38 results on '"Nav1.7"'
Search Results
2. Intranasal CRMP2-Ubc9 inhibitor regulates NaV1.7 to alleviate trigeminal neuropathic pain.
- Author
-
Loya-Lopez, Santiago I., Allen, Heather N., Duran, Paz, Calderon-Rivera, Aida, Gomez, Kimberly, Kumar, Upasana, Shields, Rory, Zeng, Rui, Dwivedi, Akshat, Saurabh, Saumya, Korczeniewska, Olga A., and Khanna, Rajesh
- Subjects
- *
NEURALGIA , *DORSAL root ganglia , *SENSORY neurons , *SENSORY ganglia , *TRIGEMINAL nerve diseases , *SODIUM channels , *OROFACIAL pain - Abstract
Dysregulation of voltage-gated sodium NaV1.7 channels in sensory neurons contributes to chronic pain conditions, including trigeminal neuropathic pain. We previously reported that chronic pain results in part from increased SUMOylation of collapsin response mediator protein 2 (CRMP2), leading to an increased CRMP2/NaV1.7 interaction and increased functional activity of NaV1.7. Targeting this feed-forward regulation, we developed compound 194, which inhibits CRMP2 SUMOylation mediated by the SUMO-conjugating enzyme Ubc9. We further demonstrated that 194 effectively reduces the functional activity of NaV1.7 channels in dorsal root ganglia neurons and alleviated inflammatory and neuropathic pain. Here, we used a comprehensive array of approaches, encompassing biochemical, pharmacological, genetic, electrophysiological, and behavioral analyses, to assess the functional implications of NaV1.7 regulation by CRMP2 in trigeminal ganglia (TG) neurons. We confirmed the expression of Scn9a, Dpysl2, and UBE2I within TG neurons. Furthermore, we found an interaction between CRMP2 and NaV1.7, with CRMP2 being SUMOylated in these sensory ganglia. Disrupting CRMP2 SUMOylation with compound 194 uncoupled the CRMP2/NaV1.7 interaction, impeded NaV1.7 diffusion on the plasma membrane, and subsequently diminished NaV1.7 activity. Compound 194 also led to a reduction in TG neuron excitability. Finally, when intranasally administered to rats with chronic constriction injury of the infraorbital nerve, 194 significantly decreased nociceptive behaviors. Collectively, our findings underscore the critical role of CRMP2 in regulating NaV1.7 within TG neurons, emphasizing the importance of this indirect modulation in trigeminal neuropathic pain. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Ih current stabilizes excitability in rodent DRG neurons and reverses hyperexcitability in a nociceptive neuron model of inherited neuropathic pain.
- Author
-
Vasylyev, Dmytro V., Liu, Shujun, and Waxman, Stephen G.
- Subjects
- *
DORSAL root ganglia , *ACTION potentials , *HEART cells , *DRUG target , *GENETIC models - Abstract
We show here that hyperpolarization‐activated current (Ih) unexpectedly acts to inhibit the activity of dorsal root ganglion (DRG) neurons expressing WT Nav1.7, the largest inward current and primary driver of DRG neuronal firing, and hyperexcitable DRG neurons expressing a gain‐of‐function Nav1.7 mutation that causes inherited erythromelalgia (IEM), a human genetic model of neuropathic pain. In this study we created a kinetic model of Ih and used it, in combination with dynamic‐clamp, to study Ih function in DRG neurons. We show, for the first time, that Ih increases rheobase and reduces the firing probability in small DRG neurons, and demonstrate that the amplitude of subthreshold oscillations is reduced by Ih. Our results show that Ih, due to slow gating, is not deactivated during action potentials (APs) and has a striking damping action, which reverses from depolarizing to hyperpolarizing, close to the threshold for AP generation. Moreover, we show that Ih reverses the hyperexcitability of DRG neurons expressing a gain‐of‐function Nav1.7 mutation that causes IEM. In the aggregate, our results show that Ih unexpectedly has strikingly different effects in DRG neurons as compared to previously‐ and well‐studied cardiac cells. Within DRG neurons where Nav1.7 is present, Ih reduces depolarizing sodium current inflow due to enhancement of Nav1.7 channel fast inactivation and creates additional damping action by reversal of Ih direction from depolarizing to hyperpolarizing close to the threshold for AP generation. These actions of Ih limit the firing of DRG neurons expressing WT Nav1.7 and reverse the hyperexcitability of DRG neurons expressing a gain‐of‐function Nav1.7 mutation that causes IEM. Key points: Hyperpolarization‐activated cyclic nucleotide‐gated (HCN) channels, the molecular determinants of hyperpolarization‐activated current (Ih) have been characterized as a 'pain pacemaker', and thus considered to be a potential molecular target for pain therapeutics.Dorsal root ganglion (DRG) neurons express Nav1.7, a channel that is not present in central neurons or cardiac tissue. Gain‐of‐function mutations (GOF) of Nav1.7 identified in inherited erythromelalgia (IEM), a human genetic model of neuropathic pain, produce DRG neuron hyperexcitability, which in turn produces severe pain.We found that Ih increases rheobase and reduces firing probability in small DRG neurons expressing WT Nav1.7, and demonstrate that the amplitude of subthreshold oscillations is reduced by Ih.We also demonstrate that Ih reverses the hyperexcitability of DRG neurons expressing a GOF Nav1.7 mutation (L858H) that causes IEM.Our results show that, in contrast to cardiac cells and CNS neurons, Ih acts to stabilize DRG neuron excitability and prevents excessive firing. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
4. Ectopic expression of Nav1.7 in spinal dorsal horn neurons induced by NGF contributes to neuropathic pain in a mouse spinal cord injury model.
- Author
-
Yan Fu, Liting Sun, Fengting Zhu, Wei Xia, Ting Wen, Ruilong Xia, Xin Yu, Dan Xu, and Changgeng Peng
- Subjects
NEURALGIA ,DORSAL root ganglia ,NEURONS ,NEUROTROPHIN receptors ,MICE - Abstract
Neuropathic pain (NP) induced by spinal cord injury (SCI) often causes long-term disturbance for patients, but the mechanisms behind remains unclear. Here, our study showed SCI-induced ectopic expression of Nav1.7 in abundant neurons located in deep and superficial laminae layers of the spinal dorsal horn (SDH) and upregulation of Nav1.7 expression in dorsal root ganglion (DRG) neurons in mice. Pharmacologic studies demonstrated that the efficacy of the blood-brain-barrier (BBB) permeable Nav1.7 inhibitor GNE-0439 for attenuation of NP in SCI mice was significantly better than that of the BBB non-permeable Nav1.7 inhibitor PF-05089771. Moreover, more than 20% of Nav1.7-expressing SDH neurons in SCI mice were activated to express FOS when there were no external stimuli, suggesting that the ectopic expression of Nav1.7 made SDH neurons hypersensitive and Nav1.7-expressing SDH neurons participated in central sensitization and in spontaneous pain and/or walking-evoked mechanical pain. Further investigation showed that NGF, a strong activator of Nav1.7 expression, and its downstream JUN were upregulated after SCI in SDH neurons with similar distribution patterns and in DRG neurons too. In conclusion, our findings showed that the upregulation of Nav1.7 was induced by SCI in both SDH and DRG neurons through increased expression of NGF/JUN, and the inhibition of Nav1.7 in both peripheral and spinal neurons alleviated mechanical pain in SCI mice. These data suggest that BBB permeable Nav1.7 blockers might relieve NP in patients with SCI and that blocking the upregulation of Nav1.7 in the early stage of SCI via selective inhibition of the downstream signaling pathways of NGF or Nav1.7-targeted RNA drugs could be a strategy for therapy of SCI-induced NP. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. Nav1.7、Nav1.8 和交感芽生在大鼠 SNI 模型神经 病理性疼痛形成过程中的作用.
- Author
-
李笑笑, 陈 涵, 李妍妍, 朱玉静, 张 谭, and 唐 俊
- Abstract
Objective To investigate the relationship between Nav1.7, Nav1.8 and sympathetic sprouting in dorsal root ganglion (DRG) during neuropathic pain induced by spared nerve injury (SNI). Methods The experimental rats were randomly divided into Control group (n=18) and SNI group (n= 18). Mechanical withdrawal threshold (MWT) was measured on the 3rd, 7th, 14th and 21st days after surgery. The mRNA expressions and protein contents of Nav1.7, Nav1.8, growth-associated protein 43 (GAP43) and tyrosine hydroxylase (TH) were detected respectively from operative side lumbar DRGs which were extracted on the 7th and 21st days, and immunofluorescence staining were performed concurrently. Results Compared with Control group, MWT in SNI group was reduced time-dependently. On the 7th day and 21st day in the SNI group, mRNA expressions and protein contents of Nav1.7 and Nav1.8 in the DRGs increased synchronously with the mRNA expand protein contents of GAP43 and TH. Immunofluorescence staining showed that large-diameter neurons which expressed Nav1.7, Nav1.8 and GAP43 abnormally increased on the postoperative 21st day. Especially, the positive neurons of Nav1.7 and Nav1.8 respectively were surrounded by TH positive fibers which formed basketlike structures. Conclusion Sympathetic sprouting and basket structure on the medium and large neurons in DRG after sciatic nerve injury were related to the synchronous increasing of Nav1.7 and Nav1.8. This phenomenon may be one of the formation factors of neuropathic pain. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. Reversal of Peripheral Neuropathic Pain by the Small-Molecule Natural Product Narirutin via Block of Na v 1.7 Voltage-Gated Sodium Channel.
