1
|
Liu X, Xiang J, Fan S, Chen X, Peng C, Xu Z. 20S-Ginsenoside Rh2, the major bioactive saponin in Panax notoginseng flowers, ameliorates cough by inhibition of NaV1.7 and TRPV1 channel currents and downregulation of TRPV1 expression. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118716. [PMID: 39179055 DOI: 10.1016/j.jep.2024.118716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax notoginseng flowers, which are the buds of the traditional Chinese medicinal herb Sanqi, are widely used in China for their cough-ameliorating properties, with demonstrated therapeutic effects in the treatment of both acute and chronic coughs. However, both the antitussive mechanism and active compound basis of P. notoginseng flowers remain poorly understood. AIM OF THE STUDY We investigated the antitussive effects of P. notoginseng flowers, identified the bioactive constituents responsible for alleviating cough symptoms, and elucidated the underlying pharmacological mechanisms. MATERIALS AND METHODS We analyzed the major chemical constituents of aqueous extracts of P. notoginseng flowers using liquid chromatography-mass spectrometry and quantitatively analyzed the key component, 20S-ginsenoside Rh2, using high-performance liquid chromatography. Using a cough reflex model in healthy mice and an ovalbumin-induced, highly sensitive guinea pig cough model, we verified the suppressive effects of P. notoginseng flowers and their saponin constituents on coughing. Furthermore, we explored the mechanisms of action of the key ion channels, NaV1.7 and TRPV1, using whole-cell patch-clamp techniques and molecular docking. Finally, the therapeutic mechanisms of P. notoginseng flowers on pathological cough were revealed using hematoxylin and eosin staining, immunohistochemistry, and western blotting. RESULTS The active components of P. notoginseng flowers were primarily protopanaxadiol-type saponins, among which 20S-ginsenoside Rh2 had the highest content (51.46 mg/g). In the mouse model, P. notoginseng flowers exhibited antitussive effects comparable to those of pentoxyverine citrate. Although its main saponin component, 20S-ginsenoside Rh2, showed slightly weaker effects, it still demonstrated concentration-dependent inhibition of channel activity. The whole-cell patch-clamp technique and virtual molecular docking showed that Rh2 might exert its effects by directly binding to the NaV1.7 and TRPV1 channels. In the guinea pig model, P. notoginseng flowers and their saponin components not only reduced cough frequency and prolonged the latency period before cough onset, but also significantly inhibited tracheal and pulmonary inflammation and the overexpression of TRPV1. CONCLUSIONS 20S-Ginsenoside Rh2, the major bioactive saponin in P. notoginseng flowers, exhibits potent antitussive effects. The potential mechanism of action of 20S-Ginsenoside Rh2 in the treatment of cough may involve inhibiting NaV1.7 and TRPV1 channel currents through direct binding to core protein active sites and downregulating TRPV1 expression.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Jie Xiang
- Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Shuyuan Fan
- Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Xumin Chen
- Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Chengzhan Peng
- Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Zhengxin Xu
- Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou, 225009, China; Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou, 225009, China; Yeda Institute of Gene and Cell Therapy, Taizhou, 318000, China.
| |
Collapse
|
2
|
Kantarci H, Elvira PD, Thottumkara AP, O'Connell EM, Iyer M, Donovan LJ, Dugan MQ, Ambiel N, Granados A, Zeng H, Saw NL, Brosius Lutz A, Sloan SA, Gray EE, Tran KV, Vichare A, Yeh AK, Münch AE, Huber M, Agrawal A, Morri M, Zhong H, Shamloo M, Anderson TA, Tawfik VL, Du Bois J, Zuchero JB. Schwann cell-secreted PGE 2 promotes sensory neuron excitability during development. Cell 2024; 187:4690-4712.e30. [PMID: 39142281 DOI: 10.1016/j.cell.2024.07.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/18/2024] [Accepted: 06/21/2024] [Indexed: 08/16/2024]
Abstract
Electrical excitability-the ability to fire and propagate action potentials-is a signature feature of neurons. How neurons become excitable during development and whether excitability is an intrinsic property of neurons remain unclear. Here, we demonstrate that Schwann cells, the most abundant glia in the peripheral nervous system, promote somatosensory neuron excitability during development. We find that Schwann cells secrete prostaglandin E2, which is necessary and sufficient to induce developing somatosensory neurons to express normal levels of genes required for neuronal function, including voltage-gated sodium channels, and to fire action potential trains. Inactivating this signaling pathway in Schwann cells impairs somatosensory neuron maturation, causing multimodal sensory defects that persist into adulthood. Collectively, our studies uncover a neurodevelopmental role for prostaglandin E2 distinct from its established role in inflammation, revealing a cell non-autonomous mechanism by which glia regulate neuronal excitability to enable the development of normal sensory functions.
Collapse
Affiliation(s)
- Husniye Kantarci
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Pablo D Elvira
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | | | - Emma M O'Connell
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Manasi Iyer
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lauren J Donovan
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Micaela Quinn Dugan
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nicholas Ambiel
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Hong Zeng
- Transgenic, Knockout and Tumor model Center (TKTC), Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nay L Saw
- Behavioral and Functional Neuroscience Laboratory, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amanda Brosius Lutz
- Department of Obstetrics and Gynecology, University Hospital, Bern, Switzerland
| | - Steven A Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Erin E Gray
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Khanh V Tran
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aditi Vichare
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ashley K Yeh
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alexandra E Münch
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Max Huber
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Aditi Agrawal
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | | | - Haining Zhong
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Mehrdad Shamloo
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Behavioral and Functional Neuroscience Laboratory, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Thomas Anthony Anderson
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Vivianne L Tawfik
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - J Du Bois
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA.
| | - J Bradley Zuchero
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
3
|
Mondal R, Vaissier Welborn V. Dynamics accelerate the kinetics of ion diffusion through channels: Continuous-time random walk models beyond the mean field approximation. J Chem Phys 2024; 160:144109. [PMID: 38597306 DOI: 10.1063/5.0188469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 03/14/2024] [Indexed: 04/11/2024] Open
Abstract
Ion channels are proteins that play a significant role in physiological processes, including neuronal excitability and signal transduction. However, the precise mechanisms by which these proteins facilitate ion diffusion through cell membranes are not well understood. This is because experimental techniques to characterize ion channel activity operate on a time scale too large to understand the role of the various protein conformations on diffusion. Meanwhile, computational approaches operate on a time scale too short to rationalize the observed behavior at the microscopic scale. In this paper, we present a continuous-time random walk model that aims to bridge the scales between the atomistic models of ion channels and the experimental measurement of their conductance. We show how diffusion slows down in complex systems by using 3D lattices that map out the pore geometry of two channels: Nav1.7 and gramicidin. We also introduce spatial and dynamic site disorder to account for system heterogeneity beyond the mean field approximation. Computed diffusion coefficients show that an increase in spatial disorder slows down diffusion kinetics, while dynamic disorder has the opposite effect. Our results imply that microscopic or phenomenological models based on the potential of mean force data overlook the functional importance of protein dynamics on ion diffusion through channels.
Collapse
Affiliation(s)
- Ronnie Mondal
- Department of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, USA
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, Virginia 24061, USA
| | - Valerie Vaissier Welborn
- Department of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, USA
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, Virginia 24061, USA
| |
Collapse
|
4
|
Du WJ, Yang H, Tong F, Liu S, Zhang C, Chen Y, Yan Y, Xiang YW, Hua LY, Gong Y, Xu ZX, Liu X, Jiang X, Lu M, Guan JS, Han Q. Ash1L ameliorates psoriasis via limiting neuronal activity-dependent release of miR-let-7b. Br J Pharmacol 2024; 181:1107-1127. [PMID: 37766518 DOI: 10.1111/bph.16254] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/12/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND AND PURPOSE Psoriasis is a common autoimmune skin disease that significantly diminishes patients' quality of life. Interactions between primary afferents of the somatosensory system and the cutaneous immune system mediate the pathogenesis of psoriasis. This study aims to elucidate the molecular mechanisms of how primary sensory neurons regulate psoriasis formation. EXPERIMENTAL APPROACH Skin and total RNA were extracted from wild-type (WT) and ASH1-like histone lysine methyltransferase (Ash1l+/- ) mice in both naive and imiquimod (IMQ)-induced psoriasis models. Immunohistochemistry, quantitative real-time polymerase chain reaction (qRT-PCR) and fluorescence-activated cell sorting (FACS) were then performed. Microfluidic chamber coculture was used to investigate the interaction between somatosensory neurons and bone marrow dendritic cells (BMDCs) ex vivo. Whole-cell patch clamp recordings were used to evaluate neuronal excitability after Ash1L haploinsufficiency in primary sensory neurons. KEY RESULTS The haploinsufficiency of ASH1L, a histone methyltransferase, in primary sensory neurons causes both neurite hyperinnervation and increased neuronal excitability, which promote miR-let-7b release from primary afferents in the skin in a neuronal activity-dependent manner. With a 'GUUGUGU' core sequence, miR-let-7b functions as an endogenous ligand of toll-like receptor 7 (TLR7) and stimulates the activation of dermal dendritic cells (DCs) and interleukin (IL)-23/IL-17 axis, ultimately exacerbating the symptoms of psoriasis. Thus, by limiting miR-let-7b release from primary afferents, ASH1L prevents dermal DC activation and ameliorates psoriasis. CONCLUSION AND IMPLICATIONS Somatosensory neuron ASH1L modulates the cutaneous immune system by limiting neuronal activity-dependent release of miR-let-7b, which can directly activate dermal DCs via TLR7 and ultimately lead to aggravated psoriatic lesion.
Collapse
Affiliation(s)
- Wan-Jie Du
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Huan Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Fang Tong
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuai Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Chen Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yeying Chen
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Yuze Yan
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yan-Wei Xiang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ling-Yang Hua
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ye Gong
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhi-Xiang Xu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoyan Liu
- Department of Biomedical Engineering, Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Xingyu Jiang
- Department of Biomedical Engineering, Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Mingfang Lu
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Ji-Song Guan
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Qingjian Han
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Lu H, Cao P. Neural Mechanisms Underlying the Coughing Reflex. Neurosci Bull 2023; 39:1823-1839. [PMID: 37606821 PMCID: PMC10661548 DOI: 10.1007/s12264-023-01104-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/15/2023] [Indexed: 08/23/2023] Open
Abstract
Breathing is an intrinsic natural behavior and physiological process that maintains life. The rhythmic exchange of gases regulates the delicate balance of chemical constituents within an organism throughout its lifespan. However, chronic airway diseases, including asthma and chronic obstructive pulmonary disease, affect millions of people worldwide. Pathological airway conditions can disrupt respiration, causing asphyxia, cardiac arrest, and potential death. The innervation of the respiratory tract and the action of the immune system confer robust airway surveillance and protection against environmental irritants and pathogens. However, aberrant activation of the immune system or sensitization of the nervous system can contribute to the development of autoimmune airway disorders. Transient receptor potential ion channels and voltage-gated Na+ channels play critical roles in sensing noxious stimuli within the respiratory tract and interacting with the immune system to generate neurogenic inflammation and airway hypersensitivity. Although recent studies have revealed the involvement of nociceptor neurons in airway diseases, the further neural circuitry underlying airway protection remains elusive. Unraveling the mechanism underpinning neural circuit regulation in the airway may provide precise therapeutic strategies and valuable insights into the management of airway diseases.
Collapse
Affiliation(s)
- Haicheng Lu
- National Institute of Biological Sciences, Beijing, 102206, China.
