1
|
Vasylyev DV, Zhao P, Schulman BR, Waxman SG. Interplay of Nav1.8 and Nav1.7 channels drives neuronal hyperexcitability in neuropathic pain. J Gen Physiol 2024; 156:e202413596. [PMID: 39378238 PMCID: PMC11465073 DOI: 10.1085/jgp.202413596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/09/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
While voltage-gated sodium channels Nav1.7 and Nav1.8 both contribute to electrogenesis in dorsal root ganglion (DRG) neurons, details of their interactions have remained unexplored. Here, we studied the functional contribution of Nav1.8 in DRG neurons using a dynamic clamp to express Nav1.7L848H, a gain-of-function Nav1.7 mutation that causes inherited erythromelalgia (IEM), a human genetic model of neuropathic pain, and demonstrate a profound functional interaction of Nav1.8 with Nav1.7 close to the threshold for AP generation. At the voltage threshold of -21.9 mV, we observed that Nav1.8 channel open-probability exceeded Nav1.7WT channel open-probability ninefold. Using a kinetic model of Nav1.8, we showed that a reduction of Nav1.8 current by even 25-50% increases rheobase and reduces firing probability in small DRG neurons expressing Nav1.7L848H. Nav1.8 subtraction also reduces the amplitudes of subthreshold membrane potential oscillations in these cells. Our results show that within DRG neurons that express peripheral sodium channel Nav1.7, the Nav1.8 channel amplifies excitability at a broad range of membrane voltages with a predominant effect close to the AP voltage threshold, while Nav1.7 plays a major role at voltages closer to resting membrane potential. Our data show that dynamic-clamp reduction of Nav1.8 conductance by 25-50% can reverse hyperexcitability of DRG neurons expressing a gain-of-function Nav1.7 mutation that causes pain in humans and suggests, more generally, that full inhibition of Nav1.8 may not be required for relief of pain due to DRG neuron hyperexcitability.
Collapse
Affiliation(s)
- Dmytro V. Vasylyev
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Peng Zhao
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Betsy R. Schulman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Stephen G. Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
2
|
Gualdani R, Barbeau S, Yuan JH, Jacobs DS, Gailly P, Dib-Hajj SD, Waxman SG. TRPV1 corneal neuralgia mutation: Enhanced pH response, bradykinin sensitization, and capsaicin desensitization. Proc Natl Acad Sci U S A 2024; 121:e2406186121. [PMID: 39226353 PMCID: PMC11406256 DOI: 10.1073/pnas.2406186121] [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: 03/26/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024] Open
Abstract
The factors that contribute to pain after nerve injury remain incompletely understood. Laser-assisted in situ keratomileusis (LASIK) and photorefractive keratectomy (PRK) are common surgical techniques to correct refractive errors. After LASIK or PRK, a subset of patients suffers intense and persistent pain, of unknown origin, described by patients as feeling like shards of glass in their eye. Here, we evaluated a TRPV1 variant, p.V527M, found in a 49-y-old woman who developed corneal pain after LASIK and subsequent PRK enhancement, reporting an Ocular Surface Disease Index score of 100. Using patch-clamp and Ca2+ imaging, we found that the V527M mutation enhances the response to acidic pH. Increasing proton concentration induced a stronger leftward shift in the activation curve of V527M compared to WT, resulting in channel activity of the mutant in acidic pH at more physiological membrane potentials. Finally, comparing the responses to consecutive applications of different agonists, we found in V527M channels a reduced capsaicin-induced desensitization and increased sensitization by the arachidonic acid metabolite 12-hydroxyeicosatetraenoic acid (12-HETE). We hypothesize that the increased response in V527M channels to protons and enhanced sensitization by 12-HETE, two inflammatory mediators released in the cornea after tissue damage, may contribute to the pathogenesis of corneal neuralgia after refractive surgery.
