401
|
Greenbaum L, Tegeder I, Barhum Y, Melamed E, Roditi Y, Djaldetti R. Contribution of genetic variants to pain susceptibility in Parkinson disease. Eur J Pain 2012; 16:1243-50. [PMID: 22473870 DOI: 10.1002/j.1532-2149.2012.00134.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2012] [Indexed: 02/03/2023]
Abstract
BACKGROUND Pain is a one of the most disturbing non-motor symptoms of Parkinson disease (PD). The susceptibility to pain varies substantially among patients with PD. The aim of this study was to assess a potential association of genetic variants to PD-related pain. METHODS We analysed 20 candidate SNPs from 12 genes previously reported to be associated with various pain phenotypes in a homogeneous group of 229 Israeli Jewish PD patients, with and without pain (n = 165 and 64, respectively). RESULTS The statistical analysis accounted for the potential influence of demographic and clinical factors. The non-synonymous rs6746030 single nucleotide polymorphism (SNP) of the SCN9A gene, which alters the coding sequence of the sodium channel Nav1.7 (arginine to tryptophan), was nominally associated with PD-related pain susceptibility (p = 0.037), as well as with central and musculoskeletal pain subtypes independently. The synonymous rs324419 SNP of the FAAH gene which encodes fatty acid amide hydrolase, a cannabinoid metabolizing enzyme, was associated with PD-related pain (p = 0.006) and specifically with the musculoskeletal subtype. The FAAH haplotype of rs324419 and rs2295633 SNPs, which was previously associated with the variability in pain response in humans, was also associated with PD-related pain (p = 0.012) and specifically with PD-related musculoskeletal pain. CONCLUSIONS Variants within in the SCN9A and FAAH genes were associated with the risk of pain in PD patients. These findings may contribute to our understanding of pain mechanisms of PD and to direct future therapies.
Collapse
Affiliation(s)
- L Greenbaum
- Laboratory of Biological Psychiatry, Department of Psychiatry, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | |
Collapse
|
402
|
Mogil JS. Pain genetics: past, present and future. Trends Genet 2012; 28:258-66. [PMID: 22464640 DOI: 10.1016/j.tig.2012.02.004] [Citation(s) in RCA: 226] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 02/16/2012] [Accepted: 02/22/2012] [Indexed: 01/01/2023]
Abstract
Chronic pain is a classic example of gene × environment interaction: inflammatory and/or nerve injuries are known or suspected to be the etiology of most chronic pain syndromes, but only a small minority of those subjected to such injuries actually develop chronic pain. Once chronic pain has developed, pain severity and analgesic response are also highly variable among individuals. Although animal genetics studies have been ongoing for over two decades, only recently have comprehensive human twin studies and large-scale association studies been performed. Here, I review recent and accelerating progress in, and continuing challenges to, the identification of genes contributing to such variability. Success in this endeavor will hopefully lead to both better management of pain using currently available therapies and the development and/or prioritizing of new ones.
Collapse
Affiliation(s)
- Jeffrey S Mogil
- Department of Psychology, McGill University, Montreal, QC, H3A 1B1, Canada.
| |
Collapse
|
403
|
Chattopadhyay M, Zhou Z, Hao S, Mata M, Fink DJ. Reduction of voltage gated sodium channel protein in DRG by vector mediated miRNA reduces pain in rats with painful diabetic neuropathy. Mol Pain 2012; 8:17. [PMID: 22439790 PMCID: PMC3388457 DOI: 10.1186/1744-8069-8-17] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 03/22/2012] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Painful neuropathy is a common complication of diabetes. Previous studies have identified significant increases in the amount of voltage gated sodium channel isoforms Na(V)1.7 and Na(V)1.3 protein in the dorsal root ganglia (DRG) of rats with streptozotocin (STZ)-induced diabetes. We found that gene transfer-mediated release of the inhibitory neurotransmitters enkephalin or gamma amino butyric acid (GABA) from DRG neurons in diabetic animals reduced pain-related behaviors coincident with a reduction in Na(V)1.7 protein levels in DRG in vivo. To further evaluate the role of Na(V)α subunit levels in DRG in the pathogenesis of pain in diabetic neuropathy, we constructed a non-replicating herpes simplex virus (HSV)-based vector expressing a microRNA (miRNA) against Na(V)α subunits. RESULTS Subcutaneous inoculation of the miRNA-expressing HSV vector into the feet of diabetic rats to transduce DRG resulted in a reduction in Na(V)α subunit levels in DRG neurons, coincident with a reduction in cold allodynia, thermal hyperalgesia and mechanical hyperalgesia. CONCLUSIONS These data support the role of increased Na(V)α protein in DRG in the pathogenesis of pain in diabetic neuropathy, and provide a proof-of-principle demonstration for the development of a novel therapy that could be used to treat intractable pain in patients with diabetic neuropathy.
Collapse
Affiliation(s)
- Munmun Chattopadhyay
- Department of Neurology, University of Michigan and VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Zhigang Zhou
- Department of Neurology, University of Michigan and VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
- Department of Pathology, University of North Carolina, Chapel Hill, NC, USA
| | - Shuanglin Hao
- Department of Neurology, University of Michigan and VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
- Department of Anesthesiology, University of Miami, Miami, FL, USA
| | - Marina Mata
- Department of Neurology, University of Michigan and VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - David J Fink
- Department of Neurology, University of Michigan and VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
- 1500 E Medical Center Drive, Ann Arbor, MI 48109, USA
| |
Collapse
|
404
|
Lin Z, Chen Q, Lee M, Cao X, Zhang J, Ma D, Chen L, Hu X, Wang H, Wang X, Zhang P, Liu X, Guan L, Tang Y, Yang H, Tu P, Bu D, Zhu X, Wang K, Li R, Yang Y. Exome sequencing reveals mutations in TRPV3 as a cause of Olmsted syndrome. Am J Hum Genet 2012; 90:558-64. [PMID: 22405088 DOI: 10.1016/j.ajhg.2012.02.006] [Citation(s) in RCA: 250] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 12/30/2011] [Accepted: 02/07/2012] [Indexed: 01/27/2023] Open
Abstract
Olmsted syndrome (OS) is a rare congenital disorder characterized by palmoplantar and periorificial keratoderma, alopecia in most cases, and severe itching. The genetic basis for OS remained unidentified. Using whole-exome sequencing of case-parents trios, we have identified a de novo missense mutation in TRPV3 that produces p.Gly573Ser in an individual with OS. Nucleotide sequencing of five additional affected individuals also revealed missense mutations in TRPV3 (which produced p.Gly573Ser in three cases and p.Gly573Cys and p.Trp692Gly in one case each). Encoding a transient receptor potential vanilloid-3 cation channel, TRPV3 is primarily expressed in the skin, hair follicles, brain, and spinal cord. In transfected HEK293 cells expressing TRPV3 mutants, much larger inward currents were recorded, probably because of the constitutive opening of the mutants. These gain-of-function mutations might lead to elevated apoptosis of keratinocytes and consequent skin hyperkeratosis in the affected individuals. Our findings suggest that TRPV3 plays essential roles in skin keratinization, hair growth, and possibly itching sensation in humans and selectively targeting TRPV3 could provide therapeutic potential for keratinization or itching-related skin disorders.
Collapse
|
405
|
Imaging the neural correlates of neuropathic pain and pleasurable relief associated with inherited erythromelalgia in a single subject with quantitative arterial spin labelling. Pain 2012; 153:1122-1127. [PMID: 22365309 PMCID: PMC3438450 DOI: 10.1016/j.pain.2011.12.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 12/05/2011] [Accepted: 12/12/2011] [Indexed: 11/23/2022]
Abstract
We identified a patient with severe inherited erythromelalgia secondary to an L858F mutation in the voltage-gated sodium channel Nav1.7. The patient reported severe ongoing foot pain, which was exquisitely sensitive to limb cooling. We confirmed this heat hypersensitivity using quantitative sensory testing. Additionally, we employed a novel perfusion imaging technique in a simple block design to assess her baseline erythromelalgia pain vs cooling relief. Robust activations of key pain, pain-affect, and reward-related centres were observed. This combined approach allowed us to confirm the presence of a temperature-sensitive channelopathy of peripheral neurons and to investigate the neural correlates of tonic neuropathic pain and relief in a single subject.