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Peng Cao
- National Institute of Biological Sciences, Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China
| |
Collapse
|
6
|
Nair SS, Pavelkova N, Murphy CM, Kollarik M, Taylor-Clark TE. Action potential conduction in the mouse and rat vagus nerve is dependent on multiple voltage-gated sodium channels (Na V1s). J Neurophysiol 2023; 130:684-693. [PMID: 37584077 PMCID: PMC10635471 DOI: 10.1152/jn.00041.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 08/14/2023] [Accepted: 08/14/2023] [Indexed: 08/17/2023] Open
Abstract
Action potential (AP) conduction depends on voltage-gated sodium channels, of which there are nine subtypes. The vagus nerve, comprising sensory afferent fibers and efferent parasympathetic fibers, provides autonomic regulation of visceral organs, but the voltage-gated sodium channels (NaV1) subtypes involved in its AP conduction are poorly defined. We studied the A- and C-waves of electrically stimulated compound action potentials (CAPs) of the mouse and rat vagus nerves with and without NaV1 inhibitor administration: tetrodotoxin (TTX), PF-05089771 (mouse NaV1.7), ProTX-II (NaV1.7), ICA-121341 (NaV1.1, NaV1.3, and NaV1.6), LSN-3049227 (NaV1.2, NaV1.6, and NaV1.7), and A-803467 (NaV1.8). We show that TTX-sensitive NaV1 channels are essential for all vagal AP conduction. PF-05089771 but not ICA-121341 inhibited the mouse A-wave, which was abolished by LSN-3049227, suggesting roles for NaV1.7 and NaV1.2. The mouse C-wave was abolished by LSN-3049227 and a combination of PF-05089771 and ICA-121341, suggesting roles for NaV1.7 and NaV1.6. The rat A-wave was inhibited by ProTX-II, ICA-121341, and a combination of these inhibitors but only abolished by LSN-3049227, suggesting roles for NaV1.7, NaV1.6, and NaV1.2. The rat C-wave was abolished by LSN-3049227 and a combination of ProTX-II and ICA-121341, suggesting roles for NaV1.7 and NaV1.6. A-803467 also inhibited the mouse and rat CAP suggesting a cooperative role for the TTX-resistant NaV1.8. Overall, our data demonstrate that multiple NaV1 subtypes contribute to vagal CAPs, with NaV1.7 and NaV1.8 playing predominant roles and NaV1.6 and NaV1.2 contributing to a different extent based on nerve fiber type and species. Inhibition of these NaV1 may impact autonomic regulation of visceral organs.NEW & NOTEWORTHY Distinct NaV1 channels are involved in action potential (AP) initiation and conduction from afferent terminals within specific organs. Here, we have identified the NaV1 necessary for AP conduction in the entire murine and rat vagus nerve. We show TTX-sensitive channels are essential for all AP conduction, predominantly NaV1.7 with NaV1.2 and NaV1.6 playing lesser roles depending on the species and fiber type. In addition, we show that NaV1.8 is also essential for most axonal AP conduction.
Collapse
Affiliation(s)
- Sanjay S Nair
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States
| | - Nikoleta Pavelkova
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States
| | - Claire M Murphy
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States
| | - Marian Kollarik
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States
| | - Thomas E Taylor-Clark
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States
| |
Collapse
|
7
|
Finol-Urdaneta RK, Zhorov BS, Baden DG, Adams DJ. Brevetoxin versus Brevenal Modulation of Human Nav1 Channels. Mar Drugs 2023; 21:396. [PMID: 37504927 PMCID: PMC10382042 DOI: 10.3390/md21070396] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023] Open
Abstract
Brevetoxins (PbTx) and brevenal are marine ladder-frame polyethers. PbTx binds to and activates voltage-gated sodium (Nav) channels in native tissues, whereas brevenal antagonizes these actions. However, the effects of PbTx and brevenal on recombinant Nav channel function have not been systematically analyzed. In this study, the PbTx-3 and brevenal modulation of tissue-representative Nav channel subtypes Nav1.2, Nav1.4, Nav1.5, and Nav1.7 were examined using automated patch-clamp. While PbTx-3 and brevenal elicit concentration-dependent and subtype-specific modulatory effects, PbTx-3 is >1000-fold more potent than brevenal. Consistent with effects observed in native tissues, Nav1.2 and Nav1.4 channels were PbTx-3- and brevenal-sensitive, whereas Nav1.5 and Nav1.7 appeared resistant. Interestingly, the incorporation of brevenal in the intracellular solution caused Nav channels to become less sensitive to PbTx-3 actions. Furthermore, we generated a computational model of PbTx-2 bound to the lipid-exposed side of the interface between domains I and IV of Nav1.2. Our results are consistent with competitive antagonism between brevetoxins and brevenal, setting a basis for future mutational analyses of Nav channels' interaction with brevetoxins and brevenal. Our findings provide valuable insights into the functional modulation of Nav channels by brevetoxins and brevenal, which may have implications for the development of new Nav channel modulators with potential therapeutic applications.
Collapse
Affiliation(s)
- Rocio K Finol-Urdaneta
- Illawarra Health & Medical Research Institute (IHMRI), Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Boris S Zhorov
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 Saint Petersburg, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Daniel G Baden
- Center for Marine Science, University of North Carolina Wilmington MARBIONC, Wilmington, NC 28409, USA
| | - David J Adams
- Illawarra Health & Medical Research Institute (IHMRI), Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
8
|
Hernández-Plata E, Cruz AA, Becerril C. Na V1.7 channels are expressed in the lower airways of the human respiratory tract. Respir Physiol Neurobiol 2023; 311:104034. [PMID: 36792043 DOI: 10.1016/j.resp.2023.104034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/01/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023]
Abstract
NaV channels expression have been reported in upper airways and tracheal smooth muscle cells controlling the generation and propagation of action potentials in the respiratory tract sensory neurons, but information about the presence of these proteins in the bronchioalveolar structures in human lungs was missing. The main objective covered in this work was to determine whether the NaV1.7 channels are expressed in lower airways, and to identify the cellular identities expressing these proteins. We detected high levels of the mRNA coding for NaV1.7 channels in isolated lung fibroblasts obtained from both normal lungs, and fibrotic lungs of patients with respiratory diseases. The protein was detected with two different antibodies in the bronchioalveolar tissue, alveolar endothelium, and capillary endothelium, in normal and pathologic lungs. These evidences are useful in the dissection of molecular mechanisms of pulmonary pathologies, and lead to consider the NaV1.7 channels as potential therapeutic targets for the treatment of pulmonary diseases.
Collapse
Affiliation(s)
- Everardo Hernández-Plata
- Investigador por México, Consejo Nacional de Ciencia y Tecnología, and Instituto Nacional de Medicina Genómica, Mexico City, Mexico.
| | - Ana Alfaro Cruz
- Departamento de Patología, Hospital General de México, "Dr. Eduardo Liceaga", Mexico City, Mexico
| | - Carina Becerril
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico
| |
Collapse
|
9
|
Kim JS, Sun H, Meeker S, Undem BJ. Role of Na V 1.9 in inflammatory mediator-induced activation of mouse airway vagal C-fibres. J Physiol 2023; 601:1139-1150. [PMID: 36750759 PMCID: PMC10023385 DOI: 10.1113/jp283751] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 01/23/2023] [Indexed: 02/09/2023] Open
Abstract
The influence of NaV 1.9 on inflammatory mediator-induced activation of airway vagal nodose C-fibres was evaluated by comparing responses in wild-type versus NaV 1.9-/- mice. A single-cell RT-PCR analysis indicated that virtually all nodose C-fibre neurons expressed NaV 1.9 (SCN11A) mRNA. Using extracellular electrophysiological recordings in an isolated vagally innervated mouse trachea-lung preparation, it was noted that mediators acting via G protein-coupled receptors (PAR2), or ionotropic receptors (P2×3) were 70-85% less effective in evoking action potential discharge in the absence of NaV 1.9. However, there was no difference in action potential discharge between wild-type and NaV 1.9-/- when the stimulus was a rapid punctate mechanical stimulus. An analysis of the passive and active properties of isolated nodose neurons revealed no difference between neurons from wild-type and NaV 1.9-/- mice, with the exception of a modest difference in the duration of the afterhyperpolarization. There was also no difference in the amount of current required to evoke action potentials (rheobase) or the action potential voltage threshold. The inward current evoked by the chemical mediator by a P2×3 agonist was the same in wild-type versus NaV 1.9-/- neurons. However, the current was sufficient to evoke action potential only in the wild-type neurons. The data support the speculation that NaV 1.9 could be an attractive therapeutic target for inflammatory airway disease by selectively inhibiting inflammatory mediator-associated vagal C-fibre activation. KEY POINTS: Inflammatory mediators were much less effective in activating the terminals of vagal airway C-fibres in mice lacking NaV 1.9. The active and passive properties of nodose neurons were the same between wild-type neurons and NaV 1.9-/- neurons. Nerves lacking NaV 1.9 responded, normally, with action potential discharge to rapid punctate mechanical stimulation of the terminals or the rapid stimulation of the cell bodies with inward current injections. NaV 1.9 channels could be an attractive target to selectively inhibit vagal nociceptive C-fibre activation evoked by inflammatory mediators without blocking the nerves' responses to the potentially hazardous stimuli associated with aspiration.
Collapse
Affiliation(s)
- Joyce S Kim
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hui Sun
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sonya Meeker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bradley J Undem
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
10
|
Loya-López SI, Duran P, Ran D, Calderon-Rivera A, Gomez K, Moutal A, Khanna R. Cell specific regulation of NaV1.7 activity and trafficking in rat nodose ganglia neurons. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2022; 12:100109. [PMID: 36531612 PMCID: PMC9755031 DOI: 10.1016/j.ynpai.2022.100109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 06/17/2023]
Abstract
The voltage-gated sodium NaV1.7 channel sets the threshold for electrogenesis. Mutations in the gene encoding human NaV1.7 (SCN9A) cause painful neuropathies or pain insensitivity. In dorsal root ganglion (DRG) neurons, activity and trafficking of NaV1.7 are regulated by the auxiliary collapsin response mediator protein 2 (CRMP2). Specifically, preventing addition of a small ubiquitin-like modifier (SUMO), by the E2 SUMO-conjugating enzyme Ubc9, at lysine-374 (K374) of CRMP2 reduces NaV1.7 channel trafficking and activity. We previously identified a small molecule, designated 194, that prevented CRMP2 SUMOylation by Ubc9 to reduce NaV1.7 surface expression and currents, leading to a reduction in spinal nociceptive transmission, and culminating in normalization of mechanical allodynia in models of neuropathic pain. In this study, we investigated whether NaV1.7 control via CRMP2-SUMOylation is conserved in nodose ganglion (NG) neurons. This study was motivated by our desire to develop 194 as a safe, non-opioid substitute for persistent pain, which led us to wonder how 194 would impact NaV1.7 in NG neurons, which are responsible for driving the cough reflex. We found functioning NaV1.7 channels in NG neurons; however, they were resistant to downregulation via either CRMP2 knockdown or pharmacological inhibition of CRMP2 SUMOylation by 194. CRMP2 SUMOylation and interaction with NaV1.7 was consered in NG neurons but the endocytic machinery was deficient in the endocytic adaptor protein Numb. Overexpression of Numb rescued CRMP2-dependent regulation on NaV1.7, rendering NG neurons sensitive to 194. Altogether, these data point at the existence of cell-specific mechanisms regulating NaV1.7 trafficking.