Collapse
Affiliation(s)
- Roberta Gualdani
- Laboratory of Cell Physiology, Institute of Neuroscience, Université catholique de Louvain, BrusselsB-1200, Belgium
| | - Solène Barbeau
- Laboratory of Cell Physiology, Institute of Neuroscience, Université catholique de Louvain, BrusselsB-1200, Belgium
| | - Jun-Hui Yuan
- Department of Neurology, Yale School of Medicine, New Haven, CT06520
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT06520
- Neurorehabilitation Research Center, Veterans Affairs Medical Center, West Haven, CT06516
| | - Deborah S. Jacobs
- Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA02114
| | - Philippe Gailly
- Laboratory of Cell Physiology, Institute of Neuroscience, Université catholique de Louvain, BrusselsB-1200, Belgium
| | - Sulayman D. Dib-Hajj
- Department of Neurology, Yale School of Medicine, New Haven, CT06520
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT06520
- Neurorehabilitation Research Center, Veterans Affairs Medical Center, West Haven, CT06516
| | - Stephen G. Waxman
- Department of Neurology, Yale School of Medicine, New Haven, CT06520
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT06520
- Neurorehabilitation Research Center, Veterans Affairs Medical Center, West Haven, CT06516
| |
Collapse
|
3
|
Tyagi S, Higerd-Rusli GP, Akin EJ, Baker CA, Liu S, Dib-Hajj FB, Waxman SG, Dib-Hajj SD. Real-time imaging of axonal membrane protein life cycles. Nat Protoc 2024; 19:2771-2802. [PMID: 38831222 PMCID: PMC11721981 DOI: 10.1038/s41596-024-00997-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 02/12/2024] [Indexed: 06/05/2024]
Abstract
The construction of neuronal membranes is a dynamic process involving the biogenesis, vesicular packaging, transport, insertion and recycling of membrane proteins. Optical imaging is well suited for the study of protein spatial organization and transport. However, various shortcomings of existing imaging techniques have prevented the study of specific types of proteins and cellular processes. Here we describe strategies for protein tagging and labeling, cell culture and microscopy that enable the real-time imaging of axonal membrane protein trafficking and subcellular distribution as they progress through some stages of their life cycle. First, we describe a process for engineering membrane proteins with extracellular self-labeling tags (either HaloTag or SNAPTag), which can be labeled with fluorescent ligands of various colors and cell permeability, providing flexibility for investigating the trafficking and spatiotemporal regulation of multiple membrane proteins in neuronal compartments. Next, we detail the dissection, transfection and culture of dorsal root ganglion sensory neurons in microfluidic chambers, which physically compartmentalizes cell bodies and distal axons. Finally, we describe four labeling and imaging procedures that utilize these enzymatically tagged proteins, flexible fluorescent labels and compartmentalized neuronal cultures to study axonal membrane protein anterograde and retrograde transport, the cotransport of multiple proteins, protein subcellular localization, exocytosis and endocytosis. Additionally, we generated open-source software for analyzing the imaging data in a high throughput manner. The experimental and analysis workflows provide an approach for studying the dynamics of neuronal membrane protein homeostasis, addressing longstanding challenges in this area. The protocol requires 5-7 days and expertise in cell culture and microscopy.
Collapse
Affiliation(s)
- Sidharth Tyagi
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA
- Center for Neuroscience and Regeneration Research, West Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Grant P Higerd-Rusli
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA
- Center for Neuroscience and Regeneration Research, West Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Elizabeth J Akin
- Center for Neuroscience and Regeneration Research, West Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Christopher A Baker
- Center for Neuroscience and Regeneration Research, West Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Shujun Liu
- Center for Neuroscience and Regeneration Research, West Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Fadia B Dib-Hajj
- Center for Neuroscience and Regeneration Research, West Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Stephen G Waxman
- Center for Neuroscience and Regeneration Research, West Haven, CT, USA.
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA.
| | - Sulayman D Dib-Hajj
- Center for Neuroscience and Regeneration Research, West Haven, CT, USA.