- Author
-
Yang, Haoyi, Shan, Zhiming, Guo, Weijie, Wang, Yuwei, Cai, Shuxian, Li, Fuyi, Huang, Qiaojie, Liu, Jessica Aijia, Cheung, Chi Wai, and Cai, Song
- Subjects
- *
SODIUM channels , *NEURALGIA , *SENSORY neurons , *NATURAL products , *NOCICEPTORS , *CALCITONIN gene-related peptide , *DORSAL root ganglia , *DRUG target - Abstract
Neuropathic pain is a refractory chronic disease affecting millions of people worldwide. Given that present painkillers have poor efficacy or severe side effects, developing novel analgesics is badly needed. The multiplex structure of active ingredients isolated from natural products provides a new source for phytochemical compound synthesis. Here, we identified a natural product, Narirutin, a flavonoid compound isolated from the Citrus unshiu, showing antinociceptive effects in rodent models of neuropathic pain. Using calcium imaging, whole-cell electrophysiology, western blotting, and immunofluorescence, we uncovered a molecular target for Narirutin's antinociceptive actions. We found that Narirutin (i) inhibits Veratridine-triggered nociceptor activities in L4-L6 rat dorsal root ganglion (DRG) neurons, (ii) blocks voltage-gated sodium (NaV) channels subtype 1.7 in both small-diameter DRG nociceptive neurons and human embryonic kidney (HEK) 293 cell line, (iii) does not affect tetrodotoxin-resistant (TTX-R) NaV channels, and (iv) blunts the upregulation of Nav1.7 in calcitonin gene-related peptide (CGRP)-labeled DRG sensory neurons after spared nerve injury (SNI) surgery. Identifying Nav1.7 as a molecular target of Narirutin may further clarify the analgesic mechanism of natural flavonoid compounds and provide an optimal idea to produce novel selective and efficient analgesic drugs. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
7. Lidocaine reduces pain behaviors by inhibiting the expression of Nav1.7 and Nav1.8 and diminishing sympathetic sprouting in SNI rats.
- Author
-
Li, Xiaoxiao, Chen, Han, Zhu, Yujing, Li, Yanyan, Zhang, Tan, and Tang, Jun
- Subjects
- *
DORSAL root ganglia , *GERMINATION , *SYMPATHETIC nervous system , *LIDOCAINE , *NEURALGIA - Abstract
Chronic neuropathic pain is a significant clinical challenge, and the mechanisms of neuropathic pain remain elusive. Previous studies have shown that spontaneous potential, which is triggered by Nav1.7 and Nav1.8 in the dorsal root ganglion (DRG), is crucial for the development of inflammatory and neuropathic pain. Functional coupling between the sympathetic nervous system and somatosensory nerves after a nerve injury has also been noted as an important factor in neuropathic pain. However, the relationship of sympathetic sprouting with Nav1.7 and Nav1.8 remains unclear. Therefore, we dynamically examined the mechanical withdrawal threshold (MWT), changes in Nav1.7 and Nav1.8, and sympathetic sprouting after lidocaine treatment in the spared nerve injury (SNI) model of rats. After lidocaine treatment, the MWT obviously increased, showing that hypersensitivity was significantly relieved and the abnormal expression of Nav1.7 and Nav1.8 caused by SNI was also significantly reduced. In addition, lidocaine distinctly inhibited sympathetic nerve sprouting and basket formation around the Nav1.7 and Nav1.8 neurons in the DRG. These results indicate that lidocaine may alleviate neuropathic pain by inhibiting the expression of Nav1.7 and Nav1.8, and diminishing sympathetic sprouting in DRG. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
8. Differential expression of Cdk5-phosphorylated CRMP2 following a spared nerve injury
- Author
-
Aubin Moutal, Yingshi Ji, Shreya Sai Bellampalli, and Rajesh Khanna
- Subjects
Cdk5 ,CRMP2 ,Neuropathic pain ,DRGs ,CaV2,2 ,NaV1.7 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Effective treatment of high-impact pain patients is one of the major stated goals of the National Pain Strategy in the United States. Identification of new targets and mechanisms underlying neuropathic pain will be critical in developing new target-specific medications for better neuropathic pain management. We recently discovered that peripheral nerve injury-induced upregulation of an axonal guidance phosphoprotein collapsin response mediator protein 2 (CRMP2) and the N-type voltage-gated calcium (CaV2.2) as well as the NaV1.7 voltage-gated sodium channel, correlates with the development of neuropathic pain. In our previous studies, we found that interfering with the phosphorylation status of CRMP2 is sufficient to confer protection from chronic pain. Here we examined the expression of CRMP2 and CRMP2 phosphorylated by cyclin-dependent kinase 5 (Cdk5, on serine residue 522 (S522)) in sciatic nerve, nerve terminals of the glabrous skin, and in select subpopulations of DRG neurons in the SNI model of neuropathic pain. By enhancing our understanding of the phosphoregulatory status of CRMP2 within DRG subpopulations, we may be in a better position to design novel pharmacological interventions for chronic pain.
- Published
- 2020
- Full Text
- View/download PDF
9. Non-SUMOylated CRMP2 decreases NaV1.7 currents via the endocytic proteins Numb, Nedd4-2 and Eps15.
- Author
-
Gomez, Kimberly, Ran, Dongzhi, Madura, Cynthia L., Moutal, Aubin, and Khanna, Rajesh
- Subjects
- *
UBIQUITIN ligases , *EPIDERMAL growth factor receptors , *DORSAL root ganglia , *SODIUM channels , *CHRONIC pain , *SENSORY neurons - Abstract
Voltage-gated sodium channels are key players in neuronal excitability and pain signaling. Functional expression of the voltage-gated sodium channel NaV1.7 is under the control of SUMOylated collapsin response mediator protein 2 (CRMP2). When not SUMOylated, CRMP2 forms a complex with the endocytic proteins Numb, the epidermal growth factor receptor pathway substrate 15 (Eps15), and the E3 ubiquitin ligase Nedd4-2 to promote clathrin-mediated endocytosis of NaV1.7. We recently reported that CRMP2 SUMO-null knock-in (CRMP2K374A/K374A) female mice have reduced NaV1.7 membrane localization and currents in their sensory neurons. Preventing CRMP2 SUMOylation was sufficient to reverse mechanical allodynia in CRMP2K374A/K374A female mice with neuropathic pain. Here we report that inhibiting clathrin assembly in nerve-injured male CRMP2K374A/K374A mice precipitated mechanical allodynia in mice otherwise resistant to developing persistent pain. Furthermore, Numb, Nedd4-2 and Eps15 expression was not modified in basal conditions in the dorsal root ganglia (DRG) of male and female CRMP2K374A/K374A mice. Finally, silencing these proteins in DRG neurons from female CRMP2K374A/K374A mice, restored the loss of sodium currents. Our study shows that the endocytic complex composed of Numb, Nedd4-2 and Eps15, is necessary for non-SUMOylated CRMP2-mediated internalization of sodium channels in vivo. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
10. Studies on CRMP2 SUMOylation-deficient transgenic mice identify sex-specific Nav1.7 regulation in the pathogenesis of chronic neuropathic pain.