Collapse
|
406
|
Affiliation(s)
- Henry Houlden
- UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK.
| |
Collapse
|
407
|
Nickel FT, Seifert F, Lanz S, Maihöfner C. Mechanisms of neuropathic pain. Eur Neuropsychopharmacol 2012; 22:81-91. [PMID: 21672666 DOI: 10.1016/j.euroneuro.2011.05.005] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 04/12/2011] [Accepted: 05/14/2011] [Indexed: 12/14/2022]
Abstract
Neuropathic pain is a disease of global burden. Its symptoms include spontaneous and stimulus-evoked painful sensations. Several maladaptive mechanisms underlying these symptoms have been elucidated in recent years: peripheral sensitization of nociception, abnormal excitability of afferent neurons, central sensitization comprising pronociceptive facilitation, disinhibition of nociception and central reorganization processes, and sympathetically maintained pain. This review aims to illustrate these pathophysiological principles, focussing on molecular and neurophysiological findings. Finally therapeutic options based on these findings are discussed.
Collapse
Affiliation(s)
- Florian T Nickel
- Department of Neurology, University of Erlangen-Nuremberg, Germany
| | | | | | | |
Collapse
|
408
|
Goldberg YP, Price N, Namdari R, Cohen CJ, Lamers MH, Winters C, Price J, Young CE, Verschoof H, Sherrington R, Pimstone SN, Hayden MR. Treatment of Nav1.7-mediated pain in inherited erythromelalgia using a novel sodium channel blocker. Pain 2012; 153:80-85. [DOI: 10.1016/j.pain.2011.09.008] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 08/31/2011] [Accepted: 09/12/2011] [Indexed: 11/26/2022]
|
409
|
Smith ESJ, Omerbasic D, Lechner SG, Anirudhan G, Lapatsina L, Lewin GR. The Molecular Basis of Acid Insensitivity in the African Naked Mole-Rat. Science 2011; 334:1557-60. [DOI: 10.1126/science.1213760] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
410
|
Gaudioso C, Hao J, Martin-Eauclaire MF, Gabriac M, Delmas P. Menthol pain relief through cumulative inactivation of voltage-gated sodium channels. Pain 2011; 153:473-484. [PMID: 22172548 DOI: 10.1016/j.pain.2011.11.014] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 09/19/2011] [Accepted: 11/14/2011] [Indexed: 01/02/2023]
Abstract
Menthol is a natural compound of plant origin known to produce cool sensation via the activation of the TRPM8 channel. It is also frequently part of topical analgesic drugs available in a pharmacy, although its mechanism of action is still unknown. Compelling evidence indicates that voltage-gated Na(+) channels are critical for experiencing pain sensation. We tested the hypothesis that menthol may block voltage-gated Na(+) channels in dorsal root ganglion (DRG) neurons. By use of a patch clamp, we evaluated the effects of menthol application on tetrodotoxin (TTX)-resistant Nav1.8 and Nav1.9 channel subtypes in DRG neurons, and on TTX-sensitive Na(+) channels in immortalized DRG neuron-derived F11 cells. The results indicate that menthol inhibits Na(+) channels in a concentration-, voltage-, and frequency-dependent manner. Menthol promoted fast and slow inactivation states, causing use-dependent depression of Na(+) channel activity. In current clamp recordings, menthol inhibited firing at high-frequency stimulation with minimal effects on normal neuronal activity. We found that low concentrations of menthol cause analgesia in mice, relieving pain produced by a Na(+) channel-targeting toxin. We conclude that menthol is a state-selective blocker of Nav1.8, Nav1.9, and TTX-sensitive Na(+) channels, indicating a role for Na(+) channel blockade in the efficacy of menthol as topical analgesic compound.
Collapse
Affiliation(s)
- Christelle Gaudioso
- Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Aix-Marseille Université, UMR 6231, CNRS, CS80011, Bd Pierre Dramard, 13344 Marseille Cedex 15, France
| | | | | | | | | |
Collapse
|
411
|
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
|
412
|
Abstract
Degenerative disk disease is a strong etiologic risk factor of chronic low back pain (LBP). A multidisciplinary approach to treatment is often warranted. Patient education, medication, and cognitive behavioral therapies are essential in the treatment of chronic LBP sufferers. Surgical intervention with a rehabilitation regime is sometimes advocated. Prognostic factors related to the outcome of different treatments include maladaptive pain coping and genetics. The identification of pain genes may assist in determining individuals susceptible to pain and in patient selection for appropriate therapy. Biologic therapies show promise, but clinical trials are needed before advocating their use in humans.
Collapse
|
413
|
McArthur JR, Singh G, McMaster D, Winkfein R, Tieleman DP, French RJ. Interactions of key charged residues contributing to selective block of neuronal sodium channels by μ-conotoxin KIIIA. Mol Pharmacol 2011; 80:573-84. [PMID: 21709136 DOI: 10.1124/mol.111.073460] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Voltage-gated sodium channels are important in initiating and propagating nerve impulses in various tissues, including cardiac muscle, skeletal muscle, the brain, and the peripheral nerves. Hyperexcitability of these channels leads to such disorders as cardiac arrhythmias (Na(v)1.5), myotonias (Na(v)1.4), epilepsies (Na(v)1.2), and pain (Na(v)1.7). Thus, there is strong motivation to identify isoform-specific blockers and the molecular determinants underlying their selectivity among these channels. μ-Conotoxin KIIIA blocks rNa(v)1.2 (IC(50), 5 nM), rNa(v)1.4 (37 nM), and hNa(v)1.7 (97 nM), expressed in mammalian cells, with high affinity and a maximal block at saturating concentrations of 90 to 95%. Mutations of charged residues on both the toxin and channel modulate the maximal block and/or affinity of KIIIA. Two toxin substitutions, K7A and R10A, modulate the maximal block (52-70%). KIIIA-H12A and R14A were the only derivatives tested that altered Na(v) isoform specificity. KIIIA-R14A showed the highest affinity for Na(v)1.7, a channel involved in pain signaling. Wild-type KIIIA has a 2-fold higher affinity for Na(v)1.4 than for Na(v)1.7, which can be attributed to a missing outer vestibule charge in domain III of Na(v)1.7. Reciprocal mutations Na(v)1.4 D1241I and Na(v)1.7 I1410D remove the affinity differences between these two channels for wild-type KIIIA without affecting their affinities for KIIIA-R14A. KIIIA is the first μ-conotoxin to show enhanced activity as pH is lowered, apparently resulting from titration of the free N terminus. Removal of this free amino group reduced the pH sensitivity by 10-fold. Recognition of these molecular determinants of KIIIA block may facilitate further development of subtype-specific, sodium channel blockers to treat hyperexcitability disorders.
Collapse
Affiliation(s)
- J R McArthur
- Department of Physiology and Pharmacology, and the Hotchkiss Brain Institute, Canada
| | | | | | | | | | | |
Collapse
|
414
|
Yuan R, Zhang X, Deng Q, Si D, Wu Y, Gao F, Zhou B. Two novel SCN9A gene heterozygous mutations may cause partial deletion of pain perception. PAIN MEDICINE 2011; 12:1510-4. [PMID: 21939494 DOI: 10.1111/j.1526-4637.2011.01237.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJECTIVE The physiological sensation of pain and rapid response to stimuli serve as an adaptive way to avoid harmful situations. Our purpose was to investigate why this protection disappears or almost disappears for patients with congenital indifference to pain (CIP). DESIGN The study was designed as a case report by scanning the candidate genes within CIP patients. SETTING The study was set at the Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. PATIENTS We reported patients from two Chinese families that showed insensitivity to pain and were diagnosed with CIP by a neurologist. Different from recently reported studies, our patients were not entirely painless, but demonstrated little pain sensation from injuries. MEASURES The measures made were novel mutations within SCN9A. RESULTS Sequence analysis of candidate genes of two affected individuals identified two novel heterozygous mutations (M899I and M932L) in the SCN9A gene. Furthermore, a novel nonsynonymous single-nucleotide polymorphism (SNP) within the SCN9A gene was revealed in affected proband and several unaffected family members. This polymorphism (c. 3312G&T, which produces the amino acid substitution V1104L in human Nav1.7), is present in 6.5% of healthy Chinese. CONCLUSIONS We speculate that the mutations may be the cause of partial deletion of pain perceptionin in our probands, and the novel polymorphism V1104L may have a predictive role in the pain sensation of healthy individuals.