Collapse
Affiliation(s)
- Santiago I. Loya-López
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, USA
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, USA
| | - Paz Duran
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, USA
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, USA
| | - Dongzhi Ran
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Aida Calderon-Rivera
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, USA
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, USA
| | - Kimberly Gomez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, USA
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, USA
| | - Aubin Moutal
- School of Medicine, Department of Pharmacology and Physiology, Saint Louis University, Saint Louis, MO 63104, USA
| | - Rajesh Khanna
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, USA
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, USA
| |
Collapse
|
11
|
Elleman AV, Du Bois J. Chemical and Biological Tools for the Study of Voltage-Gated Sodium Channels in Electrogenesis and Nociception. Chembiochem 2022; 23:e202100625. [PMID: 35315190 PMCID: PMC9359671 DOI: 10.1002/cbic.202100625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/22/2022] [Indexed: 12/17/2022]
Abstract
The malfunction and misregulation of voltage-gated sodium channels (NaV s) underlie in large part the electrical hyperexcitability characteristic of chronic inflammatory and neuropathic pain. NaV s are responsible for the initiation and propagation of electrical impulses (action potentials) in cells. Tissue and nerve injury alter the expression and localization of multiple NaV isoforms, including NaV 1.1, 1.3, and 1.6-1.9, resulting in aberrant action potential firing patterns. To better understand the role of NaV regulation, localization, and trafficking in electrogenesis and pain pathogenesis, a number of chemical and biological reagents for interrogating NaV function have been advanced. The development and application of such tools for understanding NaV physiology are the focus of this review.
Collapse
Affiliation(s)
- Anna V Elleman
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - J Du Bois
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
12
|
Fouda MA, Ghovanloo MR, Ruben PC. Late sodium current: incomplete inactivation triggers seizures, myotonias, arrhythmias, and pain syndromes. J Physiol 2022; 600:2835-2851. [PMID: 35436004 DOI: 10.1113/jp282768] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/12/2022] [Indexed: 11/08/2022] Open
Abstract
Acquired and inherited dysfunction in voltage-gated sodium channels underlies a wide range of diseases. "In addition to the defects in trafficking and expression, sodium channelopathies are also caused by dysfunction in one or several gating properties, for instance activation or inactivation. Disruption of the channel inactivation leads to the increased late sodium current, which is a common defect in seizure disorders, cardiac arrhythmias skeletal muscle myotonia and pain. An increase in late sodium current leads to repetitive action potential in neurons and skeletal muscles, and prolonged action potential duration in the heart. In this topical review, we compare the effects of late sodium current in brain, heart, skeletal muscle, and peripheral nerves. Abstract figure legend Shows cartoon illustration of general Nav channel transitions between (1) resting, (2) open, and (3) fast inactivated states. Disruption of the inactivation process exacerbates (4) late sodium currents. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Mohamed A Fouda
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada.,Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt
| | | | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| |
Collapse
|
13
|
Kim SH, Patil MJ, Hadley SH, Bahia PK, Butler SG, Madaram M, Taylor-Clark TE. Mapping of the Sensory Innervation of the Mouse Lung by Specific Vagal and Dorsal Root Ganglion Neuronal Subsets. eNeuro 2022; 9:ENEURO.0026-22.2022. [PMID: 35365503 PMCID: PMC9015009 DOI: 10.1523/eneuro.0026-22.2022] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/10/2022] [Accepted: 03/26/2022] [Indexed: 11/21/2022] Open
Abstract
The airways are densely innervated by sensory afferent nerves, whose activation regulates respiration and triggers defensive reflexes (e.g., cough, bronchospasm). Airway innervation is heterogeneous, and distinct afferent subsets have distinct functional responses. However, little is known of the innervation patterns of subsets within the lung. A neuroanatomical map is critical for understanding afferent activation under physiological and pathophysiological conditions. Here, we quantified the innervation of the mouse lung by vagal and dorsal root ganglion (DRG) sensory subsets defined by the expression of Pirt (all afferents), 5HT3 (vagal nodose afferents), Tac1 (tachykinergic afferents), and transient receptor potential vanilloid 1 channel (TRPV1; defensive/nociceptive afferents) using Cre-mediated reporter expression. We found that vagal afferents innervate almost all conducting airways and project into the alveolar region, whereas DRG afferents only innervate large airways. Of the two vagal ganglia, only nodose afferents project into the alveolar region, but both nodose and jugular afferents innervate conducting airways throughout the lung. Many afferents that project into the alveolar region express TRPV1. Few DRG afferents expressed TRPV1. Approximately 25% of blood vessels were innervated by vagal afferents (many were Tac1+). Approximately 10% of blood vessels had DRG afferents (some were Tac1+), but this was restricted to large vessels. Lastly, innervation of neuroepithelial bodies (NEBs) correlated with the cell number within the bodies. In conclusion, functionally distinct sensory subsets have distinct innervation patterns within the conducting airways, alveoli and blood vessels. Physiologic (e.g., stretch) and pathophysiological (e.g., inflammation, edema) stimuli likely vary throughout these regions. Our data provide a neuroanatomical basis for understanding afferent responses in vivo.
Collapse
Affiliation(s)
- Seol-Hee Kim
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Mayur J Patil
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Stephen H Hadley
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Parmvir K Bahia
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Shane G Butler
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Meghana Madaram
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Thomas E Taylor-Clark
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| |
Collapse
|
14
|
Spinal cord injury-mediated changes in electrophysiological properties of rat gastric nodose ganglion neurons. Exp Neurol 2022; 348:113927. [PMID: 34798136 PMCID: PMC8727501 DOI: 10.1016/j.expneurol.2021.113927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 10/30/2021] [Accepted: 11/11/2021] [Indexed: 02/03/2023]
Abstract
In preclinical rodent models, spinal cord injury (SCI) manifests as gastric vagal afferent dysfunction both acutely and chronically. However, the mechanism that underlies this dysfunction remains unknown. In the current study, we examined the effect of SCI on gastric nodose ganglia (NG) neuron excitability and on voltage-gated Na+ (NaV) channels expression and function in rats after an acute (i.e. 3-days) and chronic (i.e. 3-weeks) period. Rats randomly received either T3-SCI or sham control surgery 3-days or 3-weeks prior to experimentation as well as injections of 3% DiI solution into the stomach to identify gastric NG neurons. Single cell qRT-PCR was performed on acutely dissociated DiI-labeled NG neurons to measure NaV1.7, NaV1.8 and NaV1.9 expression levels. The results indicate that all 3 channel subtypes decreased. Current- and voltage-clamp whole-cell patch-clamp recordings were performed on acutely dissociated DiI-labeled NG neurons to measure active and passive properties of C- and A-fibers as well as the biophysical characteristics of NaV1.8 channels in gastric NG neurons. Acute and chronic SCI did not demonstrate deleterious effects on either passive properties of dissociated gastric NG neurons or biophysical properties of NaV1.8. These findings suggest that although NaV gene expression levels change following SCI, NaV1.8 function is not altered. The disruption throughout the entirety of the vagal afferent neuron has yet to be investigated.
Collapse
|
15
|
Wistrom E, Chase R, Smith PR, Campbell ZT. A compendium of validated pain genes. WIREs Mech Dis 2022; 14:e1570. [PMID: 35760453 PMCID: PMC9787016 DOI: 10.1002/wsbm.1570] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/28/2022] [Accepted: 06/06/2022] [Indexed: 12/30/2022]
Abstract
The development of novel pain therapeutics hinges on the identification and rigorous validation of potential targets. Model organisms provide a means to test the involvement of specific genes and regulatory elements in pain. Here we provide a list of genes linked to pain-associated behaviors. We capitalize on results spanning over three decades to identify a set of 242 genes. They support a remarkable diversity of functions spanning action potential propagation, immune response, GPCR signaling, enzymatic catalysis, nucleic acid regulation, and intercellular signaling. Making use of existing tissue and single-cell high-throughput RNA sequencing datasets, we examine their patterns of expression. For each gene class, we discuss archetypal members, with an emphasis on opportunities for additional experimentation. Finally, we discuss how powerful and increasingly ubiquitous forward genetic screening approaches could be used to improve our ability to identify pain genes. This article is categorized under: Neurological Diseases > Genetics/Genomics/Epigenetics Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Eric Wistrom
- Department of Biological SciencesUniversity of Texas at DallasRichardsonTexasUSA
| | - Rebecca Chase
- Department of Biological SciencesUniversity of Texas at DallasRichardsonTexasUSA
| | - Patrick R. Smith
- Department of Biological SciencesUniversity of Texas at DallasRichardsonTexasUSA
| | - Zachary T. Campbell
- Department of Biological SciencesUniversity of Texas at DallasRichardsonTexasUSA,Center for Advanced Pain StudiesUniversity of Texas at DallasRichardsonTexasUSA
| |
Collapse
|
16
|
Kollarik M, Ru F, Pavelkova N, Mulcahy J, Hunter J, Undem BJ. Role of Na V 1.7 in action potential conduction along human bronchial vagal afferent C-fibres. Br J Pharmacol 2022; 179:242-251. [PMID: 34634134 DOI: 10.1111/bph.15686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/20/2021] [Accepted: 09/17/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND AND PURPOSE The purpose of this study was to determine the role of NaV 1.7 in action potential conduction in C-fibres in the bronchial branches of the human vagus nerve. EXPERIMENTAL APPROACH Bronchial branches of the vagus nerve were dissected from human donor tissue. The C-wave of the electrically evoked compound action potential was quantified in the absence and presence of increasing concentrations of the selective NaV 1.7 blocking drugs, PF-05089771 and ST-2262, as well as the NaV 1.1, 1.2, and 1.3 blocking drug ICA121-431. The efficacy and potency of these inhibitors were compared to the standard NaV 1 blocker, tetrodotoxin. We then compared the relative potencies of the NaV 1 blockers in inhibiting the C-wave of the compound action potential, with their ability to inhibit parasympathetic cholinergic contraction of human isolated bronchi, a response previously shown to be strictly dependent on NaV 1.7 channels. KEY RESULTS The selective NaV 1.7 blockers inhibited the C-wave of the compound action potential with potencies similar to that observed in the NaV 1.7 bronchial contractions assay. Using rt-PCR, we noted that NaV 1.7 mRNA was strongly expressed and transported down the vagus nerve bundles. CONCLUSIONS AND IMPLICATIONS NaV 1.7 blockers can prevent action potential conduction in the majority of vagal C-fibres arising from human bronchi. Blockers of NaV 1.7 channels may therefore have value in inhibiting the responses to excessive airway C-fibre activation in inflammatory airway disease, responses that include coughing as well as reflex bronchoconstriction and secretions.
Collapse
Affiliation(s)
- Marian Kollarik
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - Fei Ru
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Nikoleta Pavelkova
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - John Mulcahy
- SiteOne Therapeutics, South San Francisco, CA, USA
| | - John Hunter
- SiteOne Therapeutics, South San Francisco, CA, USA
| | - Bradley J Undem
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Taylor-Clark TE, Undem BJ. Neural control of the lower airways: Role in cough and airway inflammatory disease. HANDBOOK OF CLINICAL NEUROLOGY 2022; 188:373-391. [PMID: 35965034 PMCID: PMC10688079 DOI: 10.1016/b978-0-323-91534-2.00013-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Airway function is under constant neurophysiological control, in order to maximize airflow and gas exchange and to protect the airways from aspiration, damage, and infection. There are multiple sensory nerve subtypes, whose disparate functions provide a wide array of sensory information into the CNS. Activation of these subtypes triggers specific reflexes, including cough and alterations in autonomic efferent control of airway smooth muscle, secretory cells, and vasculature. Importantly, every aspect of these reflex arcs can be impacted and altered by local inflammation caused by chronic lung disease such as asthma, bronchitis, and infections. Excessive and inappropriate activity in sensory and autonomic nerves within the airways is thought to contribute to the morbidity and symptoms associated with lung disease.
