsstrasse 17, 91054, Erlangen, Germany.
| | | | | | | |
Collapse
|
41
|
McLaughlin CR, Acosta MC, Luna C, Liu W, Belmonte C, Griffith M, Gallar J. Regeneration of functional nerves within full thickness collagen-phosphorylcholine corneal substitute implants in guinea pigs. Biomaterials 2009; 31:2770-8. [PMID: 20042235 DOI: 10.1016/j.biomaterials.2009.12.031] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 12/11/2009] [Indexed: 12/17/2022]
Abstract
Our objective was to evaluate promotion of tissue and nerve regeneration by extracellular matrix (ECM) mimics, using corneal implantation as a model system. Porcine type I collagen and 2-methacryloyloxyethyl phosphorylcholine (MPC) were crosslinked using 1-ethyl-3-(3-dimethyl aminopropyl) carbodiimide (EDC) and N-hydroxysuccinimide (NHS) and moulded into appropriate corneal dimensions to serve as substitutes for natural corneal ECM. These were implanted as full thickness grafts by penetrating keratoplasty into the corneas of guinea pigs after removal of the host tissue, and tracked over eight months, by clinical examination, slit-lamp biomicroscopy, and esthesiometry. Histopathology and ex vivo nerve terminal impulse recordings were performed at three months and at eight months. The implants promoted regeneration of corneal cells, nerves and the tear film, while retaining optical clarity. After three months, electrophysiological recordings showed evidence of mechano-nociceptors, and polymodal units inside the implants, while cold-sensitive units were present only on the peripheral host cornea. Following eight months, the incidence of nerve activity and the frequency of spontaneous firing were higher than in control eyes as reported for regenerating fibers. Active cold nerve terminals also innervated the implant area. We show that ECM mimetic materials can promote regeneration of corneal cells and functional nerves. The simplicity in fabrication and demonstrated functionality shows potential for ECM substitutes in future clinical applications.
Collapse
Affiliation(s)
- Christopher R McLaughlin
- University of Ottawa Eye Institute, The Ottawa Hospital, General Campus, 501 Smyth Road, Ottawa, ONK1H 8L6, Canada
| | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
The nervous system detects and interprets a wide range of thermal and mechanical stimuli, as well as environmental and endogenous chemical irritants. When intense, these stimuli generate acute pain, and in the setting of persistent injury, both peripheral and central nervous system components of the pain transmission pathway exhibit tremendous plasticity, enhancing pain signals and producing hypersensitivity. When plasticity facilitates protective reflexes, it can be beneficial, but when the changes persist, a chronic pain condition may result. Genetic, electrophysiological, and pharmacological studies are elucidating the molecular mechanisms that underlie detection, coding, and modulation of noxious stimuli that generate pain.
Collapse
Affiliation(s)
- Allan I Basbaum
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA.
| | | | | | | |
Collapse
|
43
|
Bhattacharya A, Wickenden AD, Chaplan SR. Sodium channel blockers for the treatment of neuropathic pain. Neurotherapeutics 2009; 6:663-78. [PMID: 19789071 PMCID: PMC5084288 DOI: 10.1016/j.nurt.2009.08.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Drugs that block voltage-gated sodium channels are efficacious in the management of neuropathic pain. Accordingly, this class of ion channels has been a major focus of analgesic research both in academia and in the pharmaceutical/biotechnology industry. In this article, we review the history of the use of sodium channel blockers, describe the current status of sodium channel drug discovery, highlight the challenges and hurdles to attain sodium channel subtype selectivity, and review the potential usefulness of selective sodium channel blockers in neuropathic pain.
Collapse
Affiliation(s)
- Anindya Bhattacharya
- grid.417429.dPain & Related Disorders Team, Johnson & Johnson Pharmaceutical Research & Development, LLC, 3210 Merryfield Row, 92121 San Diego, CA
| | - Alan D. Wickenden
- grid.417429.dPain & Related Disorders Team, Johnson & Johnson Pharmaceutical Research & Development, LLC, 3210 Merryfield Row, 92121 San Diego, CA
| | - Sandra R. Chaplan
- grid.417429.dPain & Related Disorders Team, Johnson & Johnson Pharmaceutical Research & Development, LLC, 3210 Merryfield Row, 92121 San Diego, CA
| |
Collapse
|
44
|
Schestatsky P, Nascimento OJM. What do general neurologists need to know about neuropathic pain? ARQUIVOS DE NEURO-PSIQUIATRIA 2009; 67:741-9. [DOI: 10.1590/s0004-282x2009000400039] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Neuropathic pain (NP) is defined as pain caused by lesion or dysfunction of the somatosensory system, as a result of abnormal activation of the nociceptive pathway (small fibers and spinothalamic tracts). The most common causes of this syndrome are the following: diabetes, post-herpetic neuralgia, trigeminal neuralgia, stroke, multiple sclerosis, spinal cord injury, HIV infection, cancer. In the last few years, the NP has been receiving special attention for two main reasons: (1) therapeutical refractoriness of a variety of pain syndromes with predominant neuropathic characteristics and (2) the development of diagnostic tools for neuropathic pain complaints. The present review article provides relevant information on the understanding and recognition of NP, as well as evidence-based therapeutic approaches.
Collapse
|