Collapse
Affiliation(s)
- Ruimei Yuan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | |
Collapse
|
415
|
Erythromelalgia presenting after neurosurgical intervention in a patient with multiple malformations of the spinal cord. J Am Acad Dermatol 2011; 65:e120-e122. [PMID: 21920230 DOI: 10.1016/j.jaad.2011.03.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 02/28/2011] [Accepted: 03/10/2011] [Indexed: 11/22/2022]
|
416
|
Abstract
OBJECTIVES In the clinical setting, there is marked intersubject variability in the intensity of pain reported by patients with apparently similar pain states, as well as widely differing analgesic dosing requirements between individuals to produce satisfactory pain relief with tolerable side-effects. Genetic and environmental factors as well as their interaction are implicated, and these are discussed in this review. KEY FINDINGS Pioneering work undertaken in mice more than a decade ago, showed a strong genetic contribution to levels of nociception/hypersensitivity as well as levels of antinociception produced by commonly available analgesic agents. To date more than 300 candidate 'pain' genes have been identified as potentially contributing to heritable differences in pain sensitivity and analgesic responsiveness in animals and humans, with this information available in a publicly accessible database http://www.jbldesign.com/jmogil/enter.html. Since then, many genetic association studies have been conducted in humans to investigate the possibility that single nucleotide polymorphisms (SNPs) in an individual gene may explain drug inefficacy or excessive toxicity experienced by a small subset of the whole population who have the rare allele for a particular SNP. SUMMARY Despite the fact that SNPs in more than 20 genes that affect pain sensitivity or contribute to interindividual variability in responses to analgesic medications have been identified in the human genome, much of the data is conflicting. Apart from deficiencies in the design and conduct of human genetic association studies, recent research from other fields has implicated epigenetic mechanisms that facilitate dynamic gene-environment communication, as a possible explanation.
Collapse
Affiliation(s)
- Arjun Muralidharan
- The University of Queensland, Centre for Integrated Preclinical Drug Development and School of Pharmacy, Steele Building, St Lucia Campus, Brisbane, Queensland, Australia
| | | |
Collapse
|
417
|
Abstract
PURPOSE OF REVIEW The causes of inflammatory pain and neuropathic pain are fundamentally different. There are, however, common mechanisms underlying the generation of each pain state. We will discuss some specific elements observed in both tissue and nerve injury pain states and consider the hypothesis that these two states actually demonstrate a convergence over time. RECENT FINDINGS The increased pain sensation following tissue and nerve injury results from several mechanisms, including altered ion channel expression in dorsal root ganglion neurons, enhanced dorsal horn glutamate release from primary afferents, enhanced glutamate receptor function in second-order neurons, disinhibition in the dorsal horn and glia cell activation. The role of specific subtypes of receptors, ion channels and glutamate transporters is revealed at peripheral and central sites. Importantly over time, a number of changes, in the dorsal root ganglion and in dorsal horn observed after tissue injury resemble changes observed after nerve injury. SUMMARY Recognition of mechanisms common to both inflammatory pain and neuropathic pain might shed light on the understanding of the transition from acute pain to persistent pain.
Collapse
Affiliation(s)
- Qinghao Xu
- Department of Anesthesiology, University of California-San Diego, La Jolla, CA 92093, USA.
| | | |
Collapse
|
418
|
Fryatt AG, Mulheran M, Egerton J, Gunthorpe MJ, Grubb BD. Ototrauma induces sodium channel plasticity in auditory afferent neurons. Mol Cell Neurosci 2011; 48:51-61. [PMID: 21708262 PMCID: PMC3176910 DOI: 10.1016/j.mcn.2011.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Revised: 06/02/2011] [Accepted: 06/08/2011] [Indexed: 12/19/2022] Open
Abstract
Exposure to intense sound can cause damage to the delicate sensory and neuronal components of the cochlea leading to hearing loss. Such damage often causes the dendrites of the spiral ganglion neurons (SGN), the neurons that provide the afferent innervation of the hair cells, to swell and degenerate thus damaging the synapse. In models of neuropathic pain, axotomy, another form of afferent nerve damage, is accompanied by altered voltage-gated sodium channel (VGSC) expression, leading to neuronal hyperactivity. In this study, adult Wistar rats were exposed to noise which produced a mild, 20 dB hearing threshold elevation and their VGSC expression was investigated. Quantitative PCR showed decreased NaV1.1 and NaV1.6 mRNA expression in the SGN following noise exposure (29% and 56% decrease respectively) while NaV1.7 mRNA expression increased by approximately 20% when compared to control rats. Immunohistochemistry extended these findings, revealing increased staining for NaV1.1 along the SGN dendrites and NaV1.7 in the cell bodies after noise. These results provide the first evidence for selective changes in VGSC expression following moderate noise-induced hearing loss and could contribute to elevated hearing thresholds and to the generation of perceptual anomalies commonly associated with cochlear damage, such as tinnitus and hyperacusis.
Collapse
Affiliation(s)
- Alistair G Fryatt
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, LE1 9HN UK.
| | | | | | | | | |
Collapse
|
419
|
Bregman H, Berry L, Buchanan JL, Chen A, Du B, Feric E, Hierl M, Huang L, Immke D, Janosky B, Johnson D, Li X, Ligutti J, Liu D, Malmberg A, Matson D, McDermott J, Miu P, Nguyen HN, Patel VF, Waldon D, Wilenkin B, Zheng XM, Zou A, McDonough SI, DiMauro EF. Identification of a Potent, State-Dependent Inhibitor of Nav1.7 with Oral Efficacy in the Formalin Model of Persistent Pain. J Med Chem 2011; 54:4427-45. [DOI: 10.1021/jm200018k] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Howard Bregman
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Loren Berry
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - John L. Buchanan
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - April Chen
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Bingfan Du
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Elma Feric
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Markus Hierl
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Liyue Huang
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - David Immke
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Brett Janosky
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Danielle Johnson
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Xingwen Li
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Joseph Ligutti
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Dong Liu
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Annika Malmberg
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - David Matson
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Jeff McDermott
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Peter Miu
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Hanh Nho Nguyen
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Vinod F. Patel
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Daniel Waldon
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Ben Wilenkin
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Xiao Mei Zheng
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Anruo Zou
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Stefan I. McDonough
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Erin F. DiMauro
- Department of Chemistry Research and Discovery, ‡Department of Pharmacokinetics and Drug Metabolism, and §Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department of Lead Discovery, and ⊥Department of Neuroscience, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| |
Collapse
|
420
|
Chowdhury S, Chafeev M, Liu S, Sun J, Raina V, Chui R, Young W, Kwan R, Fu J, Cadieux JA. Discovery of XEN907, a spirooxindole blocker of NaV1.7 for the treatment of pain. Bioorg Med Chem Lett 2011; 21:3676-81. [DOI: 10.1016/j.bmcl.2011.04.088] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 04/14/2011] [Accepted: 04/19/2011] [Indexed: 12/19/2022]
|
421
|
Macsari I, Sandberg L, Besidski Y, Gravenfors Y, Ginman T, Bylund J, Bueters T, Eriksson AB, Lund PE, Venyike E, Arvidsson PI. Phenyl isoxazole voltage-gated sodium channel blockers: structure and activity relationship. Bioorg Med Chem Lett 2011; 21:3871-6. [PMID: 21641215 DOI: 10.1016/j.bmcl.2011.05.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 05/11/2011] [Accepted: 05/12/2011] [Indexed: 11/26/2022]
Abstract
Blocking of certain sodium channels is considered to be an attractive mechanism to treat chronic pain conditions. Phenyl isoxazole carbamate 1 was identified as a potent and selective Na(V)1.7 blocker. Structural analogues of 1, both carbamates, ureas and amides, were proven to be useful in establishing the structure-activity relationship and improving ADME related properties. Amide 24 showed a good overall in vitro profile, that translated well to rat in vivo PK.