- Author
-
Moutal, Aubin, Song Cai, Jie Yu, Stratton, Harrison J., Chefdeville, Aude, Gomez, Kimberly, Dongzhi Ran, Madura, Cynthia L., Boinon, Lisa, Soto, Maira, Yuan Zhou, Zhiming Shan, Chew, Lindsey A., Rodgers, Kathleen E., Khanna, Rajesh, Cai, Song, Yu, Jie, Ran, Dongzhi, Zhou, Yuan, and Shan, Zhiming
- Subjects
- *
PROTEINS , *HUMAN reproduction , *CHRONIC pain , *NERVE tissue proteins , *GROWTH factors , *NEURALGIA , *SENSORY receptors , *ANIMAL experimentation , *RATS , *MEMBRANE transport proteins , *RESEARCH funding , *MICE - Abstract
The sodium channel Nav1.7 is a master regulator of nociceptive input into the central nervous system. Mutations in this channel can result in painful conditions and produce insensitivity to pain. Despite being recognized as a "poster child" for nociceptive signaling and human pain, targeting Nav1.7 has not yet produced a clinical drug. Recent work has illuminated the Nav1.7 interactome, offering insights into the regulation of these channels and identifying potentially new druggable targets. Among the regulators of Nav1.7 is the cytosolic collapsin response mediator protein 2 (CRMP2). CRMP2, modified at lysine 374 (K374) by addition of a small ubiquitin-like modifier (SUMO), bound Nav1.7 to regulate its membrane localization and function. Corollary to this, preventing CRMP2 SUMOylation was sufficient to reverse mechanical allodynia in rats with neuropathic pain. Notably, loss of CRMP2 SUMOylation did not compromise other innate functions of CRMP2. To further elucidate the in vivo role of CRMP2 SUMOylation in pain, we generated CRMP2 K374A knock-in (CRMP2) mice in which Lys374 was replaced with Ala. CRMP2 mice had reduced Nav1.7 membrane localization and function in female, but not male, sensory neurons. Behavioral appraisal of CRMP2 mice demonstrated no changes in depressive or repetitive, compulsive-like behaviors and a decrease in noxious thermal sensitivity. No changes were observed in CRMP2 mice to inflammatory, acute, or visceral pain. By contrast, in a neuropathic model, CRMP2 mice failed to develop persistent mechanical allodynia. Our study suggests that CRMP2 SUMOylation-dependent control of peripheral Nav1.7 is a hallmark of chronic, but not physiological, neuropathic pain. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
11. Differential expression of Cdk5-phosphorylated CRMP2 following a spared nerve injury.
- Author
-
Moutal, Aubin, Ji, Yingshi, Bellampalli, Shreya Sai, and Khanna, Rajesh
- Subjects
NERVE endings ,PAIN management ,SCIATIC nerve ,CHRONIC pain ,SODIUM channels ,PERIPHERAL nervous system ,CONOTOXINS - Abstract
Effective treatment of high-impact pain patients is one of the major stated goals of the National Pain Strategy in the United States. Identification of new targets and mechanisms underlying neuropathic pain will be critical in developing new target-specific medications for better neuropathic pain management. We recently discovered that peripheral nerve injury-induced upregulation of an axonal guidance phosphoprotein collapsin response mediator protein 2 (CRMP2) and the N-type voltage-gated calcium (CaV2.2) as well as the NaV1.7 voltage-gated sodium channel, correlates with the development of neuropathic pain. In our previous studies, we found that interfering with the phosphorylation status of CRMP2 is sufficient to confer protection from chronic pain. Here we examined the expression of CRMP2 and CRMP2 phosphorylated by cyclin-dependent kinase 5 (Cdk5, on serine residue 522 (S522)) in sciatic nerve, nerve terminals of the glabrous skin, and in select subpopulations of DRG neurons in the SNI model of neuropathic pain. By enhancing our understanding of the phosphoregulatory status of CRMP2 within DRG subpopulations, we may be in a better position to design novel pharmacological interventions for chronic pain. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
12. Nav1.7 and Nav1.8: Role in the pathophysiology of pain.
- Author
-
Hameed, Shaila
- Subjects
- *
SODIUM channels , *SCIENTIFIC literature , *CHRONIC pain , *PATHOLOGICAL physiology , *PAIN - Abstract
Chronic pain is a significant unmet medical problem. Current research regarding sodium channel function in pathological pain is advancing with the hope that it will enable the development of isoform-specific sodium channel blockers, a promising treatment for chronic pain. Before advancements in the pharmacological field, an elucidation of the roles of Nav1.7 and Nav1.8 in the pathophysiology of pain states is required. Thus, the aim of this report is to present what is currently known about the contributions of these sodium channel subtypes in the pathophysiology of neuropathic and inflammatory pain. The electrophysiological properties and localisation of sodium channel isoforms is discussed. Research concerning the genetic links of Nav1.7 and Nav1.8 in acquired neuropathic and inflammatory pain states from the scientific literature in this field is reported. The role of Nav1.7 and Nav1.8 in the generation and maintenance of abnormal neuronal electrogenesis and hyperexcitability highlights the importance of these channels in the development of pathological pain. However, further research in this area is required to fully elucidate the roles of Nav1.7 and Nav1.8 in the pathophysiology of pain for the development of subtype-specific sodium channel blockers. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
13. Inhibition of Nav1.7 channel by a novel blocker QLS-81 for alleviation of neuropathic pain
- Author
-
Niu, He-ling, Liu, Ya-ni, Xue, Deng-qi, Dong, Li-ying, Liu, Hui-jie, Wang, Jing, Zheng, Yi-lin, Zou, An-ruo, Shao, Li-ming, and Wang, KeWei
- Published
- 2021
- Full Text
- View/download PDF
14. A novel gain-of-function Nav1.7 mutation in a carbamazepine-responsive patient with adult-onset painful peripheral neuropathy.
- Author
-
Adi, Talia, Estacion, Mark, Schulman, Betsy R., Vernino, Steven, Dib-Haj, Sulayman D., and Waxman, Stephen G.
- Subjects
- *
SODIUM channels , *NEURONS , *PERIPHERAL neuropathy , *PAIN , *GENETIC mutation - Abstract
Voltage-gated sodium channel Nav1.7 is a threshold channel in peripheral dorsal root ganglion (DRG), trigeminal ganglion, and sympathetic ganglion neurons. Gain-of-function mutations in Nav1.7 have been shown to increase excitability in DRG neurons and have been linked to rare Mendelian and more common pain disorders. Discovery of Nav1.7 variants in patients with pain disorders may expand the spectrum of painful peripheral neuropathies associated with a well-defined molecular target, thereby providing a basis for more targeted approaches for treatment. We screened the genome of a patient with adult-onset painful peripheral neuropathy characterized by severe burning pain and report here the new Nav1.7-V810M variant. Voltage-clamp recordings were used to assess the effects of the mutation on biophysical properties of Nav1.7 and the response of the mutant channel to treatment with carbamazepine (CBZ), and multi-electrode array (MEA) recordings were used to assess the effects of the mutation on the excitability of neonatal rat pup DRG neurons. The V810M variant increases current density, shifts activation in a hyperpolarizing direction, and slows kinetics of deactivation, all gain-of-function attributes. We also show that DRG neurons that express the V810M variant become hyperexcitable. The patient responded to treatment with CBZ. Although CBZ did not depolarize activation of the mutant channel, it enhanced use-dependent inhibition. Our results demonstrate the presence of a novel gain-of-function variant of Nav1.7 in a patient with adult-onset painful peripheral neuropathy and the responsiveness of that patient to treatment with CBZ, which is likely due to the classical mechanism of use-dependent inhibition. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
15. Inhibition of the Ubc9 E2 SUMO-conjugating enzyme-CRMP2 interaction decreases NaV1.7 currents and reverses experimental neuropathic pain.
- Author
-
François-Moutal, Liberty, Dustrude, Erik T., Yue Wang, Brustovetsky, Tatiana, Dorame, Angie, Weina Ju, Moutal, Aubin, Perez-Miller, Samantha, Brustovetsky, Nickolay, Gokhale, Vijay, Khanna, May, Khanna, Rajesh, Wang, Yue, and Ju, Weina
- Subjects
- *
UBIQUITIN , *MEMBRANE proteins , *PAIN management , *SODIUM channels , *ION channels , *T cells , *SENSORY neurons , *SODIUM metabolism , *ENZYME metabolism , *ANIMAL experimentation , *ANIMALS , *BIOLOGICAL models , *CELL culture , *CYTOLOGY , *ENZYMES , *SENSORY ganglia , *GENES , *GENETICS , *GROWTH factors , *HYPERALGESIA , *NERVE tissue proteins , *NEURALGIA , *RATS , *RESEARCH funding , *SENSORY receptors , *PRECIPITIN tests , *MEMBRANE transport proteins - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
16. Synthesis and Analgesic Effects of μ-TRTX-Hhn1b on Models of Inflammatory and Neuropathic Pain
- Author
-
Yu Liu, Jianguang Tang, Yunxiao Zhang, Xiaohong Xun, Dongfang Tang, Dezheng Peng, Jianming Yi, Zhonghua Liu, and Xiaoliu Shi
- Subjects
μ-TRTX-Hhn1b ,Nav1.7 ,analgesic ,inflammatory pain ,neuropathic pain ,Medicine - Abstract
μ-TRTX-Hhn1b (HNTX-IV) is a 35-amino acid peptide isolated from the venom of the spider, Ornithoctonus hainana. It inhibits voltage-gated sodium channel Nav1.7, which has been considered as a therapeutic target for pain. The goal of the present study is to elucidate the analgesic effects of synthetic μ-TRTX-Hhn1b on animal models of pain. The peptide was first synthesized and then successfully refolded/oxidized. The synthetic peptide had the same inhibitory effect on human Nav1.7 current transiently expressed in HEK 293 cells as the native toxin. Furthermore, the analgesic potentials of the synthetic peptide were examined on models of inflammatory pain and neuropathic pain. μ-TRTX-Hhn1b produced an efficient reversal of acute nociceptive pain in the abdominal constriction model, and significantly reduced the pain scores over the 40-min period in the formalin model. The efficiency of μ-TRTX-Hhn1b on both models was equivalent to that of morphine. In the spinal nerve model, the reversal effect of μ-TRTX-Hhn1b on allodynia was longer and higher than mexiletine. These results demonstrated that μ-TRTX-Hhn1b efficiently alleviated acute inflammatory pain and chronic neuropathic pain in animals and provided an attractive template for further clinical analgesic drug design.