Collapse
Affiliation(s)
- Thomas E Taylor-Clark
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Bradley J Undem
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
18
|
Brozmanova M, Buday T, Konarska M, Plevkova J. The effect of the voltage-gated sodium channel NaV1.7 blocker PF-05089771 on cough in the guinea pig. Respir Physiol Neurobiol 2022; 299:103856. [DOI: 10.1016/j.resp.2022.103856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/17/2022] [Accepted: 01/30/2022] [Indexed: 11/24/2022]
|
19
|
Alles SRA, Smith PA. Peripheral Voltage-Gated Cation Channels in Neuropathic Pain and Their Potential as Therapeutic Targets. FRONTIERS IN PAIN RESEARCH 2021; 2:750583. [PMID: 35295464 PMCID: PMC8915663 DOI: 10.3389/fpain.2021.750583] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/10/2021] [Indexed: 11/25/2022] Open
Abstract
The persistence of increased excitability and spontaneous activity in injured peripheral neurons is imperative for the development and persistence of many forms of neuropathic pain. This aberrant activity involves increased activity and/or expression of voltage-gated Na+ and Ca2+ channels and hyperpolarization activated cyclic nucleotide gated (HCN) channels as well as decreased function of K+ channels. Because they display limited central side effects, peripherally restricted Na+ and Ca2+ channel blockers and K+ channel activators offer potential therapeutic approaches to pain management. This review outlines the current status and future therapeutic promise of peripherally acting channel modulators. Selective blockers of Nav1.3, Nav1.7, Nav1.8, Cav3.2, and HCN2 and activators of Kv7.2 abrogate signs of neuropathic pain in animal models. Unfortunately, their performance in the clinic has been disappointing; some substances fail to meet therapeutic end points whereas others produce dose-limiting side effects. Despite this, peripheral voltage-gated cation channels retain their promise as therapeutic targets. The way forward may include (i) further structural refinement of K+ channel activators such as retigabine and ASP0819 to improve selectivity and limit toxicity; use or modification of Na+ channel blockers such as vixotrigine, PF-05089771, A803467, PF-01247324, VX-150 or arachnid toxins such as Tap1a; the use of Ca2+ channel blockers such as TTA-P2, TTA-A2, Z 944, ACT709478, and CNCB-2; (ii) improving methods for assessing "pain" as opposed to nociception in rodent models; (iii) recognizing sex differences in pain etiology; (iv) tailoring of therapeutic approaches to meet the symptoms and etiology of pain in individual patients via quantitative sensory testing and other personalized medicine approaches; (v) targeting genetic and biochemical mechanisms controlling channel expression using anti-NGF antibodies such as tanezumab or re-purposed drugs such as vorinostat, a histone methyltransferase inhibitor used in the management of T-cell lymphoma, or cercosporamide a MNK 1/2 inhibitor used in treatment of rheumatoid arthritis; (vi) combination therapy using drugs that are selective for different channel types or regulatory processes; (vii) directing preclinical validation work toward the use of human or human-derived tissue samples; and (viii) application of molecular biological approaches such as clustered regularly interspaced short palindromic repeats (CRISPR) technology.
Collapse
Affiliation(s)
- Sascha R A Alles
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Peter A Smith
- Department of Pharmacology, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
20
|
Hadley S, Patil MJ, Pavelkova N, Kollarik M, Taylor-Clark TE. Contribution of tetrodotoxin-sensitive, voltage-gated sodium channels (Na V1) to action potential discharge from mouse esophageal tension mechanoreceptors. Am J Physiol Regul Integr Comp Physiol 2021; 321:R672-R686. [PMID: 34523364 PMCID: PMC8616622 DOI: 10.1152/ajpregu.00199.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/02/2021] [Accepted: 09/07/2021] [Indexed: 11/22/2022]
Abstract
Action potentials depend on voltage-gated sodium channels (NaV1s), which have nine α subtypes. NaV1 inhibition is a target for pathologies involving excitable cells such as pain. However, because NaV1 subtypes are widely expressed, inhibitors may inhibit regulatory sensory systems. Here, we investigated specific NaV1s and their inhibition in mouse esophageal mechanoreceptors-non-nociceptive vagal sensory afferents that are stimulated by low threshold mechanical distension, which regulate esophageal motility. Using single fiber electrophysiology, we found mechanoreceptor responses to esophageal distension were abolished by tetrodotoxin. Single-cell RT-PCR revealed that esophageal-labeled TRPV1-negative vagal neurons expressed multiple tetrodotoxin-sensitive NaV1s: NaV1.7 (almost all neurons) and NaV1.1, NaV1.2, and NaV1.6 (in ∼50% of neurons). Inhibition of NaV1.7, using PF-05089771, had a small inhibitory effect on mechanoreceptor responses to distension. Inhibition of NaV1.1 and NaV1.6, using ICA-121341, had a similar small inhibitory effect. The combination of PF-05089771 and ICA-121341 inhibited but did not eliminate mechanoreceptor responses. Inhibition of NaV1.2, NaV1.6, and NaV1.7 using LSN-3049227 inhibited but did not eliminate mechanoreceptor responses. Thus, all four tetrodotoxin-sensitive NaV1s contribute to action potential initiation from esophageal mechanoreceptors terminals. This is different to those NaV1s necessary for vagal action potential conduction, as demonstrated using GCaMP6s imaging of esophageal vagal neurons during electrical stimulation. Tetrodotoxin-sensitive conduction was abolished in many esophageal neurons by PF-05089771 alone, indicating a critical role of NaV1.7. In summary, multiple NaV1 subtypes contribute to electrical signaling in esophageal mechanoreceptors. Thus, inhibition of individual NaV1s would likely have minimal effect on afferent regulation of esophageal motility.
Collapse
Affiliation(s)
- Stephen Hadley
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Mayur J Patil
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Nikoleta Pavelkova
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Marian Kollarik
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Thomas E Taylor-Clark
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| |
Collapse
|
21
|
Yu L, Tsuji K, Ujihara I, Liu Q, Pavelkova N, Tsujimura T, Inoue M, Meeker S, Nisenbaum E, McDermott JS, Krajewski J, Undem BJ, Kollarik M, Canning BJ. Antitussive effects of Na V 1.7 blockade in Guinea pigs. Eur J Pharmacol 2021; 907:174192. [PMID: 34010618 DOI: 10.1016/j.ejphar.2021.174192] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 05/07/2021] [Accepted: 05/12/2021] [Indexed: 01/25/2023]
Abstract
Our previous studies implicated the voltage-gated sodium channel subtype NaV 1.7 in the transmission of action potentials by the vagal afferent nerves regulating cough and thus identified this channel as a rational therapeutic target for antitussive therapy. But it is presently unclear whether a systemically administered small molecule inhibitor of NaV 1.7 conductance can achieve therapeutic benefit in the absence of side effects on cardiovascular function, gastrointestinal motility or respiration. To this end, we have evaluated the antitussive effects of the NaV 1.7 selective blocker Compound 801 administered systemically in awake guinea pigs or administered topically in anesthetized guinea pigs. We also evaluated the antitussive effects of ambroxol, a low affinity NaV blocker modestly selective for tetrodotoxin resistant NaV subtypes. Both Compound 801 and ambroxol dose-dependently inhibited action potential conduction in guinea pig vagus nerves (assessed by compound potential), with ambroxol nearly 100-fold less potent than the NaV 1.7 selective Compound 801 in this and other NaV 1.7-dependent guinea pig and human tissue-based assays. Both drugs also inhibited citric acid evoked coughing in awake or anesthetized guinea pigs, with potencies supportive of an NaV 1.7-dependent mechanism. Notably, however, the antitussive effects of systemically administered Compound 801 were accompanied by hypotension and respiratory depression. Given the antitussive effects of topically administered Compound 801, we speculate that the likely insurmountable side effects on blood pressure and respiratory drive associated with systemic dosing make topical formulations a viable and perhaps unavoidable therapeutic strategy for targeting NaV 1.7 in cough.
Collapse
Affiliation(s)
- Li Yu
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongii University School of Medicine, Shanghai, 200065, China
| | - Kojun Tsuji
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Izumi Ujihara
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Qi Liu
- Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Baltimore, MD, 21224, USA
| | - Nikoleta Pavelkova
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Takanori Tsujimura
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Makoto Inoue
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Sonya Meeker
- Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Baltimore, MD, 21224, USA
| | - Eric Nisenbaum
- Lilly Research Laboratories, Indianapolis, IN, 46285, USA
| | | | - Jeff Krajewski
- Lilly Research Laboratories, Indianapolis, IN, 46285, USA
| | - Bradley J Undem
- Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Baltimore, MD, 21224, USA
| | - Marian Kollarik
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Brendan J Canning
- Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Baltimore, MD, 21224, USA.
| |
Collapse
|
22
|
Tetrodotoxin: A New Strategy to Treat Visceral Pain? Toxins (Basel) 2021; 13:toxins13070496. [PMID: 34357968 PMCID: PMC8310099 DOI: 10.3390/toxins13070496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 12/13/2022] Open
Abstract
Visceral pain is one of the most common symptoms associated with functional gastrointestinal (GI) disorders. Although the origin of these symptoms has not been clearly defined, the implication of both the central and peripheral nervous systems in visceral hypersensitivity is well established. The role of several pathways in visceral nociception has been explored, as well as the influence of specific receptors on afferent neurons, such as voltage-gated sodium channels (VGSCs). VGSCs initiate action potentials and dysfunction of these channels has recently been associated with painful GI conditions. Current treatments for visceral pain generally involve opioid based drugs, which are associated with important side-effects and a loss of effectiveness or tolerance. Hence, efforts have been intensified to find new, more effective and longer-lasting therapies. The implication of VGSCs in visceral hypersensitivity has drawn attention to tetrodotoxin (TTX), a relatively selective sodium channel blocker, as a possible and promising molecule to treat visceral pain and related diseases. As such, here we will review the latest information regarding this toxin that is relevant to the treatment of visceral pain and the possible advantages that it may offer relative to other treatments, alone or in combination.
Collapse
|
23
|
Tochitsky I, Jo S, Andrews N, Kotoda M, Doyle B, Shim J, Talbot S, Roberson D, Lee J, Haste L, Jordan SM, Levy BD, Bean BP, Woolf CJ. Inhibition of inflammatory pain and cough by a novel charged sodium channel blocker. Br J Pharmacol 2021; 178:3905-3923. [PMID: 33988876 DOI: 10.1111/bph.15531] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/22/2021] [Accepted: 04/27/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Many pain-triggering nociceptor neurons express TRPV1 or TRPA1, cation-selective channels with large pores that enable permeation of QX-314, a cationic analogue of lidocaine. Co-application of QX-314 with TRPV1 or TRPA1 activators can silence nociceptors. In this study, we describe BW-031, a novel more potent cationic sodium channel inhibitor, and test whether its application alone can inhibit pain associated with tissue inflammation and whether this strategy can also inhibit cough. EXPERIMENTAL APPROACH We tested the ability of BW-031 to inhibit pain in three models of tissue inflammation:- inflammation in rat paws produced by complete Freund's adjuvant or by surgical incision and a mouse ultraviolet (UV) burn model. We tested the ability of BW-031 to inhibit cough induced by inhalation of dilute citric acid in guinea pigs. KEY RESULTS BW-031 inhibited Nav 1.7 and Nav 1.1 channels with approximately sixfold greater potency than QX-314 when introduced inside cells. BW-031 inhibited inflammatory pain in all three models tested, producing more effective and longer-lasting inhibition of pain than QX-314 in the mouse UV burn model. BW-031 was effective in reducing cough counts by 78%-90% when applied intratracheally under isoflurane anaesthesia or by aerosol inhalation in guinea pigs with airway inflammation produced by ovalbumin sensitization. CONCLUSION AND IMPLICATIONS BW-031 is a novel cationic sodium channel inhibitor that can be applied locally as a single agent to inhibit inflammatory pain. BW-031 can also effectively inhibit cough in a guinea pig model of citric acid-induced cough, suggesting a new clinical approach to treating cough.