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
4
|
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
|
5
|
Tyagi S, Higerd-Rusli GP, Ghovanloo MR, Dib-Hajj F, Zhao P, Liu S, Kim DH, Shim JS, Park KS, Waxman SG, Choi JS, Dib-Hajj SD. Compartment-specific regulation of Na V1.7 in sensory neurons after acute exposure to TNF-α. Cell Rep 2024; 43:113685. [PMID: 38261513 PMCID: PMC10947185 DOI: 10.1016/j.celrep.2024.113685] [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: 08/08/2023] [Revised: 11/09/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024] Open
Abstract
Tumor necrosis factor α (TNF-α) is a major pro-inflammatory cytokine, important in many diseases, that sensitizes nociceptors through its action on a variety of ion channels, including voltage-gated sodium (NaV) channels. We show here that TNF-α acutely upregulates sensory neuron excitability and current density of threshold channel NaV1.7. Using electrophysiological recordings and live imaging, we demonstrate that this effect on NaV1.7 is mediated by p38 MAPK and identify serine 110 in the channel's N terminus as the phospho-acceptor site, which triggers NaV1.7 channel insertion into the somatic membrane. We also show that the N terminus of NaV1.7 is sufficient to mediate this effect. Although acute TNF-α treatment increases NaV1.7-carrying vesicle accumulation at axonal endings, we did not observe increased channel insertion into the axonal membrane. These results identify molecular determinants of TNF-α-mediated regulation of NaV1.7 in sensory neurons and demonstrate compartment-specific effects of TNF-α on channel insertion in the neuronal plasma membrane.
Collapse
Affiliation(s)
- Sidharth Tyagi
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT 06511, USA; Center for Neuroscience and Regeneration Research, West Haven, CT 06516, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06516, USA; Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT 06516, USA.
| | - Grant P Higerd-Rusli
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT 06511, USA; Center for Neuroscience and Regeneration Research, West Haven, CT 06516, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06516, USA; Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Mohammad-Reza Ghovanloo
- Center for Neuroscience and Regeneration Research, West Haven, CT 06516, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06516, USA; Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Fadia Dib-Hajj
- Center for Neuroscience and Regeneration Research, West Haven, CT 06516, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06516, USA; Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Peng Zhao
- Center for Neuroscience and Regeneration Research, West Haven, CT 06516, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06516, USA; Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Shujun Liu
- Center for Neuroscience and Regeneration Research, West Haven, CT 06516, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06516, USA; Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Dong-Hyun Kim
- Integrated Research Institute of Pharmaceutical Science, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, South Korea; New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, South Korea
| | - Ji Seon Shim
- Department of Physiology, Kyung Hee University School of Medicine, Seoul 02447, South Korea
| | - Kang-Sik Park
- Department of Physiology, Kyung Hee University School of Medicine, Seoul 02447, South Korea
| | - Stephen G Waxman
- Center for Neuroscience and Regeneration Research, West Haven, CT 06516, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06516, USA; Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT 06516, USA.
| | - Jin-Sung Choi
- Integrated Research Institute of Pharmaceutical Science, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, South Korea.
| | - Sulayman D Dib-Hajj
- Center for Neuroscience and Regeneration Research, West Haven, CT 06516, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06516, USA; Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT 06516, USA.
| |
Collapse
|
6
|
Loose S, Lischka A, Kuehs S, Nau C, Heinemann SH, Kurth I, Leipold E. Peripheral temperature dysregulation associated with functionally altered Na V1.8 channels. Pflugers Arch 2023; 475:1343-1355. [PMID: 37695396 PMCID: PMC10567936 DOI: 10.1007/s00424-023-02856-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/23/2023] [Accepted: 09/01/2023] [Indexed: 09/12/2023]
Abstract
The voltage-gated sodium channel NaV1.8 is prominently expressed in the soma and axons of small-caliber sensory neurons, and pathogenic variants of the corresponding gene SCN10A are associated with peripheral pain and autonomic dysfunction. While most disease-associated SCN10A variants confer gain-of-function properties to NaV1.8, resulting in hyperexcitability of sensory neurons, a few affect afferent excitability through a loss-of-function mechanism. Using whole-exome sequencing, we here identify a rare heterozygous SCN10A missense variant resulting in alteration p.V1287I in NaV1.8 in a patient with a 15-year history of progressively worsening temperature dysregulation in the distal extremities, particularly in the feet. Further symptoms include increasingly intensifying tingling and numbness in the fingers and increased sweating. To assess the impact of p.V1287I on channel function, we performed voltage-clamp recordings demonstrating that the alteration confers loss- and gain-of-function characteristics to NaV1.8 characterized by a right-shifted voltage dependence of channel activation and inactivation. Current-clamp recordings from transfected mouse dorsal root ganglion neurons further revealed that NaV1.8-V1287I channels broaden the action potentials of sensory neurons and increase their firing rates in response to depolarizing current stimulations, indicating a gain-of-function mechanism of the variant at the cellular level in a heterozygous setting. The data support the hypothesis that the properties of NaV1.8 p.V1287I are causative for the patient's symptoms and that nonpainful peripheral paresthesias should be considered part of the clinical spectrum of NaV1.8-associated disorders.