Collapse
Affiliation(s)
- Istvan Macsari
- Medicinal Chemistry, CNSP iMed Science, AstraZeneca R&D, Innovative Medicines, SE-15185 Södertälje, Sweden.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
422
|
Erythema and burning pain in the vulva: a possible phenotype of erythromelalgia. Case Rep Med 2011; 2011:374167. [PMID: 21541228 PMCID: PMC3085391 DOI: 10.1155/2011/374167] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Accepted: 02/23/2011] [Indexed: 12/02/2022] Open
Abstract
We report a case of burning vulvar pain accompanied by erythema responding to an oral combination of a benzodiazepine and a beta blocker. The positive response to two medication classes used in the treatment of erythromelalgia supports the possibility of a localized manifestation of this disorder in the genital region.
Collapse
|
423
|
Abstract
Axons are generally considered as reliable transmission cables in which stable propagation occurs once an action potential is generated. Axon dysfunction occupies a central position in many inherited and acquired neurological disorders that affect both peripheral and central neurons. Recent findings suggest that the functional and computational repertoire of the axon is much richer than traditionally thought. Beyond classical axonal propagation, intrinsic voltage-gated ionic currents together with the geometrical properties of the axon determine several complex operations that not only control signal processing in brain circuits but also neuronal timing and synaptic efficacy. Recent evidence for the implication of these forms of axonal computation in the short-term dynamics of neuronal communication is discussed. Finally, we review how neuronal activity regulates both axon morphology and axonal function on a long-term time scale during development and adulthood.
Collapse
Affiliation(s)
- Dominique Debanne
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| | - Emilie Campanac
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| | - Andrzej Bialowas
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| | - Edmond Carlier
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| | - Gisèle Alcaraz
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| |
Collapse
|
424
|
Hughes JP, Rees S, Kalindjian SB, Philpott KL. Principles of early drug discovery. Br J Pharmacol 2011; 162:1239-49. [PMID: 21091654 PMCID: PMC3058157 DOI: 10.1111/j.1476-5381.2010.01127.x] [Citation(s) in RCA: 1428] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 10/07/2010] [Accepted: 11/08/2010] [Indexed: 12/13/2022] Open
Abstract
Developing a new drug from original idea to the launch of a finished product is a complex process which can take 12-15 years and cost in excess of $1 billion. The idea for a target can come from a variety of sources including academic and clinical research and from the commercial sector. It may take many years to build up a body of supporting evidence before selecting a target for a costly drug discovery programme. Once a target has been chosen, the pharmaceutical industry and more recently some academic centres have streamlined a number of early processes to identify molecules which possess suitable characteristics to make acceptable drugs. This review will look at key preclinical stages of the drug discovery process, from initial target identification and validation, through assay development, high throughput screening, hit identification, lead optimization and finally the selection of a candidate molecule for clinical development.
Collapse
Affiliation(s)
- J P Hughes
- MedImmune Inc, Granta Park, Cambridge, UK
| | | | | | | |
Collapse
|
425
|
Staud R, Price DD, Janicke D, Andrade E, Hadjipanayis AG, Eaton WT, Kaplan L, Wallace MR. Two novel mutations of SCN9A (Nav1.7) are associated with partial congenital insensitivity to pain. Eur J Pain 2011; 15:223-30. [PMID: 20692858 PMCID: PMC2978801 DOI: 10.1016/j.ejpain.2010.07.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Revised: 06/13/2010] [Accepted: 07/03/2010] [Indexed: 10/19/2022]
Abstract
Insensitivity to pain is a rare disorder that is commonly associated with Hereditary Sensory and Autonomic Neuropathies (HSAN I-V) resulting often in autonomic dysfunction and premature death. Very few individuals have been reported with pain insensitivity lacking such autonomic neuropathies. We performed genetic, neurologic, psychological, and psychophysical evaluations in such an individual (OMIM 243000) and her first degree relatives. Sequence analysis of genomic DNA revealed two novel SCN9A mutations in this index case (IC). One was a non-conservative missense mutation (C1719R) in exon 26 present only in the IC and one parent. Further sequence analysis of the child's DNA revealed a 1-bp splice donor deletion in intron 17 which was also present in the other parent and one sibling. Detailed psychophysical testing was used to phenotypically characterize the IC, her family members, and 10 matched normal controls. Similar to family members and controls the IC showed normal somatosensory functioning for non-nociceptive mechanoreception and warmth. However, she demonstrated diminished ability to detect cool temperatures combined with profound deficits in heat and mechanical nociception. Congenital insensitivity to pain in our IC was associated with two novel SCN9A mutations which most likely resulted in a Nav1.7 channelopathy. However, in contrast to individuals with other SCN9A mutations, the observed pain insensitivity was relative and not absolute, which may be consistent with hypomorphic effects of one or both mutations. The ability to sense at least some danger signals may be advantageous and ameliorate the otherwise increased morbidity and mortality of some individuals with congenital insensitivity to pain.
Collapse
Affiliation(s)
- Roland Staud
- Department of Medicine, University of Florida, Gainesville, FL 32610, United States.
| | | | | | | | | | | | | | | |
Collapse
|
426
|
Dickinson BD, Head CA, Gitlow S, Osbahr AJ. Maldynia: pathophysiology and management of neuropathic and maladaptive pain--a report of the AMA Council on Science and Public Health. PAIN MEDICINE 2011; 11:1635-53. [PMID: 21044254 DOI: 10.1111/j.1526-4637.2010.00986.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Because of disparate taxonomic arrays for classification, the American Academy of Pain Medicine has proposed categorizing pain on a neurobiologic basis as eudynia (nociceptive pain), Greek for "good pain," or maldynia (maladaptive pain), Greek for "bad pain." The latter has been viewed as maladaptive because it may occur in the absence of ongoing noxious stimuli and does not promote healing and repair. OBJECTIVE To address recent findings on the pathogenesis of pain following neural injury and consider whether the development of maladaptive pain justifies its classification as a disease and to briefly discuss the scope of pharmacologic and non-pharmacologic approaches employed in patients with such pain. METHODS English language reports on studies using human subjects were selected from a PubMed search of the literature from 1995 to August 2010 and from the Cochrane Library. Further information was obtained from Internet sites of medical specialty and other societies devoted to pain management. RESULTS Neural damage to either the peripheral or central nervous system provokes multiple processes including peripheral and central sensitization, ectopic activity, neuronal cell death, disinhibition, altered gene expression, and abnormal sprouting and cellular connectivity. A series of neuro-immune interactions underlie many of these mechanisms. Imaging studies have shown that such damage is characterized by functional, structural, and chemical changes in the brain. Such pain is maladaptive in the sense that it occurs in the absence of ongoing noxious stimuli and does not promote healing and repair. CONCLUSION As defined, maldynia is a multidimensional process that may warrant consideration as a chronic disease not only affecting sensory and emotional processing but also producing an altered brain state based on both functional imaging and macroscopic measurements. However, the absolute clinical value of this definition is not established.