Collapse
Affiliation(s)
- Ivan Tochitsky
- F.M. Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Sooyeon Jo
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Nick Andrews
- F.M. Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Masakazu Kotoda
- F.M. Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Benjamin Doyle
- F.M. Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Jaehoon Shim
- F.M. Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Sebastien Talbot
- F.M. Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, Massachusetts, USA.,Départément de Pharmacologie et Physiologie, Université de Montréal, Montreal, Canada
| | - David Roberson
- F.M. Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Jinbo Lee
- Sage Partner International, Andover, Massachusetts, USA
| | - Louise Haste
- Pharmacology Department, Covance Inc., Huntingdon, UK
| | | | - Bruce D Levy
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bruce P Bean
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Arinze JT, Verhamme KMC, Luik AI, Stricker B, van Meurs JBJ, Brusselle GG. The interrelatedness of chronic cough and chronic pain. Eur Respir J 2021; 57:13993003.02651-2020. [PMID: 33122337 DOI: 10.1183/13993003.02651-2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/16/2020] [Indexed: 12/13/2022]
Abstract
Since chronic cough has common neurobiological mechanisms and pathophysiology with chronic pain, both clinical disorders might be interrelated. Hence, we examined the association between chronic cough and chronic pain in adult subjects in the Rotterdam Study, a large prospective population-based cohort study.Using a standardised questionnaire, chronic pain was defined as pain lasting up to 6 months and grouped into a frequency of weekly/monthly or daily pain. Chronic cough was described as daily coughing for at least 3 months duration. The longitudinal and cross-sectional associations were investigated bi-directionally.Of 7141 subjects in the study, 54% (n=3888) reported chronic pain at baseline. The co-prevalence of daily chronic pain and chronic cough was 4.4%. Chronic cough was more prevalent in subjects with daily and weekly/monthly chronic pain compared with those without chronic pain (13.8% and 10.3% versus 8.2%; p<0.001). After adjustment for potential confounders, prevalent chronic pain was significantly associated with incident chronic cough (OR 1.47, 95% CI 1.08-1.99). The association remained significant in subjects with daily chronic pain (OR 1.49, 95% CI 1.06-2.11) with a similar effect estimate, albeit non-significant in those with weekly/monthly chronic pain (OR 1.43, 95% CI 0.98-2.10). After adjustment for covariables, subjects with chronic cough had a significant risk of developing chronic pain (OR 1.63, 95% CI 1.02-2.62) compared with those without chronic cough.Chronic cough and chronic pain confer risk on each other among adult subjects, indicating that both conditions might share common risk factors and/or pathophysiologic mechanisms.
Collapse
Affiliation(s)
- Johnmary T Arinze
- Dept of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium.,Dept of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Katia M C Verhamme
- Dept of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands.,Dept of Bioanalysis, Ghent University, Ghent, Belgium
| | - Annemarie I Luik
- Dept of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Bruno Stricker
- Dept of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Joyce B J van Meurs
- Dept of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Guy G Brusselle
- Dept of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium.,Dept of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands.,Dept of Respiratory Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
25
|
Goodwin G, McMahon SB. The physiological function of different voltage-gated sodium channels in pain. Nat Rev Neurosci 2021; 22:263-274. [PMID: 33782571 DOI: 10.1038/s41583-021-00444-w] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2021] [Indexed: 02/01/2023]
Abstract
Evidence from human genetic pain disorders shows that voltage-gated sodium channel α-subtypes Nav1.7, Nav1.8 and Nav1.9 are important in the peripheral signalling of pain. Nav1.7 is of particular interest because individuals with Nav1.7 loss-of-function mutations are congenitally insensitive to acute and chronic pain, and there is considerable hope that phenocopying these effects with a pharmacological antagonist will produce a new class of analgesic drug. However, studies in these rare individuals do not reveal how and where voltage-gated sodium channels contribute to pain signalling, which is of critical importance for drug development. More than a decade of research utilizing rodent genetic models and pharmacological tools to study voltage-gated sodium channels in pain has begun to unravel the role of different subtypes. Here, we review the contribution of individual channel subtypes in three key physiological processes necessary for transmission of sensory information to the CNS: transduction of stimuli at peripheral nerve terminals, axonal transmission of action potentials and neurotransmitter release from central terminals. These data suggest that drugs seeking to recapitulate the analgesic effects of loss of function of Nav1.7 will need to be brain-penetrant - which most of those developed to date are not.
Collapse
Affiliation(s)
- George Goodwin
- Pain and Neurorestoration Group, King's College London, London, UK.
| | | |
Collapse
|
26
|
Sapio MR, Vazquez FA, Loydpierson AJ, Maric D, Kim JJ, LaPaglia DM, Puhl HL, Lu VB, Ikeda SR, Mannes AJ, Iadarola MJ. Comparative Analysis of Dorsal Root, Nodose and Sympathetic Ganglia for the Development of New Analgesics. Front Neurosci 2021; 14:615362. [PMID: 33424545 PMCID: PMC7793666 DOI: 10.3389/fnins.2020.615362] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022] Open
Abstract
Interoceptive and exteroceptive signals, and the corresponding coordinated control of internal organs and sensory functions, including pain, are received and orchestrated by multiple neurons within the peripheral, central and autonomic nervous systems. A central aim of the present report is to obtain a molecularly informed basis for analgesic drug development aimed at peripheral rather than central targets. We compare three key peripheral ganglia: nodose, sympathetic (superior cervical), and dorsal root ganglia in the rat, and focus on their molecular composition using next-gen RNA-Seq, as well as their neuroanatomy using immunocytochemistry and in situ hybridization. We obtained quantitative and anatomical assessments of transmitters, receptors, enzymes and signaling pathways mediating ganglion-specific functions. Distinct ganglionic patterns of expression were observed spanning ion channels, neurotransmitters, neuropeptides, G-protein coupled receptors (GPCRs), transporters, and biosynthetic enzymes. The relationship between ganglionic transcript levels and the corresponding protein was examined using immunohistochemistry for select, highly expressed, ganglion-specific genes. Transcriptomic analyses of spinal dorsal horn and intermediolateral cell column (IML), which form the termination of primary afferent neurons and the origin of preganglionic innervation to the SCG, respectively, disclosed pre- and post-ganglionic molecular-level circuits. These multimodal investigations provide insight into autonomic regulation, nodose transcripts related to pain and satiety, and DRG-spinal cord and IML-SCG communication. Multiple neurobiological and pharmacological contexts can be addressed, such as discriminating drug targets and predicting potential side effects, in analgesic drug development efforts directed at the peripheral nervous system.
Collapse
Affiliation(s)
- Matthew R Sapio
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Fernando A Vazquez
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Amelia J Loydpierson
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Jenny J Kim
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Danielle M LaPaglia
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Henry L Puhl
- Section on Neurotransmitter Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Van B Lu
- Section on Neurotransmitter Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Stephen R Ikeda
- Section on Neurotransmitter Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Andrew J Mannes
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Michael J Iadarola
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| |
Collapse
|
27
|
Brozmanova M, Pavelkova N. The Prospect for Potent Sodium Voltage-Gated Channel Blockers to Relieve an Excessive Cough. Physiol Res 2021; 69:S7-S18. [PMID: 32228007 DOI: 10.33549/physiolres.934395] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
An excessive, irritable, productive or non-productive coughing associated with airway inflammation belongs to pathological cough. Increased activation of airway vagal nociceptors in pathological conditions results from dysregulation of the neural pathway that controls cough. A variety of mediators associated with airway inflammation overstimulate these vagal airway fibers including C-fibers leading to hypersensitivity and hyperreactivity. Because current antitussives have limited efficacy and unwanted side effects there is a continual demand for the development of a novel more effective antitussives for a new efficacious and safe cough treatment. Therefore, inhibiting the activity of these vagal C-fibers represents a rational approach to the development of effective antitussive drugs. This may be achieved by blocking inflammatory mediator receptors or by blocking the generator potential associated with the specific ion channels. Because voltage-gated sodium channels (NaVs) are absolutely required for action potentials initiation and conduction irrespective of the stimulus, NaVs become a promising neural target. There is evidence that NaV1.7, 1.8 and 1.9 subtypes are predominantly expressed in airway cough-triggering nerves. The advantage of blocking these NaVs is suppressing C-fiber irrespective to stimuli, but the disadvantage is that by suppressing the nerves is may also block beneficial sensations and neuronal reflex behavior. The concept is that new antitussive drugs would have the benefit of targeting peripheral airway nociceptors without inhibiting the protective cough reflex.
Collapse
Affiliation(s)
- M Brozmanova
- Department of Pathophysiology, Jessenius Faculty of Medicine, Comenius University, Martin, Slovakia.
| | | |
Collapse
|
28
|
Ru F, Pavelkova N, Krajewski JL, McDermott JS, Undem BJ, Kollarik M. Stimulus intensity-dependent recruitment of Na V1 subunits in action potential initiation in nerve terminals of vagal C-fibers innervating the esophagus. Am J Physiol Gastrointest Liver Physiol 2020; 319:G443-G453. [PMID: 32726130 PMCID: PMC7654645 DOI: 10.1152/ajpgi.00122.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We investigated voltage-gated sodium channel (NaV1) subunits that regulate action potential initiation in the nerve terminals of vagal nodose C-fibers innervating the esophagus. Extracellular single fiber recordings were made from the nodose C-fibers, with mechanically sensitive nerve terminals in the isolated innervated guinea pig esophagus. NaV1 inhibitors were selectively delivered to the tissue-containing nerve terminals. Graded esophageal distention was used for mechanical stimulation. The NaV1.7 inhibitor PF-05089771 nearly abolished action potential initiation in response to low levels of esophageal distention but only partially inhibited the response to higher levels of esophageal distention. The PF-05089771-insensitive component of the response progressively increased (up to ≈50%) with increasing esophageal distention and was abolished by tetrodotoxin (TTX). In addition to NaV1.7, nodose C-fiber [transient receptor potential channel-vanilloid subfamily member 1 (TRPV1)-positive] neurons retrogradely labeled from the esophagus expressed mRNA for multiple TTX-sensitive NaV1s. The group NaV1.1, NaV1.2, and NaV1.3 inhibitor ICA-121431 inhibited but did not abolish the PF-05089771-insensitive component of the response to high level of esophageal distention. However, combination of ICA-121431 with compound 801, which also inhibits NaV1.7 and NaV1.6, nearly abolished the response to the high level of esophageal distention. Our data indicate that the action potential initiation in esophageal nodose C-fibers evoked by low (innocuous) levels of esophageal distention is mediated by NaV1.7. However, the response evoked by higher (noxious) levels of esophageal distention has a progressively increasing NaV1.7-independent component that involves multiple TTX-sensitive NaV1s. The stimulus intensity-dependent recruitment of NaV1s may offer novel opportunities for strategic targeting of NaV1 subunits for inhibition of nociceptive signaling in visceral C-fibers.NEW & NOTEWORTHY We report that pharmacologically distinguishable voltage-gated sodium channels (NaV1) mediate action potential initiation at low (innocuous) versus high (noxious) intensity of esophageal distention in nerve terminals of vagal nodose C-fibers. Action potential initiation at low intensity is entirely dependent on NaV1.7; however, additional tetrodotoxin (TTX)-sensitive NaV1s are recruited at higher intensity of distention. This is the first demonstration that NaV1s underlying action potential initiation in visceral C-fibers depend on the intensity of the stimulus.