Collapse
Affiliation(s)
- Simon Loose
- Department of Anesthesiology and Intensive Care & CBBM - Center of Brain, Behavior and Metabolism, University of Luebeck, Ratzeburger Allee 160, 23562, Luebeck, Germany
| | - Annette Lischka
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Samuel Kuehs
- Department of Anesthesiology and Intensive Care & CBBM - Center of Brain, Behavior and Metabolism, University of Luebeck, Ratzeburger Allee 160, 23562, Luebeck, Germany
| | - Carla Nau
- Department of Anesthesiology and Intensive Care & CBBM - Center of Brain, Behavior and Metabolism, University of Luebeck, Ratzeburger Allee 160, 23562, Luebeck, Germany
| | - Stefan H Heinemann
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena and Jena University Hospital, Jena, Germany
| | - Ingo Kurth
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Enrico Leipold
- Department of Anesthesiology and Intensive Care & CBBM - Center of Brain, Behavior and Metabolism, University of Luebeck, Ratzeburger Allee 160, 23562, Luebeck, Germany.
| |
Collapse
|
7
|
Tyagi S, Sarveswaran N, Higerd-Rusli GP, Liu S, Dib-Hajj FB, Waxman SG, Dib-Hajj SD. Conserved but not critical: Trafficking and function of NaV1.7 are independent of highly conserved polybasic motifs. Front Mol Neurosci 2023; 16:1161028. [PMID: 37008789 PMCID: PMC10060856 DOI: 10.3389/fnmol.2023.1161028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
Non-addictive treatment of chronic pain represents a major unmet clinical need. Peripheral voltage-gated sodium (NaV) channels are an attractive target for pain therapy because they initiate and propagate action potentials in primary afferents that detect and transduce noxious stimuli. NaV1.7 sets the gain on peripheral pain-signaling neurons and is the best validated peripheral ion channel involved in human pain, and previous work has shown that it is transported in vesicles in sensory axons which also carry Rab6a, a small GTPase known to be involved in vesicular packaging and axonal transport. Understanding the mechanism of the association between Rab6a and NaV1.7 could inform therapeutic modalities to decrease trafficking of NaV1.7 to the distal axonal membrane. Polybasic motifs (PBM) have been shown to regulate Rab-protein interactions in a variety of contexts. In this study, we explored whether two PBMs in the cytoplasmic loop that joins domains I and II of human NaV1.7 were responsible for association with Rab6a and regulate axonal trafficking of the channel. Using site-directed mutagenesis we generated NaV1.7 constructs with alanine substitutions in the two PBMs. Voltage-clamp recordings showed that the constructs retain wild-type like gating properties. Optical Pulse-chase Axonal Long-distance (OPAL) imaging in live sensory axons shows that mutations of these PBMs do not affect co-trafficking of Rab6a and NaV1.7, or the accumulation of the channel at the distal axonal surface. Thus, these polybasic motifs are not required for interaction of NaV1.7 with the Rab6a GTPase, or for trafficking of the channel to the plasma membrane.