Collapse
Affiliation(s)
- Barry D Dickinson
- Council on Science and Public Health, American Medical Association, Chicago, Illinois 60654, USA.
| | | | | | | |
Collapse
|
427
|
Gurkiewicz M, Korngreen A, Waxman SG, Lampert A. Kinetic modeling of Nav1.7 provides insight into erythromelalgia-associated F1449V mutation. J Neurophysiol 2011; 105:1546-57. [PMID: 21289137 DOI: 10.1152/jn.00703.2010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Gain-of-function mutations of the voltage-gated sodium channel (VGSC) Na(v)1.7 have been linked to human pain disorders. The mutation F1449V, located at the intracellular end of transmembrane helix S6 of domain III, induces the inherited pain syndrome erythromelalgia. A kinetic model of wild-type (WT) and F1449V Na(v)1.7 may provide a basis for predicting putative intraprotein interactions. We semiautomatically constrained a Markov model using stochastic search algorithms and whole cell patch-clamp recordings from human embryonic kidney cells transfected with Na(v)1.7 and its F1449V mutation. The best models obtained simulated known differences in action potential thresholds and firing patterns in spinal sensory neurons expressing WT and F1449V. The most suitable Markov model consisted of three closed, one open, and two inactivated states. The model predicted that the F1449V mutation shifts occupancy of the closed states closer to the open state, making it easier for the channel pore to open. It also predicted that F1449V's second inactivated state is more than four times more likely to be occupied than the equivalent state in WT at hyperpolarized potentials, although the mutation still lowered the firing threshold of action potentials. The differences between WT and F1449V were not limited to a single transition. Thus a point mutation in position F1449, while phenotypically most probably affecting the activation gate, may also modify channel functions mediated by structures in more distant areas of the channel protein.
Collapse
Affiliation(s)
- Meron Gurkiewicz
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 52900, Israel.
| | | | | | | |
Collapse
|
428
|
Recent Advances Toward Pain Therapeutics. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2011. [DOI: 10.1016/b978-0-12-386009-5.00025-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
429
|
Binshtok AM. Mechanisms of nociceptive transduction and transmission: a machinery for pain sensation and tools for selective analgesia. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2011; 97:143-77. [PMID: 21708310 DOI: 10.1016/b978-0-12-385198-7.00006-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Many surgical and dental procedures depend on use of local anesthetics to reversibly eliminate pain. By the blockade of voltage-gated sodium channels, local anesthetics prevent the transmission of nociceptive information. However, since all local anesthetics act non-selectively on all types of axons they also cause a loss of innocuous sensation, motor paralysis and autonomic block. Thus, approaches that produce only a selective blockade of pain fibers are of great potential clinical importance. In this chapter we will review the recent findings describing mechanisms of pain transduction and transmission and introduce novel therapeutic approaches to produce pain-selective analgesia.
Collapse
Affiliation(s)
- Alexander M Binshtok
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada and Center for Research on Pain, The Hebrew University Medical School, Jerusalem, Israel
| |
Collapse
|
430
|
|
431
|
Saez NJ, Senff S, Jensen JE, Er SY, Herzig V, Rash LD, King GF. Spider-venom peptides as therapeutics. Toxins (Basel) 2010; 2:2851-71. [PMID: 22069579 PMCID: PMC3153181 DOI: 10.3390/toxins2122851] [Citation(s) in RCA: 222] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 12/17/2010] [Accepted: 12/17/2010] [Indexed: 01/01/2023] Open
Abstract
Spiders are the most successful venomous animals and the most abundant terrestrial predators. Their remarkable success is due in large part to their ingenious exploitation of silk and the evolution of pharmacologically complex venoms that ensure rapid subjugation of prey. Most spider venoms are dominated by disulfide-rich peptides that typically have high affinity and specificity for particular subtypes of ion channels and receptors. Spider venoms are conservatively predicted to contain more than 10 million bioactive peptides, making them a valuable resource for drug discovery. Here we review the structure and pharmacology of spider-venom peptides that are being used as leads for the development of therapeutics against a wide range of pathophysiological conditions including cardiovascular disorders, chronic pain, inflammation, and erectile dysfunction.
Collapse
Affiliation(s)
- Natalie J Saez
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, 4072, Australia.
| | | | | | | | | | | | | |
Collapse
|
432
|
Kim TH, Kim HI, Kim J, Park M, Song JH. Effects of minocycline on Na+ currents in rat dorsal root ganglion neurons. Brain Res 2010; 1370:34-42. [PMID: 21081117 DOI: 10.1016/j.brainres.2010.11.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 11/08/2010] [Accepted: 11/09/2010] [Indexed: 10/18/2022]
Abstract
Minocycline is an inhibitor of microglial activation and proliferation. Minocycline suppresses pain-related behaviors in many different pain states, which correlates closely with its inhibition of microglial activation and subsequent release of pro-inflammatory mediators in the spinal cord. Na(+) channels in dorsal root ganglion (DRG) neurons are implicated in the generation of inflammatory and neuropathic pain. To elucidate a possible peripheral mechanism of minocycline analgesia, effects of minocycline on tetrodotoxin-sensitive and tetrodotoxin-resistant Na(+) currents in rat DRG neurons were investigated. Minocycline potently inhibited both types of Na(+) currents with IC(50) values of 350 nM and 410 nM, respectively. The inhibition was accompanied by a depolarizing shift of the activation voltage. However, minocycline slowed the inactivation and speeded up the recovery from inactivation. These results suggest minocycline may exert analgesia peripherally thorough Na(+) channel inhibition in the primary afferent neurons as well as centrally through microglial inhibition in the spinal cord.
Collapse
Affiliation(s)
- Tae Hoon Kim
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul 156-756, Republic of Korea
| | | | | | | | | |
Collapse
|
433
|
Dabby R, Sadeh M, Gilad R, Lampl Y, Cohen S, Inbar S, Leshinsky-Silver E. Chronic non-paroxysmal neuropathic pain - Novel phenotype of mutation in the sodium channel SCN9A gene. J Neurol Sci 2010; 301:90-2. [PMID: 21094958 DOI: 10.1016/j.jns.2010.10.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2010] [Revised: 09/29/2010] [Accepted: 10/04/2010] [Indexed: 12/19/2022]
Abstract
BACKGROUND Gain-of-function mutations in the SCN9A gene (encoding to NaV1.7 voltage-gated sodium channel) cause two rare paroxysmal pain disorders: inherited erythromelalgia (IEM) and paroxysmal extreme pain disorder (PEDP). These phenotypes are characterized by episodic extreme localized pain with cutaneous autonomic signs. So far, no other phenotypes have been associated with mutation in the SCN9A gene. OBJECTIVE To investigate mutations in the SCN9A gene in patients with chronic non-paroxysmal neuropathic pain. PATIENTS 9 patients with chronic severe unexplained neuropathic pain. RESULTS Of the nine patients one had predicted pathologic mutations in the SCN9A gene. This patient had a heterozygous change of n.4648 T-C in exon 27 resulting in a substitution of W1550R, a highly conserved amino acid, predicting damage in the transmembrane S2 region, repeat IV. This mutation was not found in 50 controls. CONCLUSIONS SCN9A mutations cause pain syndromes other than IEM and PEPD. These mutations should be considered in patients with resistant unexplained chronic neuropathic pain.
Collapse
Affiliation(s)
- Ron Dabby
- Department of Neurology, Wolfson Medical Center, Holon, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | | | | | | | | | | | | |
Collapse
|
434
|
Abstract
Mendelian heritable pain disorders have provided insights into human pain mechanisms and suggested new analgesic drug targets. Interestingly, many of the heritable monogenic pain disorders have been mapped to mutations in genes encoding ion channels. Studies in transgenic mice have also implicated many ion channels in damage sensing and pain modulation. It seems likely that aberrant peripheral or central ion channel activity underlies or initiates many pathological pain conditions. Understanding the mechanistic basis of ion channel malfunction in terms of trafficking, localization, biophysics, and consequences for neurotransmission is a potential route to new pain therapies.
Collapse
Affiliation(s)
- Ramin Raouf
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, UK.
| | | | | |
Collapse
|
435
|
Abstract
Inherited episodic neurological disorders are often due to mutations in ion channels or their interacting proteins, termed channelopathies. There are a wide variety of such disorders, from those causing paralysis, to extreme pain, to ataxia. A common theme in these is alteration of action potential properties or synaptic transmission and a resulting increased propensity of the resulting tissue to enter into or stay in an altered excitability state. Manifestations of these disorders are triggered by an array of precipitants, all of which stress the particular affected tissue in some way and aid in propelling its activity into an aberrant state. Study of these disorders has aided in the understanding of disease risk factors and elucidated the cause of clinically related sporadic disorders. The findings from study of these disorders will aid in the diagnosis and efficient targeted treatment of affected patients.