Collapse
Affiliation(s)
- Fei Ru
- 1Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nikoleta Pavelkova
- 2University of South Florida, Morsani College of Medicine, Tampa, Florida
| | | | | | - Bradley J. Undem
- 1Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marian Kollarik
- 2University of South Florida, Morsani College of Medicine, Tampa, Florida
| |
Collapse
|
29
|
Kowalski CW, Ragozzino FJ, Lindberg JEM, Peterson B, Lugo JM, McLaughlin RJ, Peters JH. Cannabidiol activation of vagal afferent neurons requires TRPA1. J Neurophysiol 2020; 124:1388-1398. [PMID: 32965166 DOI: 10.1152/jn.00128.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Vagal afferent neurons abundantly express excitatory transient receptor potential (TRP) channels, which strongly influence afferent signaling. Cannabinoids have been identified as direct agonists of TRP channels, including TRPA1 and TRPV1, suggesting that exogenous cannabinoids may influence vagal signaling via TRP channel activation. The diverse therapeutic effects of electrical vagus nerve stimulation also result from administration of the nonpsychotropic cannabinoid, cannabidiol (CBD); however, the direct effects of CBD on vagal afferent signaling remain unknown. We investigated actions of CBD on vagal afferent neurons, using calcium imaging and electrophysiology. CBD produced strong excitatory effects in neurons expressing TRPA1. CBD responses were prevented by removal of bath calcium, ruthenium red, and the TRPA1 antagonist A967079, but not the TRPV1 antagonist SB366791, suggesting an essential role for TRPA1. These pharmacological experiments were confirmed using genetic knockouts where TRPA1 KO mice lacked CBD responses, whereas TRPV1 knockout (KO) mice exhibited CBD-induced activation. We also characterized CBD-provoked inward currents at resting potentials in vagal afferents expressing TRPA1 that were absent in TRPA1 KO mice, but persisted in TRPV1 KO mice. CBD also inhibited voltage-activated sodium conductances in A-fiber, but not in C-fiber afferents. To simulate adaptation, resulting from chronic cannabis use, we administered cannabis extract vapor daily for 3 wk. Cannabis exposure reduced the magnitude of CBD responses, likely due to a loss of TRPA1 signaling. Together, these findings detail a novel excitatory action of CBD at vagal afferent neurons, which requires TRPA1 and may contribute to the vagal mimetic effects of CBD and adaptation following chronic cannabis use.NEW & NOTEWORTHY CBD usage has increased with its legalization. The clinical efficacy of CBD has been demonstrated for conditions including some forms of epilepsy, depression, and anxiety that are also treatable by vagus nerve stimulation. We found CBD exhibited direct excitatory effects on vagal afferent neurons that required TRPA1, were augmented by TRPV1, and attenuated following chronic cannabis vapor exposure. These effects may contribute to vagal mimetic effects of CBD and adaptation after chronic cannabis use.
Collapse
Affiliation(s)
- Cody W Kowalski
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Forrest J Ragozzino
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Jonathan E M Lindberg
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - BreeAnne Peterson
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Janelle M Lugo
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Ryan J McLaughlin
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - James H Peters
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| |
Collapse
|
30
|
Mazzone SB, McGarvey L. Mechanisms and Rationale for Targeted Therapies in Refractory and Unexplained Chronic Cough. Clin Pharmacol Ther 2020; 109:619-636. [PMID: 32748976 PMCID: PMC7983941 DOI: 10.1002/cpt.2003] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/24/2020] [Indexed: 12/22/2022]
Abstract
Chronic cough, defined as a cough lasting > 8 weeks, is a common medical condition that exerts a substantial physical, mental, and social burden on patients. A subset of patients with chronic cough are troubled with a cough that persists despite optimal treatment of presumed associated common and uncommon conditions (refractory chronic cough; RCC) or in which no diagnosable cause for cough can be identified despite extensive assessment (unexplained chronic cough; UCC). Many of these patients exhibit clinical features of cough hypersensitivity, including laryngeal paresthesia, hypertussia, and allotussia. Over-the-counter cough remedies are ineffective and can lead to intolerable side effects when used for RCC/UCC, and the lack of approved treatments indicated for these conditions reflects a major unmet need. An increased understanding of the anatomy and neurophysiology of protective and pathologic cough has fostered a robust clinical development pipeline of several targeted therapies for RCC/UCC. This manuscript reviews the mechanisms presumed to underly RCC/UCC together with the rationale and clinical evidence for several targeted therapies currently under clinical investigation, including transient receptor potential channel antagonists, P2X3-receptor antagonists, voltage-gated sodium channel blockers, neuromodulators, and neurokinin-1-receptor antagonists. Finally, we provide an overview of targets that have been investigated in preclinical models of cough and other airway diseases that may hold future promise for clinical studies in RCC/UCC. Development of targeted therapies with different sites of action may foster a precision medicine approach to treat this heterogeneous, underserved patient population.
Collapse
Affiliation(s)
- Stuart B Mazzone
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Victoria, Australia
| | - Lorcan McGarvey
- Wellcome-Wolfson Institute of Experimental Medicine, Queen's University Belfast, Belfast, UK
| |
Collapse
|
31
|
Wang JT, Zheng YM, Chen YT, Gu M, Gao ZB, Nan FJ. Discovery of aryl sulfonamide-selective Nav1.7 inhibitors with a highly hydrophobic ethanoanthracene core. Acta Pharmacol Sin 2020; 41:293-302. [PMID: 31316182 PMCID: PMC7471454 DOI: 10.1038/s41401-019-0267-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/30/2019] [Indexed: 01/19/2023] Open
Abstract
Nav1.7 channels are mainly distributed in the peripheral nervous system. Blockade of Nav1.7 channels with small-molecule inhibitors in humans might provide pain relief without affecting the central nervous system. Based on the facts that many reported Nav1.7-selective inhibitors contain aryl sulfonamide fragments, as well as a tricyclic antidepressant, maprotiline, has been found to inhibit Nav1.7 channels, we designed and synthesized a series of compounds with ethanoanthracene and aryl sulfonamide moieties. Their inhibitory activity on sodium channels were detected with electrophysiological techniques. We found that compound 10o potently inhibited Nav1.7 channels stably expressed in HEK293 cells (IC50 = 0.64 ± 0.30 nmol/L) and displayed a high Nav1.7/Nav1.5 selectivity. In mouse small-sized dorsal root ganglion neurons, compound 10o (10, 100 nmol/L) dose-dependently decreased the sodium currents and dramatically suppressed depolarizing current-elicited neuronal discharge. Preliminary in vivo experiments showed that compound 10o possessed good analgesic activity: in a mouse visceral pain model, administration of compound 10o (30−100 mg/kg, i.p.) effectively and dose-dependently suppressed acetic acid-induced writhing.
Collapse
|
32
|
Undem BJ, Sun H. Molecular/Ionic Basis of Vagal Bronchopulmonary C-Fiber Activation by Inflammatory Mediators. Physiology (Bethesda) 2020; 35:57-68. [PMID: 31799905 PMCID: PMC6985783 DOI: 10.1152/physiol.00014.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 12/11/2022] Open
Abstract
Stimulation of bronchopulmonary vagal afferent C fibers by inflammatory mediators can lead to coughing, chest tightness, and changes in breathing pattern, as well as reflex bronchoconstriction and secretions. These responses serve a defensive function in healthy lungs but likely contribute to many of the signs and symptoms of inflammatory airway diseases. A better understanding of the mechanisms underlying the activation of bronchopulmonary C-fiber terminals may lead to novel therapeutics that would work in an additive or synergic manner with existing anti-inflammatory strategies.
Collapse
Affiliation(s)
| | - Hui Sun
- Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
33
|
Brozmanova M, Svajdova S, Pavelkova N, Muroi Y, Undem B, Kollarik M. The voltage-gated sodium channel NaV1.8 blocker A-803467 inhibits cough in the guinea pig. Respir Physiol Neurobiol 2019; 270:103267. [DOI: 10.1016/j.resp.2019.103267] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/17/2019] [Accepted: 08/05/2019] [Indexed: 10/26/2022]
|
34
|
Mulcahy JV, Pajouhesh H, Beckley JT, Delwig A, Bois JD, Hunter JC. Challenges and Opportunities for Therapeutics Targeting the Voltage-Gated Sodium Channel Isoform Na V1.7. J Med Chem 2019; 62:8695-8710. [PMID: 31012583 PMCID: PMC6786914 DOI: 10.1021/acs.jmedchem.8b01906] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Voltage-gated sodium ion channel subtype 1.7 (NaV1.7) is a high interest target for the discovery of non-opioid analgesics. Compelling evidence from human genetic data, particularly the finding that persons lacking functional NaV1.7 are insensitive to pain, has spurred considerable effort to develop selective inhibitors of this Na+ ion channel target as analgesic medicines. Recent clinical setbacks and disappointing performance of preclinical compounds in animal pain models, however, have led to skepticism around the potential of selective NaV1.7 inhibitors as human therapeutics. In this Perspective, we discuss the attributes and limitations of recently disclosed investigational drugs targeting NaV1.7 and review evidence that, by better understanding the requirements for selectivity and target engagement, the opportunity to deliver effective analgesic medicines targeting NaV1.7 endures.
Collapse
Affiliation(s)
- John V. Mulcahy
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - Hassan Pajouhesh
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - Jacob T. Beckley
- SiteOne Therapeutics, 351 Evergreen Drive, Suite B1, Bozeman, MT 59715
| | - Anton Delwig
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - J. Du Bois
- Stanford University, Lokey Chemistry and Biology, 337 Campus Drive, Stanford, CA 94305
| | - John C. Hunter
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| |
Collapse
|
35
|
Bisphenol A Regulates Sodium Ramp Currents in Mouse Dorsal Root Ganglion Neurons and Increases Nociception. Sci Rep 2019; 9:10306. [PMID: 31312012 PMCID: PMC6635372 DOI: 10.1038/s41598-019-46769-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 07/05/2019] [Indexed: 12/02/2022] Open
Abstract
17β-Estradiol mediates the sensitivity to pain and is involved in sex differences in nociception. The widespread environmental disrupting chemical bisphenol A (BPA) has estrogenic activity, but its implications in pain are mostly unknown. Here we show that treatment of male mice with BPA (50 µg/kg/day) during 8 days, decreases the latency to pain behavior in response to heat, suggesting increased pain sensitivity. We demonstrate that incubation of dissociated dorsal root ganglia (DRG) nociceptors with 1 nM BPA increases the frequency of action potential firing. SCN9A encodes the voltage-gated sodium channel Nav1.7, which is present in DRG nociceptors and is essential in pain signaling. Nav1.7 and other voltage-gated sodium channels in mouse DRG are considered threshold channels because they produce ramp currents, amplifying small depolarizations and enhancing electrical activity. BPA increased Nav-mediated ramp currents elicited with slow depolarizations. Experiments using pharmacological tools as well as DRG from ERβ−/− mice indicate that this BPA effect involves ERα and phosphoinositide 3-kinase. The mRNA expression and biophysical properties other than ramp currents of Nav channels, were unchanged by BPA. Our data suggest that BPA at environmentally relevant doses affects the ability to detect noxious stimuli and therefore should be considered when studying the etiology of pain conditions.