Collapse
Affiliation(s)
- Sidharth Tyagi
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, United States
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Nivedita Sarveswaran
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Grant P. Higerd-Rusli
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, United States
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Shujun Liu
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Fadia B. Dib-Hajj
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Stephen G. Waxman
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
- *Correspondence: Stephen G. Waxman,
| | - Sulayman D. Dib-Hajj
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
- Sulayman D. Dib-Hajj,
| |
Collapse
|
8
|
Ghovanloo MR, Tyagi S, Zhao P, Kiziltug E, Estacion M, Dib-Hajj SD, Waxman SG. High-throughput combined voltage-clamp/current-clamp analysis of freshly isolated neurons. CELL REPORTS METHODS 2023; 3:100385. [PMID: 36814833 PMCID: PMC9939380 DOI: 10.1016/j.crmeth.2022.100385] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/11/2022] [Accepted: 12/15/2022] [Indexed: 01/15/2023]
Abstract
The patch-clamp technique is the gold-standard methodology for analysis of excitable cells. However, throughput of manual patch-clamp is slow, and high-throughput robotic patch-clamp, while helpful for applications like drug screening, has been primarily used to study channels and receptors expressed in heterologous systems. We introduce an approach for automated high-throughput patch-clamping that enhances analysis of excitable cells at the channel and cellular levels. This involves dissociating and isolating neurons from intact tissues and patch-clamping using a robotic instrument, followed by using an open-source Python script for analysis and filtration. As a proof of concept, we apply this approach to investigate the biophysical properties of voltage-gated sodium (Nav) channels in dorsal root ganglion (DRG) neurons, which are among the most diverse and complex neuronal cells. Our approach enables voltage- and current-clamp recordings in the same cell, allowing unbiased, fast, simultaneous, and head-to-head electrophysiological recordings from a wide range of freshly isolated neurons without requiring culturing on coverslips.
Collapse
Affiliation(s)
- Mohammad-Reza Ghovanloo
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA
- Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Sidharth Tyagi
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA
- Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Medical Scientist Training Program, Yale University School of Medicine, New Haven, CT, USA
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA
- Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Emre Kiziltug
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Mark Estacion
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA
- Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Sulayman D. Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA
- Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Stephen G. Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA
- Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
9
|
Baker CA, Tyagi S, Higerd-Rusli GP, Liu S, Zhao P, Dib-Hajj FB, Waxman SG, Dib-Hajj SD. Paclitaxel effects on axonal localization and vesicular trafficking of Na V1.8. Front Mol Neurosci 2023; 16:1130123. [PMID: 36860665 PMCID: PMC9970094 DOI: 10.3389/fnmol.2023.1130123] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 01/25/2023] [Indexed: 02/16/2023] Open
Abstract
Patients treated with paclitaxel (PTX) or other antineoplastic agents can experience chemotherapy-induced peripheral neuropathy (CIPN), a debilitating side effect characterized by numbness and pain. PTX interferes with microtubule-based transport, which inhibits tumor growth via cell cycle arrest but can also affect other cellular functions including trafficking of ion channels critical to transduction of stimuli by sensory neurons of the dorsal root ganglia (DRG). We examined the effects of PTX on voltage-gated sodium channel NaV1.8, which is preferentially expressed in DRG neurons, using a microfluidic chamber culture system and chemigenetic labeling to observe anterograde channel transport to the endings of DRG axons in real time. PTX treatment increased the numbers of NaV1.8-containing vesicles traversing the axons. Vesicles in PTX-treated cells exhibited greater average velocity, along with shorter and less frequent pauses along their trajectories. These events were paralleled by greater surface accumulation of NaV1.8 channels at the distal ends of DRG axons. These results were consistent with observations that NaV1.8 is trafficked in the same vesicles containing NaV1.7 channels, which are also involved in pain syndromes in humans and are similarly affected by PTX treatment. However, unlike Nav1.7, we did not detect increased NaV1.8 current density measured at the neuronal soma, suggesting a differential effect of PTX on trafficking of NaV1.8 in soma versus axonal compartments. Therapeutic targeting of axonal vesicular traffic would affect both Nav1.7 and Nav1.8 channels and increase the possibilities of alleviating pain associated with CIPN.