- Published
- 2014
- Full Text
- View/download PDF
17. Correlative increasing expressions of KIF5b and Nav1.7 in DRG neurons of rats under neuropathic pain conditions.
- Author
-
Yin, Jun-Bin, Liu, Hai-Xia, Dong, Qin-Qin, Wu, Huang-Hui, Liang, Zhuo-Wen, Fu, Jin-Tao, Zhao, Wen-Jun, Hu, Huai-Qiang, Guo, Hong-Wei, Zhang, Ting, Lu, Ya-Cheng, Jin, Shan, Wang, Xiao-Ling, Cao, Bing-Zhen, Wang, Zhe, and Ding, Tan
- Subjects
- *
NEURALGIA , *NEURITIS , *DORSAL root ganglia , *PERIPHERAL nerve injuries , *NERVOUS system injuries , *PAIN - Abstract
• KIF5b, one isoform of kinesin-1, was necessary for the membrane localizations of Nav1.7 in DRG neurons. • KIF5b interacted with Nav1.7 and mediated the trafficking of Nav1.7. • Intrathecal injections of KIF5b shRNA moderated the SNI-induced both mechanical and thermal hyperalgesia. • The rescued analgesic effects of KIF5b shRNA also alleviated SNI-induced negative emotional behaviors. Nav1.7, one of tetrodotoxin-sensitive voltage-gated sodium channels, mainly expressed in the small diameter dorsal root ganglion (DRG) neurons. The expression and accumulation on neuronal membrane of Nav1.7 increased following peripheral tissue inflammation or nerve injury. However, the mechanisms for membrane accumulation of Nav1.7 remained unclear. We report that KIF5b, a highly expressed member of the kinesin-1 family in DRGs, promoted the translocation of Nav1.7 to the plasma membrane in DRG neurons of the rat. Following nociceptive behaviors in rats induced by peripheral spared nerve injury (SNI), synchronously increased KIF5b and Nav1.7 expressions were observed in DRGs. Immunohistochemistry staining demonstrated the co-expressions of KIF5b and Nav1.7 in the same DRG neurons. Immunoprecipitation experiments further confirmed the interactions between KIF5b and Nav1.7. Moreover, intrathecal injections of KIF5b shRNA moderated the SNI-induced both mechanical and thermal hyperalgesia. The rescued analgesic effects also alleviated SNI-induced anxiety-like behaviors. In sum, KIF5b was required for the membrane localizations of Nav1.7, which suggests a novel mechanism for the trafficking of Nav1.7 involved in neuropathic pain. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
18. Non-SUMOylated CRMP2 decreases NaV1.7 currents via the endocytic proteins Numb, Nedd4-2 and Eps15
- Author
-
Rajesh Khanna, Dongzhi Ran, Kimberly Gomez, Aubin Moutal, and Cynthia L. Madura
- Subjects
Male ,0301 basic medicine ,Nedd4 Ubiquitin Protein Ligases ,Endocytic cycle ,SUMO protein ,Neuropathic pain ,lcsh:RC346-429 ,Mice ,0302 clinical medicine ,Ganglia, Spinal ,Internalization ,media_common ,Sulfonamides ,biology ,Chemistry ,NAV1.7 Voltage-Gated Sodium Channel ,Eps15 ,Endocytosis ,Ubiquitin ligase ,Cell biology ,NaV1.7 ,Numb ,Hyperalgesia ,Intercellular Signaling Peptides and Proteins ,Thiazolidines ,Female ,Collapsin response mediator protein family ,Ion Channel Gating ,media_common.quotation_subject ,Nerve Tissue Proteins ,Models, Biological ,Clathrin ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Animals ,Gene Silencing ,Molecular Biology ,lcsh:Neurology. Diseases of the nervous system ,Adaptor Proteins, Signal Transducing ,Research ,Sodium channel ,Membrane Proteins ,Sumoylation ,Spinal Nerves ,030104 developmental biology ,CRMP2 ,NUMB ,biology.protein ,Nedd4-2 ,030217 neurology & neurosurgery - Abstract
Voltage-gated sodium channels are key players in neuronal excitability and pain signaling. Functional expression of the voltage-gated sodium channel NaV1.7 is under the control of SUMOylated collapsin response mediator protein 2 (CRMP2). When not SUMOylated, CRMP2 forms a complex with the endocytic proteins Numb, the epidermal growth factor receptor pathway substrate 15 (Eps15), and the E3 ubiquitin ligase Nedd4-2 to promote clathrin-mediated endocytosis of NaV1.7. We recently reported that CRMP2 SUMO-null knock-in (CRMP2K374A/K374A) female mice have reduced NaV1.7 membrane localization and currents in their sensory neurons. Preventing CRMP2 SUMOylation was sufficient to reverse mechanical allodynia in CRMP2K374A/K374A female mice with neuropathic pain. Here we report that inhibiting clathrin assembly in nerve-injured male CRMP2K374A/K374A mice precipitated mechanical allodynia in mice otherwise resistant to developing persistent pain. Furthermore, Numb, Nedd4-2 and Eps15 expression was not modified in basal conditions in the dorsal root ganglia (DRG) of male and female CRMP2K374A/K374A mice. Finally, silencing these proteins in DRG neurons from female CRMP2K374A/K374A mice, restored the loss of sodium currents. Our study shows that the endocytic complex composed of Numb, Nedd4-2 and Eps15, is necessary for non-SUMOylated CRMP2-mediated internalization of sodium channels in vivo.
- Published
- 2021
19. Upregulation of Nav1.7 by endogenous hydrogen sulfide contributes to maintenance of neuropathic pain
- Author
-
Jun-Jie Tian, Wen-Yan Shi, Ying Zhou, Jun-Qiang Si, Zu-Wei Qu, Qin-Yi Chen, Ke-Tao Ma, Li Li, Meng Zhang, and Chao-Yang Tan
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,SNi ,dorsal root ganglion ,hydrogen sulfide ,Endogeny ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Dorsal root ganglion ,Downregulation and upregulation ,Internal medicine ,Genetics ,medicine ,Animals ,Extracellular Signal-Regulated MAP Kinases ,Nav1.7 ,neuropathic pain ,Mitogen-Activated Protein Kinase Kinases ,cystathionine β-synthetase ,biology ,Chemistry ,NAV1.7 Voltage-Gated Sodium Channel ,General Medicine ,Articles ,Nerve injury ,Cystathionine beta synthase ,Rats ,Up-Regulation ,030104 developmental biology ,Rheobase ,Endocrinology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Neuropathic pain ,biology.protein ,Neuralgia ,medicine.symptom ,Signal Transduction - Abstract
Nav1.7 is closely associated with neuropathic pain. Hydrogen sulfide (H2S) has recently been reported to be involved in numerous biological functions, and it has been shown that H2S can enhance the sodium current density, and inhibiting the endogenous production of H2S mediated by cystathionine β-synthetase (CBS) using O-(carboxymethyl) hydroxylamine hemihydrochloride (AOAA) can significantly reduce the expression of Nav1.7 and thus the sodium current density in rat dorsal root ganglion (DRG) neurons. In the present study, it was shown that the fluorescence intensity of H2S was increased in a spared nerve injury (SNI) model and AOAA inhibited this increase. Nav1.7 is expressed in DRG neurons, and the expression of CBS and Nav1.7 were increased in DRG neurons 7, 14 and 21 days post-operation. AOAA inhibited the increase in the expression of CBS, phosphorylated (p)-MEK1/2, p-ERK1/2 and Nav1.7 induced by SNI, and U0126 (a MEK blocker) was able to inhibit the increase in p-MEK1/2, p-ERK1/2 and Nav1.7 expression. However, PF-04856264 did not inhibit the increase in CBS, p-MEK1/2, p-ERK1/2 or Nav1.7 expression induced by SNI surgery. The current density of Nav1.7 was significantly increased in the SNI model and administration of AOAA and U0126 both significantly decreased the density. In addition, AOAA, U0126 and PF-04856264 inhibited the decrease in rheobase, and the increase in action potential induced by SNI in DRG neurons. There was no significant difference in thermal withdrawal latency among each group. However, the time the animals spent with their paw lifted increased significantly following SNI, and the time the animals spent with their paw lifted decreased significantly following the administration of AOAA, U0126 and PF-04856264. In conclusion, these data show that Nav1.7 expression in DRG neurons is upregulated by CBS-derived endogenous H2S in an SNI model, contributing to the maintenance of neuropathic pain.