Collapse
|
36
|
Masi EB, Levy T, Tsaava T, Bouton CE, Tracey KJ, Chavan SS, Zanos TP. Identification of hypoglycemia-specific neural signals by decoding murine vagus nerve activity. Bioelectron Med 2019; 5:9. [PMID: 32232099 PMCID: PMC7098244 DOI: 10.1186/s42234-019-0025-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 06/06/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Glucose is a crucial energy source. In humans, it is the primary sugar for high energy demanding cells in brain, muscle and peripheral neurons. Deviations of blood glucose levels from normal levels for an extended period of time is dangerous or even fatal, so regulation of blood glucose levels is a biological imperative. The vagus nerve, comprised of sensory and motor fibres, provides a major anatomical substrate for regulating metabolism. While prior studies have implicated the vagus nerve in the neurometabolic interface, its specific role in either the afferent or efferent arc of this reflex remains elusive. METHODS Here we use recently developed methods to isolate and decode specific neural signals acquired from the surface of the vagus nerve in BALB/c wild type mice to identify those that respond robustly to hypoglycemia. We also attempted to decode neural signals related to hyperglycemia. In addition to wild type mice, we analyzed the responses to acute hypo- and hyperglycemia in transient receptor potential cation channel subfamily V member 1 (TRPV1) cell depleted mice. The decoding algorithm uses neural signals as input and reconstructs blood glucose levels. RESULTS Our algorithm was able to reconstruct the blood glucose levels with high accuracy (median error 18.6 mg/dl). Hyperglycemia did not induce robust vagus nerve responses, and deletion of TRPV1 nociceptors attenuated the hypoglycemia-dependent vagus nerve signals. CONCLUSION These results provide insight to the sensory vagal signaling that encodes hypoglycemic states and suggest a method to measure blood glucose levels by decoding nerve signals. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
| | - Todd Levy
- 2Institute of Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030 USA
| | - Tea Tsaava
- 2Institute of Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030 USA
| | - Chad E Bouton
- 2Institute of Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030 USA
| | - Kevin J Tracey
- Zucker School of Medicine at Hofstra/Northwell, Heampstead, NY USA
- 2Institute of Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030 USA
| | - Sangeeta S Chavan
- Zucker School of Medicine at Hofstra/Northwell, Heampstead, NY USA
- 2Institute of Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030 USA
| | - Theodoros P Zanos
- Zucker School of Medicine at Hofstra/Northwell, Heampstead, NY USA
- 2Institute of Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030 USA
| |
Collapse
|
37
|
Kollarik M, Sun H, Herbstsomer RA, Ru F, Kocmalova M, Meeker SN, Undem BJ. Different role of TTX-sensitive voltage-gated sodium channel (Na V 1) subtypes in action potential initiation and conduction in vagal airway nociceptors. J Physiol 2019; 596:1419-1432. [PMID: 29435993 DOI: 10.1113/jp275698] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 01/23/2018] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The action potential initiation in the nerve terminals and its subsequent conduction along the axons of afferent nerves are not necessarily dependent on the same voltage-gated sodium channel (NaV 1) subunits. The action potential initiation in jugular C-fibres within airway tissues is not blocked by TTX; nonetheless, conduction of action potentials along the vagal axons of these nerves is often dependent on TTX-sensitive channels. This is not the case for nodose airway Aδ-fibres and C-fibres, where both action potential initiation and conduction is abolished by TTX or selective NaV 1.7 blockers. The difference between the initiation of action potentials within the airways vs. conduction along the axons should be considered when developing NaV 1 blocking drugs for topical application to the respiratory tract. ABSTRACT The action potential (AP) initiation in the nerve terminals and its subsequent AP conduction along the axons do not necessarily depend on the same subtypes of voltage-gated sodium channels (NaV 1s). We evaluated the role of TTX-sensitive and TTX-resistant NaV 1s in vagal afferent nociceptor nerves derived from jugular and nodose ganglia innervating the respiratory system. Single cell RT-PCR was performed on vagal afferent neurons retrogradely labelled from the guinea pig trachea. Almost all of the jugular neurons expressed the TTX-sensitive channel NaV 1.7 along with TTX-resistant NaV 1.8 and NaV 1.9. Tracheal nodose neurons also expressed NaV 1.7 but, less frequently, NaV 1.8 and NaV 1.9. NaV 1.6 were expressed in ∼40% of the jugular and 25% of nodose tracheal neurons. Other NaV 1 α subunits were only rarely expressed. Single fibre recordings were made from the vagal nodose and jugular nerve fibres innervating the trachea or lung in the isolated perfused vagally-innervated preparations that allowed for selective drug delivery to the nerve terminal compartment (AP initiation) or to the desheathed vagus nerve (AP conduction). AP initiation in jugular C-fibres was unaffected by TTX, although it was inhibited by NaV 1.8 blocker (PF-01247324) and abolished by combination of TTX and PF-01247324. However, AP conduction in the majority of jugular C-fibres was abolished by TTX. By contrast, both AP initiation and conduction in nodose nociceptors was abolished by TTX or selective NaV 1.7 blockers. Distinction between the effect of a drug with respect to inhibiting AP in the nerve terminals within the airways vs. at conduction sites along the vagus nerve is relevant to therapeutic strategies involving inhaled NaV 1 blocking drugs.
Collapse
Affiliation(s)
- M Kollarik
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pathophysiology, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - H Sun
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - R A Herbstsomer
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - F Ru
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - M Kocmalova
- Department of Pharmacology, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - S N Meeker
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - B J Undem
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
38
|
Nicolas S, Zoukimian C, Bosmans F, Montnach J, Diochot S, Cuypers E, De Waard S, Béroud R, Mebs D, Craik D, Boturyn D, Lazdunski M, Tytgat J, De Waard M. Chemical Synthesis, Proper Folding, Na v Channel Selectivity Profile and Analgesic Properties of the Spider Peptide Phlotoxin 1. Toxins (Basel) 2019; 11:toxins11060367. [PMID: 31234412 PMCID: PMC6628435 DOI: 10.3390/toxins11060367] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/11/2019] [Accepted: 06/16/2019] [Indexed: 12/19/2022] Open
Abstract
Phlotoxin-1 (PhlTx1) is a peptide previously identified in tarantula venom (Phlogius species) that belongs to the inhibitory cysteine-knot (ICK) toxin family. Like many ICK-based spider toxins, the synthesis of PhlTx1 appears particularly challenging, mostly for obtaining appropriate folding and concomitant suitable disulfide bridge formation. Herein, we describe a procedure for the chemical synthesis and the directed sequential disulfide bridge formation of PhlTx1 that allows for a straightforward production of this challenging peptide. We also performed extensive functional testing of PhlTx1 on 31 ion channel types and identified the voltage-gated sodium (Nav) channel Nav1.7 as the main target of this toxin. Moreover, we compared PhlTx1 activity to 10 other spider toxin activities on an automated patch-clamp system with Chinese Hamster Ovary (CHO) cells expressing human Nav1.7. Performing these analyses in reproducible conditions allowed for classification according to the potency of the best natural Nav1.7 peptide blockers. Finally, subsequent in vivo testing revealed that intrathecal injection of PhlTx1 reduces the response of mice to formalin in both the acute pain and inflammation phase without signs of neurotoxicity. PhlTx1 is thus an interesting toxin to investigate Nav1.7 involvement in cellular excitability and pain.
Collapse
Affiliation(s)
- Sébastien Nicolas
- Institut du Thorax, Inserm UMR 1087/CNRS UMR 6291, LabEx "Ion Channels, Science & Therapeutics", F-44007 Nantes, France.
| | - Claude Zoukimian
- Smartox Biotechnology, 6 rue des Platanes, F-38120 Saint-Egrève, France.
- Department of Molecular Chemistry, Univ. Grenoble Alpes, CNRS, 570 rue de la chimie, CS 40700, 38000 Grenoble, France.
| | - Frank Bosmans
- Faculty of Medicine and Health Sciences, Department of Basic and Applied Medical Sciences, 9000 Gent, Belgium.
- Toxicology and Pharmacology, University of Leuven, Campus Gasthuisberg, P.O. Box 922, Herestraat 49, 3000 Leuven, Belgium.
| | - Jérôme Montnach
- Institut du Thorax, Inserm UMR 1087/CNRS UMR 6291, LabEx "Ion Channels, Science & Therapeutics", F-44007 Nantes, France.
| | - Sylvie Diochot
- Université Côte d'Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des lucioles, 6560 Valbonne, France.
| | - Eva Cuypers
- Toxicology and Pharmacology, University of Leuven, Campus Gasthuisberg, P.O. Box 922, Herestraat 49, 3000 Leuven, Belgium.
| | - Stephan De Waard
- Institut du Thorax, Inserm UMR 1087/CNRS UMR 6291, LabEx "Ion Channels, Science & Therapeutics", F-44007 Nantes, France.
| | - Rémy Béroud
- Smartox Biotechnology, 6 rue des Platanes, F-38120 Saint-Egrève, France.
| | - Dietrich Mebs
- Institute of Legal Medicine, University of Frankfurt, Kennedyallee 104, Frankfurt, Germany.
| | - David Craik
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, Australia.
| | - Didier Boturyn
- Department of Molecular Chemistry, Univ. Grenoble Alpes, CNRS, 570 rue de la chimie, CS 40700, 38000 Grenoble, France.
| | - Michel Lazdunski
- Université Côte d'Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des lucioles, 6560 Valbonne, France.
| | - Jan Tytgat
- Toxicology and Pharmacology, University of Leuven, Campus Gasthuisberg, P.O. Box 922, Herestraat 49, 3000 Leuven, Belgium.
| | - Michel De Waard
- Institut du Thorax, Inserm UMR 1087/CNRS UMR 6291, LabEx "Ion Channels, Science & Therapeutics", F-44007 Nantes, France.
- Smartox Biotechnology, 6 rue des Platanes, F-38120 Saint-Egrève, France.
| |
Collapse
|
39
|
|
40
|
Bennett DL, Clark AJ, Huang J, Waxman SG, Dib-Hajj SD. The Role of Voltage-Gated Sodium Channels in Pain Signaling. Physiol Rev 2019; 99:1079-1151. [DOI: 10.1152/physrev.00052.2017] [Citation(s) in RCA: 256] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Acute pain signaling has a key protective role and is highly evolutionarily conserved. Chronic pain, however, is maladaptive, occurring as a consequence of injury and disease, and is associated with sensitization of the somatosensory nervous system. Primary sensory neurons are involved in both of these processes, and the recent advances in understanding sensory transduction and human genetics are the focus of this review. Voltage-gated sodium channels (VGSCs) are important determinants of sensory neuron excitability: they are essential for the initial transduction of sensory stimuli, the electrogenesis of the action potential, and neurotransmitter release from sensory neuron terminals. Nav1.1, Nav1.6, Nav1.7, Nav1.8, and Nav1.9 are all expressed by adult sensory neurons. The biophysical characteristics of these channels, as well as their unique expression patterns within subtypes of sensory neurons, define their functional role in pain signaling. Changes in the expression of VGSCs, as well as posttranslational modifications, contribute to the sensitization of sensory neurons in chronic pain states. Furthermore, gene variants in Nav1.7, Nav1.8, and Nav1.9 have now been linked to human Mendelian pain disorders and more recently to common pain disorders such as small-fiber neuropathy. Chronic pain affects one in five of the general population. Given the poor efficacy of current analgesics, the selective expression of particular VGSCs in sensory neurons makes these attractive targets for drug discovery. The increasing availability of gene sequencing, combined with structural modeling and electrophysiological analysis of gene variants, also provides the opportunity to better target existing therapies in a personalized manner.
Collapse
Affiliation(s)
- David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Alex J. Clark
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Jianying Huang
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Stephen G. Waxman
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Sulayman D. Dib-Hajj
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
41
|
Patil MJ, Sun H, Ru F, Meeker S, Undem BJ. Targeting C-fibers for peripheral acting anti-tussive drugs. Pulm Pharmacol Ther 2019; 56:15-19. [PMID: 30872160 DOI: 10.1016/j.pupt.2019.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 01/09/2023]
Abstract
Activation of vagal C-fibers is likely involved in some types of pathological coughing, especially coughing that is associated with airway inflammation. This is because stimulation of vagal C-fibers leads to strong urge to cough sensations, and because C-fiber terminals can be strongly activated by mediators associated with airway inflammation. The most direct manner in which a given mediator can activate a C-fiber terminal is through interacting with its receptor expressed in the terminal membrane. The agonist-receptor interaction then must lead to the opening (or potentially closing) of ion channels that lead to a membrane depolarization. This depolarization is referred to as a generator potential. If, and only if, the generator potential reaches the voltage necessary to activate voltage-gated sodium channels, action potentials are initiated and conducted to the central terminals within the CNS. Therefore, there are three target areas to block the inflammatory mediator induced activation of C-fiber terminals. First, at the level of the mediator-receptor interaction, secondly at the level of the generator potential, and third at the level of the voltage-gated sodium channels. Here we provide a brief overview of each of these therapeutic strategies.