Collapse
Affiliation(s)
- Devon P Ryan
- Neuroscience Graduate Program, University of California-San Francisco, San Francisco, CA 94158, USA
| | | |
Collapse
|
436
|
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: 0.8] [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
|
437
|
Cox JJ, Sheynin J, Shorer Z, Reimann F, Nicholas AK, Zubovic L, Baralle M, Wraige E, Manor E, Levy J, Woods CG, Parvari R. Congenital insensitivity to pain: novel SCN9A missense and in-frame deletion mutations. Hum Mutat 2010; 31:E1670-86. [PMID: 20635406 PMCID: PMC2966863 DOI: 10.1002/humu.21325] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Accepted: 06/29/2010] [Indexed: 11/08/2022]
Abstract
SCN9Aencodes the voltage-gated sodium channel Na(v)1.7, a protein highly expressed in pain-sensing neurons. Mutations in SCN9A cause three human pain disorders: bi-allelic loss of function mutations result in Channelopathy-associated Insensitivity to Pain (CIP), whereas activating mutations cause severe episodic pain in Paroxysmal Extreme Pain Disorder (PEPD) and Primary Erythermalgia (PE). To date, all mutations in SCN9A that cause a complete inability to experience pain are protein truncating and presumably lead to no protein being produced. Here, we describe the identification and functional characterization of two novel non-truncating mutations in families with CIP: a homozygously-inherited missense mutation found in a consanguineous Israeli Bedouin family (Na(v)1.7-R896Q) and a five amino acid in-frame deletion found in a sporadic compound heterozygote (Na(v)1.7-DeltaR1370-L1374). Both of these mutations map to the pore region of the Na(v)1.7 sodium channel. Using transient transfection of PC12 cells we found a significant reduction in membrane localization of the mutant protein compared to the wild type. Furthermore, voltage clamp experiments of mutant-transfected HEK293 cells show a complete loss of function of the sodium channel, consistent with the absence of pain phenotype. In summary, this study has identified critical amino acids needed for the normal subcellular localization and function of Na(v)1.7.
Collapse
Affiliation(s)
- James J Cox
- Department of Medical Genetics, University of CambridgeUK
| | - Jony Sheynin
- Department of Virology and Developmental Genetics, Faculty of Health Sciences, Ben Gurion University of the NegevIsrael
- National Institute of Biotechnology in the Negev, Ben Gurion University of the NegevIsrael
| | - Zamir Shorer
- Division of Pediatrics, Soroka Medical Center and Faculty of Health Sciences, Ben Gurion University of the NegevIsrael
| | - Frank Reimann
- Department of Clinical Biochemistry, University of CambridgeUK
| | | | - Lorena Zubovic
- International Centre for Genetic Engineering and BiotechnologyTrieste, Italy
| | - Marco Baralle
- International Centre for Genetic Engineering and BiotechnologyTrieste, Italy
| | | | - Esther Manor
- Department of Virology and Developmental Genetics, Faculty of Health Sciences, Ben Gurion University of the NegevIsrael
- Institute of Genetics, Soroka Medical Center and Faculty of Health SciencesBeer Sheva, Israel
| | - Jacov Levy
- Division of Pediatrics, Soroka Medical Center and Faculty of Health Sciences, Ben Gurion University of the NegevIsrael
| | | | - Ruti Parvari
- Department of Virology and Developmental Genetics, Faculty of Health Sciences, Ben Gurion University of the NegevIsrael
- National Institute of Biotechnology in the Negev, Ben Gurion University of the NegevIsrael
| |
Collapse
|
438
|
Abstract
In this issue of Neuron, Kremeyer et al. describe a gain-of-function mutation of TRPA1 that produces a painful disorder, familial episodic pain syndrome. This discovery enlarges the list of ion channels that, when mutated, produce pain. The growing universe of "channelopathic pain" presents some interesting overarching concepts and questions.
Collapse
Affiliation(s)
- Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
439
|
Kurban M, Wajid M, Shimomura Y, Christiano AM. A nonsense mutation in the SCN9A gene in congenital insensitivity to pain. Dermatology 2010; 221:179-83. [PMID: 20628234 DOI: 10.1159/000314692] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 04/29/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Congenital insensitivity to pain (CIP) (OMIM 243000) is a rare autosomal-recessive disorder. Clinically, CIP is characterized by insensitivity to all modalities of pain except neuropathic pain, and recurrent injuries frequently go unnoticed. CIP is caused by mutations in the SCN9A gene encoding for the Na1.7 channel. METHODS We analyzed the DNA from members of a consanguineous Pakistani family for mutations in the SCN9A gene through direct sequencing after performing linkage studies. RESULTS We identified a novel missense mutation designated R523X in all affected individuals. A screening assay ruled out the possibility of polymorphism. CONCLUSION We identified a novel mutation in the Na1.7 channel leading to CIP, extending the spectrum of mutations in the Na1.7 channel, and enhancing our understanding of the physiology of pain.
Collapse
Affiliation(s)
- Mazen Kurban
- Department of Dermatology, Columbia University, New York, NY, USA
| | | | | | | |
Collapse
|
440
|
Kremeyer B, Lopera F, Cox JJ, Momin A, Rugiero F, Marsh S, Woods CG, Jones NG, Paterson KJ, Fricker FR, Villegas A, Acosta N, Pineda-Trujillo NG, Ramírez JD, Zea J, Burley MW, Bedoya G, Bennett DL, Wood JN, Ruiz-Linares A. A gain-of-function mutation in TRPA1 causes familial episodic pain syndrome. Neuron 2010; 66:671-80. [PMID: 20547126 PMCID: PMC4769261 DOI: 10.1016/j.neuron.2010.04.030] [Citation(s) in RCA: 339] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2010] [Indexed: 12/12/2022]
Abstract
Human monogenic pain syndromes have provided important insights into the molecular mechanisms that underlie normal and pathological pain states. We describe an autosomal-dominant familial episodic pain syndrome characterized by episodes of debilitating upper body pain, triggered by fasting and physical stress. Linkage and haplotype analysis mapped this phenotype to a 25 cM region on chromosome 8q12-8q13. Candidate gene sequencing identified a point mutation (N855S) in the S4 transmembrane segment of TRPA1, a key sensor for environmental irritants. The mutant channel showed a normal pharmacological profile but altered biophysical properties, with a 5-fold increase in inward current on activation at normal resting potentials. Quantitative sensory testing demonstrated normal baseline sensory thresholds but an enhanced secondary hyperalgesia to punctate stimuli on treatment with mustard oil. TRPA1 antagonists inhibit the mutant channel, promising a useful therapy for this disorder. Our findings provide evidence that variation in the TRPA1 gene can alter pain perception in humans.