- Published
- 2020
20. Activating Sirt1 by resveratrol suppresses Nav1.7 expression in DRG through miR-182 and alleviates neuropathic pain in rats
- Author
-
Xiaochun Yang, Wenze Dong, Qianqian Jia, and Liwei Zhang
- Subjects
Male ,0301 basic medicine ,animal structures ,Biophysics ,resveratrol ,Resveratrol ,Pharmacology ,Neuropathic pain ,Intrathecal ,Biochemistry ,Mechanical Allodynia ,Rats, Sprague-Dawley ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Sirtuin 1 ,Western blot ,Ganglia, Spinal ,medicine ,Animals ,Nav1.7 ,Sirt1 ,medicine.diagnostic_test ,business.industry ,NAV1.7 Voltage-Gated Sodium Channel ,Withdrawal latency ,Rats ,nervous system diseases ,Disease Models, Animal ,MicroRNAs ,030104 developmental biology ,chemistry ,miR-182 ,NAV1 ,Neuralgia ,Sciatic nerve ,business ,psychological phenomena and processes ,030217 neurology & neurosurgery ,Research Paper - Abstract
Neuropathic pain is clinically unsatisfactorily treated because of unclear mechanisms. The present study aims to explore the concrete mechanisms underlying the alleviation of resveratrol-activated silent information regulator 1 (Sirt1) to chronic constriction injury (CCI)-induced neuropathic pain. CCI surgery was conducted to the unilateral sciatic nerve of male Sprague-Dawley rats to induce neuropathic pain experimentally. Resveratrol with or without miR-182 antagomir were administered to CCI rats via intrathecal catheter. Behavioral tests including paw withdrawal threshold (PWT) and paw withdrawal latency (PWL) were conducted to explore mechanical allodynia and thermal hyperalgesia. Western blot, qRT-PCR were used to detect the expression levels of Sirt1, miR-182, and Nav1.7 in CCI dorsal root ganglions (DRGs). CCI rats displayed lower PWT and PWL compared with the sham control. Also, the CCI DRGs displayed lower Sirt1 and miR-182 expression as well as higher Nav1.7 expression, which would be almost reversed by resveratrol treatment for 4 successive days. We also found that miR-182 expression inhibition erased the analgesia effect of resveratrol to CCI–induced neuropathic pain possibly through upregulating Nav1.7 expression. In summary, resveratrol alleviated CCI–induced neuropathic pain, possibly through activating Sirt1 to suppress Nav1.7 expression via upregulating miR-182 expression in CCI DRGs.
- Published
- 2020
- Full Text
- View/download PDF
21. Synthesis and Evaluation of (4-Chlorobenzhydryl) Piperazine Amides as Sodium Channel Nav1.7 Inhibitors.
- Author
-
Back, Seung Keun, Kam, Yoo Lim, Oh, Jung Ae, Na, Heung Sik, Ih, Uhtaek, and Park Choo, Hea-Young
- Subjects
- *
PIPERAZINE , *SODIUM channels , *DRUG development , *ANTICONVULSANTS , *FORMALDEHYDE , *CHRONIC pain treatment - Abstract
Blockage of voltage-gated sodium channels is used to treat neuropathic pain which is chronic and can become debilitating. Sodium channels Nav1.7-1.9 are especially attractive targets for drug discovery because of the broad therapeutic potential of their modulation. For a neuropathic pain therapy, anticonvulsant like lamotrigine, carbamazepine and a topical anesthetic such as Lidocaine are used. A growing number of clinical reports suggest that selective inhibitors of Nav1.7 are likely to be the powerful analgesics for treating a broad range of pain conditions. Therefore we evaluated 108 amide derivatives synthesized on human Nav1.7 ( hNav1.7) by VIPR (voltage/ion probe reader), a fluorescence image plate reader ( FLIPR) assay that used voltage-sensor fluorescence dye and stable HEK-293 cell lines expressing hNaV1.7. Ten compounds demonstrated inhibitory activity, and the two most active compounds ( 5 and 6) had IC50 values of 8-10 μM. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
22. Pain behavior in SCN9A (Nav1.7) and SCN10A (Nav1.8) mutant rodent models
- Author
-
Celeste Chidiac, Yann Herault, Yaping Xue, Claire Gaveriaux-Ruff, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA), Herault, Yann, and Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,SCN10A ,Nociception ,[SDV.GEN] Life Sciences [q-bio]/Genetics ,medicine.disease_cause ,Bioinformatics ,Neuropathic pain ,Mice ,0302 clinical medicine ,Loss of Function Mutation ,Medicine ,Nav1.7 ,ComputingMilieux_MISCELLANEOUS ,Nav1.8 ,Mice, Knockout ,Voltage-Gated Sodium Channel Blockers ,Mutation ,Analgesics ,SCN9A ,General Neuroscience ,NAV1.7 Voltage-Gated Sodium Channel ,medicine.anatomical_structure ,Peripheral nervous system ,Gain of Function Mutation ,Rats, Transgenic ,Life Sciences & Biomedicine ,Rodent model ,Knockout ,Small Fiber Neuropathy ,Analgesic ,Pain ,NAV1.8 Voltage-Gated Sodium Channel ,03 medical and health sciences ,Noxious stimulus ,Animals ,Humans ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,Science & Technology ,Mechanism (biology) ,business.industry ,Sodium channel ,[SCCO.NEUR]Cognitive science/Neuroscience ,[SCCO.NEUR] Cognitive science/Neuroscience ,Neurosciences ,Mutant ,Pain behavior ,Voltage-Gated Sodium Channel Agonists ,Rats ,Disease Models, Animal ,030104 developmental biology ,NAV1 ,Neurosciences & Neurology ,business ,030217 neurology & neurosurgery - Abstract
The two voltage gated sodium channels Nav1.7 and Nav1.8 are expressed in the peripheral nervous system and involved in various pain conditions including inflammatory and neuropathic pain. Rodent models bearing deletions or mutations of the corresponding genes, Scn9a and Scn10a, were created in order to understand the role of these channels in the pathophysiological mechanism underlying pain symptoms. This review summarizes the pain behavior profiles reported in Scn9a and Scn10a rodent models. The complete loss-of-function or knockout (KO) of Scn9a or Scn10a and the conditional KO (cKO) of Scn9a in specific cell populations were shown to decrease sensitivity to various pain stimuli. The Possum mutant mice bearing a dominant hypermorphic mutation in Scn10a revealed higher sensitivity to noxious stimuli. Several gain-of-function mutations were identified in patients with painful small fiber neuropathy. Future knowledge obtained from preclinical models bearing these mutations will allow understanding how these mutations affect pain. In addition, the review gives perspectives for creating models that better mimic patients' pain symptoms in view to developing novel analgesic strategies. ispartof: NEUROSCIENCE LETTERS vol:753 ispartof: location:Ireland status: published
- Published
- 2021
- Full Text
- View/download PDF
23. Chemical shift perturbation mapping of the Ubc9-CRMP2 interface identifies a pocket in CRMP2 amenable for allosteric modulation of Nav1.7 channels
- Author
-
Liberty François-Moutal, May Khanna, Vijay Gokhale, David D. Scott, Samantha Perez-Miller, and Rajesh Khanna
- Subjects
0301 basic medicine ,SNi ,Allosteric regulation ,Biophysics ,SUMO protein ,Microscale thermophoresis ,Nerve Tissue Proteins ,Biochemistry ,03 medical and health sciences ,Allosteric Regulation ,Ubc9 ,medicine ,Humans ,Protein Interaction Maps ,Chemistry ,Drug discovery ,Sodium channel ,HSQC-NMR ,NAV1.7 Voltage-Gated Sodium Channel ,Chronic pain ,medicine.disease ,SUMOylation ,Cell biology ,NaV1.7 ,030104 developmental biology ,CRMP2 ,Neuropathic pain ,Ubiquitin-Conjugating Enzymes ,Intercellular Signaling Peptides and Proteins ,Collapsin response mediator protein family ,Research Paper - Abstract
Drug discovery campaigns directly targeting the voltage-gated sodium channel NaV1.7, a highly prized target in chronic pain, have not yet been clinically successful. In a differentiated approach, we demonstrated allosteric control of trafficking and activity of NaV1.7 by prevention of SUMOylation of collapsin response mediator protein 2 (CRMP2). Spinal administration of a SUMOylation incompetent CRMP2 (CRMP2 K374A) significantly attenuated pain behavior in the spared nerve injury (SNI) model of neuropathic pain, underscoring the importance of SUMOylation of CRMP2 as a pathologic event in chronic pain. Using a rational design strategy, we identified a heptamer peptide harboring CRMP2’s SUMO motif that disrupted the CRMP2-Ubc9 interaction, inhibited CRMP2 SUMOylation, inhibited NaV1.7 membrane trafficking, and specifically inhibited NaV1.7 sodium influx in sensory neurons. Importantly, this peptide reversed nerve injury-induced thermal and mechanical hypersensitivity in the SNI model, supporting the practicality of discovering pain drugs by indirectly targeting NaV1.7 via prevention of CRMP2 SUMOylation. Here, our goal was to map the unique interface between CRMP2 and Ubc9, the E2 SUMO conjugating enzyme. Using computational and biophysical approaches, we demonstrate the enzyme/substrate nature of Ubc9/CRMP2 binding and identify hot spots on CRMP2 that may form the basis of future drug discovery campaigns disrupting the CRMP2-Ubc9 interaction to recapitulate allosteric regulation of NaV1.7 for pain relief.