Collapse
Affiliation(s)
- Mayur J Patil
- Department of Medicine, Johns Hopkins University School of Medicine, USA
| | - Hui Sun
- Department of Medicine, Johns Hopkins University School of Medicine, USA
| | - Fei Ru
- Department of Medicine, Johns Hopkins University School of Medicine, USA
| | - Sonya Meeker
- Department of Medicine, Johns Hopkins University School of Medicine, USA
| | - Bradley J Undem
- Department of Medicine, Johns Hopkins University School of Medicine, USA.
| |
Collapse
|
42
|
Regulation of Cough by Voltage-Gated Sodium Channels in Airway Sensory Nerves. ACTA MEDICA MARTINIANA 2019. [DOI: 10.2478/acm-2018-0012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Abstract
Chronic cough is a significant clinical problem in many patients. Current cough suppressant therapies are largely ineffective and have many dangerous adverse effects. Therefore, the identification of novel therapeutic targets and strategies for chronic cough treatment may lead to development of novel effective antitussive therapies with fewer adverse effects. The experimental research in the area of airway sensory nerves suggests that there are two main vagal afferent nerve subtypes that can directly activate cough – extrapulmonary airway C-fibres and Aδ-fibres (described as cough receptors) innervating the trachea. There are different receptors on the vagal nerve terminals that can trigger coughing, such as TRP channels and P2X2/3 receptors. However, in many patients with chronic respiratory diseases multiple activation of these receptors could be involved and it is also difficult to target these receptors. For that reason, a strategy that would inhibit cough-triggering nerve afferents regardless of activated receptors would be of great benefit. In recent years huge progress in understanding of voltage-gated sodium channels (NaVs) leads to a hypothesis that selective targeting of NaVs in airways may represent an effective treatment of pathological cough. The NaVs (NaV1.1 – NaV1.9) are essential for initiation and conduction of action potentials in these nerve fibres. Effective blocking of NaVs will prevent communication between airways and central nervous system and that would inhibit provoked cough irrespective to stimuli. This review provides an overview of airway afferent nerve subtypes that have been described in respiratory tract of human and in animal models. Moreover, the review highlights the current knowledge about cough, the sensory nerves involved in cough, and the voltage-gated sodium channels as a novel neural target in regulation of cough.
Collapse
|
43
|
Lidocaine, a Non–selective Inhibitor of Voltage-Gated Sodium Channels, Blocks Chemically-Induced Cough in Awake Naïve Guinea Pigs. ADVANCES IN PULMONARY MEDICINE: RESEARCH AND INNOVATIONS 2019; 1160:1-9. [DOI: 10.1007/5584_2018_326] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
44
|
Xie MX, Yang J, Pang RP, Zeng WA, Ouyang HD, Liu YQ, Liu XG. Bulleyaconitine A attenuates hyperexcitability of dorsal root ganglion neurons induced by spared nerve injury: The role of preferably blocking Nav1.7 and Nav1.3 channels. Mol Pain 2018; 14:1744806918778491. [PMID: 29783906 PMCID: PMC5967161 DOI: 10.1177/1744806918778491] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background Oral administration of Bulleyaconitine A, an extracted diterpenoid alkaloid from Aconitum bulleyanum plants, is effective for treating chronic pain in rats and in human patients, but the underlying mechanisms are poorly understood. Results As the hyperexcitability of dorsal root ganglion neurons resulting from the upregulation of voltage-gated sodium (Nav) channels has been proved critical for development of chronic pain, we tested the effects of Bulleyaconitine A on Nav channels in rat spared nerve injury model of neuropathic pain. We found that Bulleyaconitine A at 5 nM increased the threshold of action potentials and reduced the firing rate of dorsal root ganglion neurons in spared nerve injury rats but not in sham rats. Bulleyaconitine A preferably blocked tetrodotoxin-sensitive Nav channels over tetrodotoxin-resistant ones in dorsal root ganglion neurons of spared nerve injury rats. Bulleyaconitine A was more potent for blocking Nav1.3 and Nav1.7 than Nav1.8 in cell lines. The half maximal inhibitory concentration (IC50) values for resting Nav1.3, Nav1.7, and Nav1.8 were 995.6 ± 139.1 nM, 125.7 ± 18.6 nM, and 151.2 ± 15.4 μM, respectively, which were much higher than those for inactivated Nav1.3 (20.3 ± 3.4 pM), Nav1.7 (132.9 ± 25.5 pM), and Nav1.8 (18.0 ± 2.5 μM). The most profound use-dependent blocking effect of Bulleyaconitine A was observed on Nav1.7, less on Nav1.3, and least on Nav1.8 at IC50 concentrations. Bulleyaconitine A facilitated the inactivation of Nav channels in each subtype. Conclusions Preferably blocking tetrodotoxin-sensitive Nav1.7 and Nav1.3 in dorsal root ganglion neurons may contribute to Bulleyaconitine A’s antineuropathic pain effect.
Collapse
Affiliation(s)
- Man-Xiu Xie
- 1 Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jie Yang
- 2 Department of Physiology, Pain Research Center, Zhongshan School of Medicine of Sun Yat-Sen University, Guangzhou, China
| | - Rui-Ping Pang
- 2 Department of Physiology, Pain Research Center, Zhongshan School of Medicine of Sun Yat-Sen University, Guangzhou, China.,3 Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong, China
| | - Wei-An Zeng
- 1 Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Han-Dong Ouyang
- 1 Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yan-Qing Liu
- 4 Department of Pain Medicine, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
| | - Xian-Guo Liu
- 2 Department of Physiology, Pain Research Center, Zhongshan School of Medicine of Sun Yat-Sen University, Guangzhou, China.,3 Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong, China
| |
Collapse
|
45
|
Jurcakova D, Ru F, Kollarik M, Sun H, Krajewski J, Undem BJ. Voltage-Gated Sodium Channels Regulating Action Potential Generation in Itch-, Nociceptive-, and Low-Threshold Mechanosensitive Cutaneous C-Fibers. Mol Pharmacol 2018; 94:1047-1056. [DOI: 10.1124/mol.118.112839] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/20/2018] [Indexed: 01/25/2023] Open
|
46
|
Abstract
Fever-associated seizures or epilepsy (FASE) is primarily characterised by the occurrence of a seizure or epilepsy usually accompanied by a fever. It is common in infants and children, and generally includes febrile seizures (FS), febrile seizures plus (FS+), Dravet syndrome (DS) and genetic epilepsy with febrile seizures plus (GEFSP). The aetiology of FASE is unclear. Genetic factors may play crucial roles in FASE. Mutations in certain genes may cause a wide spectrum of phenotypical overlap ranging from isolated FS, FS+ and GEFSP to DS. Synapse-associated proteins, postsynaptic GABAA receptor, and sodium channels play important roles in synaptic transmission. Mutations in these genes may involve in the pathogenesis of FASE. Elevated temperature promotes synaptic vesicle (SV) recycling and enlarges SV size, which may enhance synaptic transmission and contribute to FASE occurring. This review provides an overview of the loci, genes, underlying pathogenesis and the fever-inducing effect of FASE. It may provide a more comprehensive understanding of pathogenesis and contribute to the clinical diagnosis of FASE.
Collapse
|
47
|
Kocmalova M, Joskova M, Franova S, Banovcin P, Sutovska M. Airway Defense Control Mediated via Voltage-Gated Sodium Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 921:71-80. [PMID: 27161110 DOI: 10.1007/5584_2016_244] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Expression of voltage-gated sodium channels (Nav) takes place in the airways and the role of Nav1.7 and Nav1.8 in the control of airway's defense reflexes has been confirmed. The activation of Nav channels is crucial for cough initiation and airway smooth muscle reactivity, but it is unknown whether these channels regulate ciliary beating. This study evaluated the involvement of Nav1.7 and Nav1.8 channels in the airway defense mechanisms using their pharmacological blockers in healthy guinea pigs and in the experimental allergic asthma model. Asthma was modeled by ovalbumin sensitization over a period of 21 days. Blockade of Nav1.7 channels significantly decreased airway smooth muscle reactivity in vivo, the number of cough efforts, and the cilia beat frequency in healthy animals. In the allergic asthma model, blockade of Nav1.8 efficiently relieved symptoms of asthma, without adversely affecting cilia beat frequency. The study demonstrates that Nav1.8 channel antagonism has a potential to alleviate cough and bronchial hyperreactivity in asthma.
Collapse
Affiliation(s)
- M Kocmalova
- Department of Pharmacology, Division of Respirology, BioMed Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 4C Mala Hora St, 036 01, Martin, Slovakia
| | - M Joskova
- Department of Pharmacology, Division of Respirology, BioMed Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 4C Mala Hora St, 036 01, Martin, Slovakia.
| | - S Franova
- Department of Pharmacology, Division of Respirology, BioMed Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 4C Mala Hora St, 036 01, Martin, Slovakia
| | - P Banovcin
- Department of Children and Adolescents, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, and Martin University Hospital, Martin, Slovakia
| | - M Sutovska
- Department of Pharmacology, Division of Respirology, BioMed Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 4C Mala Hora St, 036 01, Martin, Slovakia
| |
Collapse
|
48
|
Blocking voltage-gated sodium channels as a strategy to suppress pathological cough. Pulm Pharmacol Ther 2017; 47:38-41. [DOI: 10.1016/j.pupt.2017.05.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/10/2017] [Accepted: 05/12/2017] [Indexed: 12/31/2022]
|
49
|
Mazzone SB, Undem BJ. Vagal Afferent Innervation of the Airways in Health and Disease. Physiol Rev 2017; 96:975-1024. [PMID: 27279650 DOI: 10.1152/physrev.00039.2015] [Citation(s) in RCA: 339] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Vagal sensory neurons constitute the major afferent supply to the airways and lungs. Subsets of afferents are defined by their embryological origin, molecular profile, neurochemistry, functionality, and anatomical organization, and collectively these nerves are essential for the regulation of respiratory physiology and pulmonary defense through local responses and centrally mediated neural pathways. Mechanical and chemical activation of airway afferents depends on a myriad of ionic and receptor-mediated signaling, much of which has yet to be fully explored. Alterations in the sensitivity and neurochemical phenotype of vagal afferent nerves and/or the neural pathways that they innervate occur in a wide variety of pulmonary diseases, and as such, understanding the mechanisms of vagal sensory function and dysfunction may reveal novel therapeutic targets. In this comprehensive review we discuss historical and state-of-the-art concepts in airway sensory neurobiology and explore mechanisms underlying how vagal sensory pathways become dysfunctional in pathological conditions.
Collapse
Affiliation(s)
- Stuart B Mazzone
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, Australia; and Department of Medicine, Johns Hopkins University Medical School, Asthma & Allergy Center, Baltimore, Maryland
| | - Bradley J Undem
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, Australia; and Department of Medicine, Johns Hopkins University Medical School, Asthma & Allergy Center, Baltimore, Maryland
| |
Collapse
|
50
|
Chung KF. Advances in mechanisms and management of chronic cough: The Ninth London International Cough Symposium 2016. Pulm Pharmacol Ther 2017; 47:2-8. [PMID: 28216388 DOI: 10.1016/j.pupt.2017.02.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 02/12/2017] [Indexed: 12/30/2022]
Abstract
At the Ninth London International Cough Symposium held in June 2016, advances in chronic cough were presented. Chronic cough has been labelled as a cough hypersensitivity syndrome (CHS) with neuroinflammatory mechanisms likely to be the underlying mechanisms. The concept is that there is a stage of peripheral sensitisation induced by inflammatory factors setting up the scene for a central component that can be visualised by functional magnetic resonance imaging. There has also been progress in assessing CHS patients in the clinic in terms of measuring cough, with an increasing interest in assessing different types of cough associated with respiratory diseases such as asthma, COPD, bronchiectasis and pulmonary fibrosis. There is an emerging area of new antitussives in the form of neuromodulators. These advances have been paralleled by improvements in the management of patients with chronic cough. However, more work is needed but the future looks promising.
Collapse
Affiliation(s)
- Kian Fan Chung
- National Heart & Lung Institute, Imperial College London, UK; Biomedical Research Unit, Royal Brompton & Harefield NHS Trust, London, UK.
| |
Collapse
|