Collapse
Affiliation(s)
- Barbara Kremeyer
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, UK
| | - Francisco Lopera
- Grupo de Neurociencias, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - James J. Cox
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
- Department of Medical Genetics, Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge CB2 0XY, UK
| | - Aliakmal Momin
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Francois Rugiero
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Steve Marsh
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - C. Geoffrey Woods
- Department of Medical Genetics, Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge CB2 0XY, UK
| | - Nicholas G. Jones
- Department of Neurorestoration, Wolfson CARD, Hodgkin Building, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Kathryn J. Paterson
- Department of Neurorestoration, Wolfson CARD, Hodgkin Building, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Florence R. Fricker
- Department of Neurorestoration, Wolfson CARD, Hodgkin Building, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Andrés Villegas
- Grupo de Neurociencias, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Natalia Acosta
- Grupo de Neurociencias, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | | | - Juan Diego Ramírez
- Grupo de Neurociencias, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Julián Zea
- Grupo de Neurociencias, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Mari-Wyn Burley
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, UK
| | - Gabriel Bedoya
- Grupo de Mapeo Genético, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - David L.H. Bennett
- Department of Neurorestoration, Wolfson CARD, Hodgkin Building, Guy's Campus, King's College London, London SE1 1UL, UK
| | - John N. Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
- World Class University Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul National University, Korea
| | - Andrés Ruiz-Linares
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, UK
- Grupo de Mapeo Genético, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
441
|
Estacion M, Waxman SG, Dib-Hajj SD. Effects of ranolazine on wild-type and mutant hNav1.7 channels and on DRG neuron excitability. Mol Pain 2010; 6:35. [PMID: 20529343 PMCID: PMC2898769 DOI: 10.1186/1744-8069-6-35] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Accepted: 06/08/2010] [Indexed: 12/19/2022] Open
Abstract
Background A direct role of sodium channels in pain has recently been confirmed by establishing a monogenic link between SCN9A, the gene which encodes sodium channel Nav1.7, and pain disorders in humans, with gain-of-function mutations causing severe pain syndromes, and loss-of-function mutations causing congenital indifference to pain. Expression of sodium channel Nav1.8 in DRG neurons has also been shown to be essential for the manifestation of mutant Nav1.7-induced neuronal hyperexcitability. These findings have confirmed key roles of Nav1.7 and Nav1.8 in pain and identify these channels as novel targets for pain therapeutic development. Ranolazine preferentially blocks cardiac late sodium currents at concentrations that do not significantly reduce peak sodium current. Ranolazine also blocks wild-type Nav1.7 and Nav1.8 channels in a use-dependent manner. However, ranolazine's effects on gain-of-function mutations of Nav1.7 and on DRG neuron excitability have not been investigated. We used voltage- and current-clamp recordings to evaluate the hypothesis that ranolazine may be effective in regulating Nav1.7-induced DRG neuron hyperexcitability. Results We show that ranolazine produces comparable block of peak and ramp currents of wild-type Nav1.7 and mutant Nav1.7 channels linked to Inherited Erythromelalgia and Paroxysmal Extreme Pain Disorder. We also show that ranolazine, at a clinically-relevant concentration, blocks high-frequency firing of DRG neurons expressing wild-type but not mutant channels. Conclusions Our data suggest that ranalozine can attenuate hyperexcitability of DRG neurons over-expressing wild-type Nav1.7 channels, as occurs in acquired neuropathic and inflammatory pain, and thus merits further study as an alternative to existing non-selective sodium channel blockers.
Collapse
Affiliation(s)
- Mark Estacion
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | |
Collapse
|
442
|
Cregg R, Momin A, Rugiero F, Wood JN, Zhao J. Pain channelopathies. J Physiol 2010; 588:1897-904. [PMID: 20142270 PMCID: PMC2901978 DOI: 10.1113/jphysiol.2010.187807] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 02/01/2010] [Indexed: 01/11/2023] Open
Abstract
Pain remains a major clinical challenge, severely afflicting around 6% of the population at any one time. Channelopathies that underlie monogenic human pain syndromes are of great clinical relevance, as cell surface ion channels are tractable drug targets. The recent discovery that loss-of-function mutations in the sodium channel Nav1.7 underlie a recessive pain-free state in otherwise normal people is particularly significant. Deletion of channel-encoding genes in mice has also provided insights into mammalian pain mechanisms. Ion channels expressed by immune system cells (e.g. P2X7) have been shown to play a pivotal role in changing pain thresholds, whilst channels involved in sensory transduction (e.g. TRPV1), the regulation of neuronal excitability (potassium channels), action potential propagation (sodium channels) and neurotransmitter release (calcium channels) have all been shown to be potentially selective analgesic drug targets in some animal pain models. Migraine and visceral pain have also been associated with voltage-gated ion channel mutations. Insights into such channelopathies thus provide us with a number of potential targets to control pain.
Collapse
Affiliation(s)
- Roman Cregg
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | | | | | | | | |
Collapse
|
443
|
Nakayama T, Ogiwara I, Ito K, Kaneda M, Mazaki E, Osaka H, Ohtani H, Inoue Y, Fujiwara T, Uematsu M, Haginoya K, Tsuchiya S, Yamakawa K. Deletions of SCN1A 5′ genomic region with promoter activity in Dravet syndrome. Hum Mutat 2010; 31:820-9. [DOI: 10.1002/humu.21275] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
444
|
Zuliani V, Rivara M, Fantini M, Costantino G. Sodium channel blockers for neuropathic pain. Expert Opin Ther Pat 2010; 20:755-79. [DOI: 10.1517/13543771003774118] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
445
|
Mantegazza M, Curia G, Biagini G, Ragsdale DS, Avoli M. Voltage-gated sodium channels as therapeutic targets in epilepsy and other neurological disorders. Lancet Neurol 2010; 9:413-24. [PMID: 20298965 DOI: 10.1016/s1474-4422(10)70059-4] [Citation(s) in RCA: 335] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Voltage-gated sodium channels (VGSCs) are key mediators of intrinsic neuronal and muscle excitability. Abnormal VGSC activity is central to the pathophysiology of epileptic seizures, and many of the most widely used antiepileptic drugs, including phenytoin, carbamazepine, and lamotrigine, are inhibitors of VGSC function. These antiepileptic drugs might also be efficacious in the treatment of other nervous system disorders, such as migraine, multiple sclerosis, neurodegenerative diseases, and neuropathic pain. In this Review, we summarise the structure and function of VGSCs and their involvement in the pathophysiology of several neurological disorders. We also describe the biophysical and molecular bases for the mechanisms of action of antiepileptic VGSC blockers and discuss the efficacy of these drugs in the treatment of epileptic and non-epileptic disorders. Overall, clinical and experimental data indicate that these drugs are efficacious for a range of diseases, and that the development of drugs with enhanced selectivity for specific VGSC isoforms might be an effective and novel approach for the treatment of several neurological diseases.
Collapse
Affiliation(s)
- Massimo Mantegazza
- Dipartimento di Neurofisiopatologia, Fondazione Istituto Neurologico C Besta, Milano, Italy
| | | | | | | | | |
Collapse
|
446
|
Abstract
Nociception is essential for survival whereas pathological pain is maladaptive and often unresponsive to pharmacotherapy. Voltage-gated sodium channels, Na(v)1.1-Na(v)1.9, are essential for generation and conduction of electrical impulses in excitable cells. Human and animal studies have identified several channels as pivotal for signal transmission along the pain axis, including Na(v)1.3, Na(v)1.7, Na(v)1.8, and Na(v)1.9, with the latter three preferentially expressed in peripheral sensory neurons and Na(v)1.3 being upregulated along pain-signaling pathways after nervous system injuries. Na(v)1.7 is of special interest because it has been linked to a spectrum of inherited human pain disorders. Here we review the contribution of these sodium channel isoforms to pain.
Collapse
Affiliation(s)
- Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | | | | | | |
Collapse
|
447
|
Reimann F, Cox JJ, Belfer I, Diatchenko L, Zaykin DV, McHale DP, Drenth JPH, Dai F, Wheeler J, Sanders F, Wood L, Wu TX, Karppinen J, Nikolajsen L, Männikkö M, Max MB, Kiselycznyk C, Poddar M, te Morsche RH, Smith S, Gibson D, Kelempisioti A, Maixner W, Gribble FM, Woods CG. Pain perception is altered by a nucleotide polymorphism in SCN9A. Proc Natl Acad Sci U S A 2010; 107:5148-53. [PMID: 20212137 PMCID: PMC2841869 DOI: 10.1073/pnas.0913181107] [Citation(s) in RCA: 221] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The gene SCN9A is responsible for three human pain disorders. Nonsense mutations cause a complete absence of pain, whereas activating mutations cause severe episodic pain in paroxysmal extreme pain disorder and primary erythermalgia. This led us to investigate whether single nucleotide polymorphisms (SNPs) in SCN9A were associated with differing pain perception in the general population. We first genotyped 27 SCN9A SNPs in 578 individuals with a radiographic diagnosis of osteoarthritis and a pain score assessment. A significant association was found between pain score and SNP rs6746030; the rarer A allele was associated with increased pain scores compared to the commoner G allele (P = 0.016). This SNP was then further genotyped in 195 pain-assessed people with sciatica, 100 amputees with phantom pain, 179 individuals after lumbar discectomy, and 205 individuals with pancreatitis. The combined P value for increased A allele pain was 0.0001 in the five cohorts tested (1277 people in total). The two alleles of the SNP rs6746030 alter the coding sequence of the sodium channel Nav1.7. Each was separately transfected into HEK293 cells and electrophysiologically assessed by patch-clamping. The two alleles showed a difference in the voltage-dependent slow inactivation (P = 0.042) where the A allele would be predicted to increase Nav1.7 activity. Finally, we genotyped 186 healthy females characterized by their responses to a diverse set of noxious stimuli. The A allele of rs6746030 was associated with an altered pain threshold and the effect mediated through C-fiber activation. We conclude that individuals experience differing amounts of pain, per nociceptive stimulus, on the basis of their SCN9A rs6746030 genotype.