- Published
- 2018
24. Recent developments regarding voltage-gated sodium channel blockers for the treatment of inherited and acquired neuropathic pain syndromes
- Author
-
Jonathan W. Theile and Theodore R Cummins
- Subjects
TRPV1 ,neuropathic pain ,Nav1.7 ,Nav1.8 ,resurgent currents ,voltage-gated sodium channel ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Chronic and neuropathic pain constitute significant health problems affecting millions of individuals each year. Pain sensations typically originate in sensory neurons of the peripheral nervous system (PNS) which relay information to the central nervous system (CNS). Pathological pain sensations can arise as result of changes in excitability of these peripheral sensory neurons. Voltage-gated sodium channels are key determinants regulating action-potential generation and propagation; thus, changes in sodium channel function can have profound effects on neuronal excitability and pain signaling. At present, most of the clinically available sodium channel blockers used to treat pain are non-selective across sodium channel isoforms and can contribute to cardio-toxicity, motor impairments and CNS side effects. Numerous strides have been made over the last decade in an effort to develop more selective and efficacious sodium channel blockers to treat pain. The purpose of this review is to highlight some of the more recent developments put forth by research universities and pharmaceutical companies alike in the pursuit of developing more targeted sodium channel therapies for the treatment of a variety of neuropathic pain conditions.
- Published
- 2011
- Full Text
- View/download PDF
25. New and Developing Drugs for the Treatment of Neuropathic Pain in Diabetes.
- Author
-
Freeman, Roy
- Abstract
A number of agents from diverse pharmacological classes are used to treat neuropathic pain associated with diabetic peripheral neuropathy. Only three of these have regulatory approval for this indication in the U.S. In this focused article, I will discuss selected drugs, newly approved or in development, to treat neuropathic pain in patients with diabetic neuropathy. These will include agonists and antagonists of the transient receptor potential channels, a family of receptor proteins that play a role in the transduction of physical stress; sodium channel isoform specific antagonists; a recently approved dual-action opioid receptor agonist-norepinephrine reuptake inhibitor; gene therapy for neuropathic pain; and anti-nerve growth factor molecules. Mechanisms of action, preclinical supporting data, clinical trial evidence, and adverse effects will be reviewed. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
26. Novel Mutations Mapping to the Fourth Sodium Channel Domain of Nav1.7 Result in Variable Clinical Manifestations of Primary Erythromelalgia.
- Author
-
Cregg, Roman, Laguda, Bisola, Werdehausen, Robert, Cox, James, Linley, John, Ramirez, Juan, Bodi, Istvan, Markiewicz, Michael, Howell, Kevin, Chen, Ya-Chun, Agnew, Karen, Houlden, Henry, Lunn, Michael, Bennett, David, Wood, John, and Kinali, Maria
- Abstract
We identified and clinically investigated two patients with primary erythromelalgia mutations (PEM), which are the first reported to map to the fourth domain of Nav1.7 ( DIV). The identified mutations (A1746G and W1538R) were cloned and transfected to cell cultures followed by electrophysiological analysis in whole-cell configuration. The investigated patients presented with PEM, while age of onset was very different (3 vs. 61 years of age). Electrophysiological characterization revealed that the early onset A1746G mutation leads to a marked hyperpolarizing shift in voltage dependence of steady-state activation, larger window currents, faster activation kinetics (time-to-peak current) and recovery from steady-state inactivation compared to wild-type Nav1.7, indicating a pronounced gain-of-function. Furthermore, we found a hyperpolarizing shift in voltage dependence of slow inactivation, which is another feature commonly found in Nav1.7 mutations associated with PEM. In silico neuron simulation revealed reduced firing thresholds and increased repetitive firing, both indicating hyperexcitability. The late-onset W1538R mutation also revealed gain-of-function properties, although to a lesser extent. Our findings demonstrate that mutations encoding for DIV of Nav1.7 can not only be linked to congenital insensitivity to pain or paroxysmal extreme pain disorder but can also be causative of PEM, if voltage dependency of channel activation is affected. This supports the view that the degree of biophysical property changes caused by a mutation may have an impact on age of clinical manifestation of PEM. In summary, these findings extent the genotype-phenotype correlation profile for SCN9A and highlight a new region of Nav1.7 that is implicated in PEM. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
27. Neuronal expression of the ubiquitin ligase Nedd4-2 in rat dorsal root ganglia: Modulation in the spared nerve injury model of neuropathic pain
- Author
-
Cachemaille, M., Laedermann, C.J., Pertin, M., Abriel, H., Gosselin, R.-D., and Decosterd, I.
- Subjects
- *
GENE expression , *UBIQUITIN ligases , *GANGLIA , *PAIN perception , *NEURONS , *NERVOUS system injuries - Abstract
Abstract: Neuronal hyperexcitability following peripheral nerve lesions may stem from altered activity of voltage-gated sodium channels (VGSCs), which gives rise to allodynia or hyperalgesia. In vitro, the ubiquitin ligase Nedd4-2 is a negative regulator of VGSC α-subunits (Nav), in particular Nav1.7, a key actor in nociceptor excitability. We therefore studied Nedd4-2 in rat nociceptors, its co-expression with Nav1.7 and Nav1.8, and its regulation in pathology. Adult rats were submitted to the spared nerve injury (SNI) model of neuropathic pain or injected with complete Freund’s adjuvant (CFA), a model of inflammatory pain. L4 dorsal root ganglia (DRG) were analyzed in sham-operated animals, seven days after SNI and 48h after CFA with immunofluorescence and Western blot. We observed Nedd4-2 expression in almost 50% of DRG neurons, mostly small and medium-sized. A preponderant localization is found in the non-peptidergic sub-population. Additionally, 55.7±2.7% and 55.0±3.6% of Nedd4-2-positive cells are co-labeled with Nav1.7 and Nav1.8 respectively. SNI significantly decreases the proportion of Nedd4-2-positive neurons from 45.9±1.9% to 33.5±0.7% (p <0.01) and the total Nedd4-2 protein to 44%±0.13% of its basal level (p <0.01, n =4 animals in each group, mean±SEM). In contrast, no change in Nedd4-2 was found after peripheral inflammation induced by CFA. These results indicate that Nedd4-2 is present in nociceptive neurons, is downregulated after peripheral nerve injury, and might therefore contribute to the dysregulation of Navs involved in the hyperexcitability associated with peripheral nerve injuries. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
28. NaV1.7 accumulates and co-localizes with phosphorylated ERK1/2 within transected axons in early experimental neuromas
- Author
-
Persson, Anna-Karin, Gasser, Andreas, Black, Joel A., and Waxman, Stephen G.
- Subjects
- *
PHOSPHORYLATION , *AXONS , *NEUROMAS , *MITOGEN-activated protein kinases , *SODIUM channels , *CHRONIC pain , *SCIATIC nerve - Abstract
Abstract: Peripheral nerve injury can result in formation of a neuroma, which is often associated with heightened sensitivity to normally innocuous stimuli as well as spontaneous dysesthesia and pain. The onset and persistence of neuropathic pain have been linked to spontaneous ectopic electrogenesis in axons within neuromas, suggesting an involvement of voltage-gated sodium channels. Sodium channel isoforms NaV1.3, NaV1.7 and NaV1.8 have been shown to accumulate in chronic painful human neuromas, while, to date, only NaV1.3 has been reported to accumulate within experimental neuromas. Although recent evidence strongly support a major contribution for NaV1.7 in nociception, the expression of NaV1.7 in injured axons within acute neuromas has not been studied. The current study examined whether NaV1.7 accumulates in experimental rat neuromas. We further investigated whether activated (phosphorylated) mitogen-activated protein (MAP) kinase ERK1/2, which is known to modulate NaV1.7 properties, is co-localized with NaV1.7 within axons in neuromas. We demonstrate increased levels of NaV1.7 in experimental rat sciatic nerve neuromas, 2weeks after nerve ligation and transaction. We further show elevated levels of phosphorylated ERK1/2 within individual neuroma axons that exhibit NaV1.7 accumulation. These results extend previous descriptions of sodium channel and MAP kinase accumulation within experimental and human neuromas, and suggest that targeted blockade of NaV1.7 or ERK1/2 may provide a strategy for amelioration of chronic pain that often follows nerve injury and formation of neuromas. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
29. The Roles of Sodium Channels in Nociception: Implications for Mechanisms of Neuropathic Pain.
- Author
-
Liu, Min and Wood, John N.