Collapse
Affiliation(s)
| | - James J. Cox
- Department of Medical Genetics, Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge CB2 0XY, United Kingdom
| | - Inna Belfer
- Molecular Epidemiology of Pain Program, Department of Anaesthesiology, University of Pittsburgh, Pittsburgh, PA 15261
| | - Luda Diatchenko
- Center for Neurosensory Disorders, School of Dentistry,University of North Carolina, Chapel Hill, NC 27599-7455
| | - Dmitri V. Zaykin
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Duncan P. McHale
- Pfizer Global Research and Development, Sandwich Laboratories, Sandwich CT13 9NJ, United Kingdom
| | - Joost P. H. Drenth
- Department of Gastroenterology and Hepatology, Radboud University Nijmegen Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Feng Dai
- Molecular Epidemiology of Pain Program, Department of Anaesthesiology, University of Pittsburgh, Pittsburgh, PA 15261
- Departments of Anesthesiology and Biostatistics and Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Jerry Wheeler
- Pfizer Global Research and Development, Sandwich Laboratories, Sandwich CT13 9NJ, United Kingdom
| | - Frances Sanders
- Pfizer Global Research and Development, Sandwich Laboratories, Sandwich CT13 9NJ, United Kingdom
| | - Linda Wood
- Pfizer Global Research and Development, Groton Laboratories, Groton, CT 06340
| | - Tian-Xia Wu
- Center for Information Technology, National Institutes of Health, Bethesda, MD 20892
| | - Jaro Karppinen
- Oulu Center for Cell–Matrix Research, Biocenter and Department of Medical Biochemistry and Molecular Biology, FIN-90014 University of Oulu, Oulu, Finland; and
| | - Lone Nikolajsen
- Musculoskeletal Centre, Finnish Institute of Occupational Health, and Institute of Clinical Sciences, Department of Physical Medicine and Rehabilitation, FI-90220 University of Oulu, Oulu, Finland
| | - Minna Männikkö
- Oulu Center for Cell–Matrix Research, Biocenter and Department of Medical Biochemistry and Molecular Biology, FIN-90014 University of Oulu, Oulu, Finland; and
| | - Mitchell B. Max
- Molecular Epidemiology of Pain Program, Department of Anaesthesiology, University of Pittsburgh, Pittsburgh, PA 15261
| | - Carly Kiselycznyk
- Center for Information Technology, National Institutes of Health, Bethesda, MD 20892
| | - Minakshi Poddar
- Molecular Epidemiology of Pain Program, Department of Anaesthesiology, University of Pittsburgh, Pittsburgh, PA 15261
| | - Rene H.M. te Morsche
- Department of Gastroenterology and Hepatology, Radboud University Nijmegen Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Shad Smith
- Center for Neurosensory Disorders, School of Dentistry,University of North Carolina, Chapel Hill, NC 27599-7455
| | - Dustin Gibson
- Center for Neurosensory Disorders, School of Dentistry,University of North Carolina, Chapel Hill, NC 27599-7455
| | - Anthi Kelempisioti
- Oulu Center for Cell–Matrix Research, Biocenter and Department of Medical Biochemistry and Molecular Biology, FIN-90014 University of Oulu, Oulu, Finland; and
| | - William Maixner
- Center for Neurosensory Disorders, School of Dentistry,University of North Carolina, Chapel Hill, NC 27599-7455
| | | | - C. Geoffrey Woods
- Department of Medical Genetics, Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge CB2 0XY, United Kingdom
| |
Collapse
|
448
|
Abstract
Drugs inhibiting voltage-gated sodium channels have long been used as analgesics, beginning with the use of local anaesthetics for sensory blockade and then with the discovery that Nav-blocking anticonvulsants also have benefit for pain therapy. These drugs were discovered without knowledge of their molecular target, using traditional pharmacological methods, and their clinical utility is limited by relatively narrow therapeutic windows. Until recently, attempts to develop improved inhibitors using modern molecular-targeted screening approaches have met with limited success. However, in the last few years there has been renewed activity following the discovery of human Nav1.7 mutations that cause striking insensitivity to pain. Together with recent advances in the technologies required to prosecute ion channels as drug targets, this has led to significant progress being made. This article reviews these developments and summarises current findings with these emerging new Nav inhibitors, highlighting some of the unanswered questions and the challenges that remain before they can be developed for clinical use.
Collapse
Affiliation(s)
- Jeffrey J Clare
- Cell-Based Assays Group, Millipore Corporation, St Charles, Missouri 63304, USA.
| |
Collapse
|
449
|
Amin A, Klemens C, Verkerk A, Meregalli P, Asghari-Roodsari A, de Bakker J, January C, Wilde A, Tan H. Fever-triggered ventricular arrhythmias in Brugada syndrome and type 2 long-QT syndrome. Neth Heart J 2010; 18:165-9. [PMID: 20390067 PMCID: PMC2848357 DOI: 10.1007/bf03091755] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The risk for lethal ventricular arrhythmias is increased in individuals who carry mutations in genes that encode cardiac ion channels. Loss-of-function mutations in SCN5A, the gene encoding the cardiac sodium channel, are linked to Brugada syndrome (BrS). Arrhythmias in BrS are often preceded by coved-type ST-segment elevation in the right-precordial leads V1 and V2. Loss-of-function mutations in KCNH2, the gene encoding the cardiac ion channel that is responsible for the rapidly activating delayed rectifying potassium current, are linked to long-QT syndrome type 2 (LQT-2). LQT-2 is characterised by delayed cardiac repolarisation and rate-corrected QT interval (QTc) prolongation. Here, we report that the risk for ventricular arrhythmias in BrS and LQT-2 is further increased during fever. Moreover, we demonstrate that fever may aggravate coved-type ST-segment elevation in BrS, and cause QTc lengthening in LQT-2. Finally, we describe molecular mechanisms that may underlie the proarrhythmic effects of fever in BrS and LQT-2. (Neth Heart J 2010;18:165-9.).
Collapse
Affiliation(s)
- A.S. Amin
- Heart Failure Research Centre, Department of Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - C.A. Klemens
- Heart Failure Research Centre, Department of Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, and Interuniversity Cardiology Institute Netherlands
| | - A.O. Verkerk
- Heart Failure Research Centre, Department of Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - P.G. Meregalli
- Department of Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - A. Asghari-Roodsari
- Heart Failure Research Centre, Department of Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, and Interuniversity Cardiology Institute Netherlands
| | - J.M.T. de Bakker
- Heart Failure Research Centre, Department of Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, and Interuniversity Cardiology Institute Netherlands
| | - C.T. January
- Departments of Medicine and Physiology, Cellular and Molecular Arrhythmia Research Program, University of Wisconsin, Madison, Wisconsin, USA
| | - A.A.M. Wilde
- Heart Failure Research Centre, Department of Experimental Cardiology, and Department of Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - H.L. Tan
- Heart Failure Research Centre, Department of Experimental Cardiology, and Department of Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
450
|
Obreja O, Schmelz M. Single-fiber recordings of unmyelinated afferents in pig. Neurosci Lett 2010; 470:175-9. [DOI: 10.1016/j.neulet.2009.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 10/01/2009] [Accepted: 10/02/2009] [Indexed: 11/28/2022]
|