- Subjects
- *
PAIN management , *ANIMALS , *BIOLOGICAL models , *CELL physiology , *CHRONIC pain , *GENETICS , *GENOMES , *PAIN , *SENSES , *SODIUM ,PERIPHERAL neuropathy diagnosis - Abstract
Animal models have provided useful insights into the development and treatment of neuropathic pain. New genetic data from both human studies and transgenic mouse models suggest that specific voltage-gated sodium channel subtypes are associated with specific types of pain and, as such, may be useful analgesic drug targets for a variety of pain types including neuropathic pain. Global voltagegated sodium channel blockers such as lidocaine have proven efficacy in treating pain but can be limited by adverse effects when administered systemically. Selective sodium channel blockers targeting channels at the periphery (Nav1.7, Nav1.8, and Nav1.9) could potentially reduce the side effect profile. Individual isoforms of voltage-gated sodium channels have been linked to particular types of pain. Nav1.7 is a useful target for ameliorating acute mechanical pain and inflammatory pain, and strong evidence also suggests that Nav1.9 could be targeted for treating inflammatory pain. Selective blockers of Nav1.8 could also have clinical benefit for visceral pain. Although there is no association between a single sodium channel isoform and neuropathic pain, combined blockade of peripherally expressed isoforms Nav1.7, Nav1.8, and Nav1.9 may prove useful. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
30. Benzazepinone Nav1.7 blockers: Potential treatments for neuropathic pain
- Author
-
Hoyt, Scott B., London, Clare, Ok, Hyun, Gonzalez, Edward, Duffy, Joseph L., Abbadie, Catherine, Dean, Brian, Felix, John P., Garcia, Maria L., Jochnowitz, Nina, Karanam, Bindhu V., Li, Xiaohua, Lyons, Kathryn A., McGowan, Erin, MacIntyre, D. Euan, Martin, William J., Priest, Birgit T., Smith, McHardy M., Tschirret-Guth, Richard, and Warren, Vivien A.
- Subjects
- *
ION channels , *SODIUM channels , *BIOCHEMISTRY , *ACTIVE biological transport - Abstract
Abstract: A series of benzazepinones were synthesized and evaluated as hNav1.7 sodium channel blockers. Several compounds from this series displayed good oral bioavailability and exposure and were efficacious in a rat model of neuropathic pain. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
31. Safety and Efficacy of a Topical Sodium Channel Inhibitor (TV-45070) in Patients With Postherpetic Neuralgia (PHN)
- Author
-
Katie Jane Webster Proctor, Judith Neville, Y P Goldberg, Robin Sherrington, Nicola Anne Price, Michael Fetell, Richard Malamut, Jeffery Vest, Rostam Namdari, Samer Kaber, and Simon N. Pimstone
- Subjects
Male ,0301 basic medicine ,Indoles ,Genotype ,Administration, Topical ,Analgesic ,Neuralgia, Postherpetic ,Subgroup analysis ,Placebo ,Proof of Concept Study ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,law ,medicine ,Clinical endpoint ,Humans ,Spiro Compounds ,Nav1.7 ,postherpetic neuralgia ,neuropathic pain ,Cross-Over Studies ,Postherpetic neuralgia ,business.industry ,NAV1.7 Voltage-Gated Sodium Channel ,Original Articles ,Middle Aged ,analgesic ,medicine.disease ,Crossover study ,Treatment Outcome ,030104 developmental biology ,Anesthesiology and Pain Medicine ,Anesthesia ,Neuralgia ,Female ,Neurology (clinical) ,business ,R1150W ,030217 neurology & neurosurgery ,Sodium Channel Blockers - Abstract
Objective The objective was to evaluate the safety and efficacy of TV-45070 ointment, as a treatment for postherpetic neuralgia, and to explore the response in patients with the Nav1.7 R1150W gain-of-function polymorphism. Materials and methods This was a randomized, placebo-controlled, 2-period, 2-treatment crossover trial. Patients with postherpetic neuralgia with moderate or greater pain received TV-45070 and placebo ointments, each applied twice daily for 3 weeks. The primary efficacy measure was the difference in change in mean daily pain score from baseline compared with the last week of placebo and active treatment. Secondary endpoints included responder rate analyses and a further exploratory analysis of response in carriers of the Nav1.7 R1150W polymorphism was conducted. Results Seventy patients were enrolled and 54 completed the study. TV-45070 was safe and well tolerated. No statistical difference was observed between treatments for the primary endpoint. However, the proportion of patients with ≥50% reduction in mean pain scores at week 3 was greater on TV-45070 than on placebo (26.8% vs. 10.7%, P=0.0039). Similarly, a greater proportion of patients on TV-45070 had a ≥30% reduction in mean pain scores at week 3 (39.3% on TV-45070 vs. 23.2% on placebo, P=0.0784). Of note, 63% of patients with the R1150W polymorphism versus 35% of wild-type carriers had a ≥30% reduction in mean pain score on TV-45070 at week 3 (no inferential analysis performed). Conclusions The 50% responder analysis suggests a subpopulation may exist with a more marked analgesic response to TV-45070.The trend toward a larger proportion of responders within Nav1.7 R1150W carriers warrants further investigation.
- Published
- 2017
- Full Text
- View/download PDF
32. The role of nociceptive synaptic transmission modulation at the spinal cord level in different pain states
- Author
-
Adámek, Pavel, Paleček, Jiří, Vaculín, Šimon, and Vlachová, Viktorie
- Subjects
nociception ,neuropatická bolest ,PI3K ,synaptický přenos ,paclitaxel ,nocicepce ,Bolest ,kapsaicin ,synaptic transmission ,neuropathic pain ,CB1 ,anandamid ,TLR4 ,anandamide ,Pain ,TRPV1 ,Nav1.7 ,capsaicin - Abstract
Pain is a common symptom of many clinical syndromes and diseases. In particular, the treatment of neuropathic pain represents a serious public health issue because currently available analgesia is ineffective in many cases or it has adverse effects. Treatment of pain-related suffering requires knowledge of how pain signals are initially generated and subsequently transmitted by the nervous system. A nociceptive system plays a key role in this process of encoding and transmission of pain signals. Modulation of the nociceptive synaptic transmission in the spinal cord dorsal horn represents an important mechanism in the development and maintenance of different pathological pain states. This doctoral thesis has aimed to investigate and clarify some of the mechanisms involved in the modulation of the spinal nociceptive processing in different pain states. The main attention was paid to study the following issues: (I.) Which is the role of Transient Receptor Potential Vanilloid type 1 channels (TRPV1), Toll-Like Receptors 4 (TLR4), and phosphatidylinositol 3-kinase (PI3K) in the development of neuropathic pain induced by paclitaxel (PAC) chemotherapy in acute in vitro, and subchronic in vivo murine model of PAC-induced peripheral neuropathy (PIPN)? (II.) How is affected spinal inhibitory synaptic control...
- Published
- 2019
33. Chronic non-paroxysmal neuropathic pain — Novel phenotype of mutation in the sodium channel SCN9A gene
- Author
-
Dabby, Ron, Sadeh, Menachem, Gilad, Ronit, Lampl, Yair, Cohen, Sarit, Inbar, Shani, and Leshinsky-Silver, Esther
- Subjects
- *
CHRONIC pain , *PHENOTYPES , *GENETIC mutation , *SODIUM channels , *ERYTHROMELALGIA , *AMINO acids - Abstract
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. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
34. Conotoxins Targeting Neuronal Voltage-Gated Sodium Channel Subtypes: Potential Analgesics?
- Author
-
Oliver Knapp, Jeffrey R. McArthur, and David J. Adams
- Subjects
Models, Molecular ,Health, Toxicology and Mutagenesis ,Molecular Sequence Data ,lcsh:Medicine ,μ-conotoxin ,Voltage-Gated Sodium Channels ,Review ,mO-conotoxin ,Pharmacology ,Toxicology ,Nav1.9 ,Drug Discovery ,voltage-gated sodium channel ,Animals ,Humans ,pain ,Amino Acid Sequence ,nociception ,Conotoxin ,Binding site ,Voltage-Gated Sodium Channel Blockers ,Nav1.7 ,Ion channel ,Nav1.8 ,Neurons ,Analgesics ,Binding Sites ,Nav1.3 ,Chemistry ,Drug discovery ,Sodium channel ,lcsh:R ,Conus Snail ,m-conotoxin ,analgesic ,nervous system ,Neuropathic pain ,Neuralgia ,Conotoxins ,μO-conotoxin - Abstract
Voltage-gated sodium channels (VGSC) are the primary mediators of electrical signal amplification and propagation in excitable cells. VGSC subtypes are diverse, with different biophysical and pharmacological properties, and varied tissue distribution. Altered VGSC expression and/or increased VGSC activity in sensory neurons is characteristic of inflammatory and neuropathic pain states. Therefore, VGSC modulators could be used in prospective analgesic compounds. VGSCs have specific binding sites for four conotoxin families: μ-, μO-, δ- and ί-conotoxins. Various studies have identified that the binding site of these peptide toxins is restricted to well-defined areas or domains. To date, only the μ- and μO-family exhibit analgesic properties in animal pain models. This review will focus on conotoxins from the μ- and μO-families that act on neuronal VGSCs. Examples of how these conotoxins target various pharmacologically important neuronal ion channels, as well as potential problems with the development of drugs from conotoxins, will be discussed.
- Published
- 2012
- Full Text
- View/download PDF
35. Pain Disorders and Erythromelalgia Caused by Voltage-Gated Sodium Channel Mutations
- Author
-
Dabby, Ron
- Published
- 2012
- Full Text
- View/download PDF
36. SCN9A Variants May be Implicated in Neuropathic Pain Associated With Diabetic Peripheral Neuropathy and Pain Severity
- Author
-
Hao Wang, Alan Wickenden, Gary Romano, Peter Cheng, Qingqin S. Li, and Reyna Favis
- Subjects
Male ,Canada ,Genotyping Techniques ,diabetic painful neuropathy ,Bioinformatics ,Severity of Illness Index ,Linkage Disequilibrium ,White People ,Cohort Studies ,Diabetic Neuropathies ,Severity of illness ,medicine ,Humans ,Genetic Predisposition to Disease ,Small Fiber Neuropathy ,Nav1.7 ,Pain Measurement ,neuropathic pain ,SCN9A ,business.industry ,NAV1.7 Voltage-Gated Sodium Channel ,Chronic pain ,Genetic Variation ,Exons ,Original Articles ,Middle Aged ,medicine.disease ,Introns ,United States ,Anesthesiology and Pain Medicine ,Peripheral neuropathy ,Pain severity ,Anesthesia ,Neuropathic pain ,Neuralgia ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Female ,Neurology (clinical) ,SCN9A Gene ,Chronic Pain ,business ,targeted deep sequencing - Abstract
Supplemental Digital Content is available in the text., Objectives: Previous studies have established the role of SCN9A in various pain conditions, including idiopathic small fiber neuropathy. In the present study, we interrogate the relationship between common and rare variants in SCN9A gene and chronic neuropathic pain associated with diabetic peripheral neuropathy. Design: Using a cohort of 938 patients of European ancestry with chronic neuropathic pain associated with diabetic peripheral neuropathy enrolled in 6 clinical studies and 2 controls (POPRES, n=2624 and Coriell, n=1029), we examined the relationship between SCN9A variants and neuropathic pain in a case-control study using a 2-stage design. The exonic regions of SCN9A were sequenced in a subset of 244 patients with neuropathic pain, and the variants discovered were compared with POPRES control (stage 1). The top associated variants were followed up by genotyping in the entire case collection and Coriell controls restricting the analysis to the matching patients from the United States and Canada only (stage 2). Results: Seven variants were found to be associated with neuropathic pain at the sequencing stage. Four variants (Asp1908Gly, Val991Leu/Met932Leu, and an intronic variant rs74449889) were confirmed by genotyping to occur at a higher frequency in cases than controls (odds ratios ∼2.1 to 2.6, P=0.05 to 0.009). Val991Leu/Met932Leu was also associated with the severity of pain as measured by pain score Numeric Rating Scale (NRS-11, P=0.047). Val991Leu/Met932Leu variants were in complete linkage disequilibrium and previously shown to cause hyperexcitability in dorsal root ganglia neurons. Conclusions: The association of SCN9A variants with neuropathic pain and pain severity suggests a role of SCN9A in the disease etiology of neuropathic pain.
- Published
- 2015
37. Novel Mutations Mapping to the Fourth Sodium Channel Domain of Nav1.7 Result in Variable Clinical Manifestations of Primary Erythromelalgia
- Author
-
Kevin Howell, Istvan Bodi, Roman Cregg, John N. Wood, Bisola Laguda, Robert Werdehausen, Karen Agnew, John E. Linley, Henry Houlden, Maria Kinali, Ya-Chun Chen, Juan D. Ramirez, Michael Markiewicz, David L.H. Bennett, Michael P. Lunn, and James J. Cox
- Subjects
Patch-Clamp Techniques ,Action Potentials ,medicine.disease_cause ,Neuropathic pain ,0302 clinical medicine ,Missense mutation ,Age of Onset ,Nav1.7 ,0303 health sciences ,Mutation ,Analgesics ,NAV1.7 Voltage-Gated Sodium Channel ,Middle Aged ,Erythromelalgia ,Phenotype ,Neurology ,Child, Preschool ,Sensation Disorders ,Molecular Medicine ,Female ,Voltage-gated sodium channels ,Congenital insensitivity to pain ,medicine.medical_specialty ,Recombinant Fusion Proteins ,Molecular Sequence Data ,Mutation, Missense ,Transfection ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Internal medicine ,medicine ,Paroxysmal extreme pain disorder ,Humans ,Point Mutation ,Amino Acid Sequence ,030304 developmental biology ,Original Paper ,Ion Transport ,Sequence Homology, Amino Acid ,business.industry ,Point mutation ,Sodium channel ,Sodium ,medicine.disease ,Protein Structure, Tertiary ,Endocrinology ,HEK293 Cells ,Gain-of-function mutations ,Mutagenesis, Site-Directed ,Primary Erythromelalgia ,business ,Neuroscience ,Sequence Alignment ,030217 neurology & neurosurgery - Abstract
We identified and clinically investigated two patients with primary erythromelalgia mutations (PEM), which are the first reported to map to the fourth domain of Nav1.7 (DIV). The identified mutations (A1746G and W1538R) were cloned and transfected to cell cultures followed by electrophysiological analysis in whole-cell configuration. The investigated patients presented with PEM, while age of onset was very different (3 vs. 61 years of age). Electrophysiological characterization revealed that the early onset A1746G mutation leads to a marked hyperpolarizing shift in voltage dependence of steady-state activation, larger window currents, faster activation kinetics (time-to-peak current) and recovery from steady-state inactivation compared to wild-type Nav1.7, indicating a pronounced gain-of-function. Furthermore, we found a hyperpolarizing shift in voltage dependence of slow inactivation, which is another feature commonly found in Nav1.7 mutations associated with PEM. In silico neuron simulation revealed reduced firing thresholds and increased repetitive firing, both indicating hyperexcitability. The late-onset W1538R mutation also revealed gain-of-function properties, although to a lesser extent. Our findings demonstrate that mutations encoding for DIV of Nav1.7 can not only be linked to congenital insensitivity to pain or paroxysmal extreme pain disorder but can also be causative of PEM, if voltage dependency of channel activation is affected. This supports the view that the degree of biophysical property changes caused by a mutation may have an impact on age of clinical manifestation of PEM. In summary, these findings extent the genotype–phenotype correlation profile for SCN9A and highlight a new region of Nav1.7 that is implicated in PEM. Electronic supplementary material The online version of this article (doi:10.1007/s12017-012-8216-8) contains supplementary material, which is available to authorized users.
- Published
- 2013
38. Nav1.7 protein and mRNA expression in the dorsal root ganglia of rats with chronic neuropathic pain
- Author
-
Chao, Liu, Jing, Cao, Xiuhua, Ren, and Weidong, Zang
- Subjects
neuropathic pain ,Research and Report: Spinal Cord Injury and Neural Regeneration ,regeneration ,dorsal root ganglia ,neural regeneration ,Nav1.7 ,sensitization ,sodium channel ,hyperalgesia - Abstract
Neuropathic pain was produced by chronic constriction injury of the sciatic nerve in rats. Behavioral tests showed that the thresholds for thermal and mechanical hyperalgesia were significantly reduced in neuropathic pain rats 3–28 days following model induction. The results of immunohistochemistry, western blot assays and reverse transcription-PCR showed that Nav1.7 protein and mRNA expression was significantly increased in the injured dorsal root ganglia. These findings indicated that Nav1.7 might play an important role in the model of chronic neuropathic pain.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.