Collapse
Affiliation(s)
- Christopher A. Baker
- Department of Neurology, Yale University, New Haven, CT, United States
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT, United States
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Sidharth Tyagi
- Department of Neurology, Yale University, New Haven, CT, United States
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT, United States
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
- MD/PhD Program, Yale University, New Haven, CT, United States
| | - Grant P. Higerd-Rusli
- Department of Neurology, Yale University, New Haven, CT, United States
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT, United States
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
- MD/PhD Program, Yale University, New Haven, CT, United States
| | - Shujun Liu
- Department of Neurology, Yale University, New Haven, CT, United States
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT, United States
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Peng Zhao
- Department of Neurology, Yale University, New Haven, CT, United States
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT, United States
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Fadia B. Dib-Hajj
- Department of Neurology, Yale University, New Haven, CT, United States
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT, United States
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Stephen G. Waxman
- Department of Neurology, Yale University, New Haven, CT, United States
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT, United States
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
- *Correspondence: Stephen G. Waxman, ; Sulayman D. Dib-Hajj,
| | - Sulayman D. Dib-Hajj
- Department of Neurology, Yale University, New Haven, CT, United States
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT, United States
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
- *Correspondence: Stephen G. Waxman, ; Sulayman D. Dib-Hajj,
| |
Collapse
|
10
|
Higerd-Rusli GP, Tyagi S, Liu S, Dib-Hajj FB, Waxman SG, Dib-Hajj SD. The fates of internalized Na V1.7 channels in sensory neurons: Retrograde cotransport with other ion channels, axon-specific recycling, and degradation. J Biol Chem 2023; 299:102816. [PMID: 36539035 PMCID: PMC9843449 DOI: 10.1016/j.jbc.2022.102816] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 12/05/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
Neuronal function relies on the maintenance of appropriate levels of various ion channels at the cell membrane, which is accomplished by balancing secretory, degradative, and recycling pathways. Neuronal function further depends on membrane specialization through polarized distribution of specific proteins to distinct neuronal compartments such as axons. Voltage-gated sodium channel NaV1.7, a threshold channel for firing action potentials in nociceptors, plays a major role in human pain, and its abundance in the plasma membrane is tightly regulated. We have recently characterized the anterograde axonal trafficking of NaV1.7 channels in Rab6A-positive vesicles, but the fate of internalized channels is not known. Membrane proteins that have undergone endocytosis can be directed into multiple pathways including those for degradation, recycling to the membrane, and transcytosis. Here, we demonstrate NaV1.7 endocytosis and dynein-dependent retrograde trafficking in Rab7-containing late endosomes together with other axonal membrane proteins using real-time imaging of live neurons. We show that some internalized NaV1.7 channels are delivered to lysosomes within the cell body, and that there is no evidence for NaV1.7 transcytosis. In addition, we show that NaV1.7 is recycled specifically to the axonal membrane as opposed to the soma membrane, suggesting a novel mechanism for the development of neuronal polarity. Together, these results shed light on the mechanisms by which neurons maintain excitable membranes and may inform efforts to target ion channel trafficking for the treatment of disorders of excitability.
Collapse
Affiliation(s)
- Grant P Higerd-Rusli
- MD/PhD Program, Yale University School of Medicine, New Haven, Connecticut, USA; Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Sidharth Tyagi
- MD/PhD Program, Yale University School of Medicine, New Haven, Connecticut, USA; Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Shujun Liu
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Fadia B Dib-Hajj
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Stephen G Waxman
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA.
| | - Sulayman D Dib-Hajj
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA.
| |
Collapse
|
11
|
Upregulation of DRG protein TMEM100 facilitates dry-skin-induced pruritus by enhancing TRPA1 channel function. Acta Biochim Biophys Sin (Shanghai) 2022; 55:404-416. [PMID: 36514220 PMCID: PMC10160222 DOI: 10.3724/abbs.2022180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The dry skin tortures numerous patients with severe itch. The transient receptor potential cation channel V member 1 (TRPV1) and A member 1 (TRPA1) are two essential receptors for peripheral neural coding of itch sensory, mediating histaminergic and nonhistaminergic itch separately. In the dorsal root ganglion, transmembrane protein 100 (TMEM100) is structurally related to both TRPV1 and TRPA1 receptors, but the exact role of TMEM100 in itch sensory coding is still unknown. Here, in this study, we find that TMEM100 + DRG neurons account for the majority of activated neurons in an acetone-ether-water (AEW)-induced dry skin itch model, and some TMEM100 + DRG neurons are colocalized with both TRPA1 and the chloroquine-related Mrgpr itch receptor family. Both the expression and function of TRPA1 channels, but not TRPV1 channels, are upregulated in the AEW model, and specific DRG Tmem100 gene knockdown alleviates AEW-induced itch and rescues the expression and functional changes of TRPA1. Our results strongly suggest that TMEM100 protein in DRG is the main facilitating factor for dry-skin-related chronic itch, and specific suppression of TMEM100 in DRG could be a novel effective treatment strategy for patients who suffer from dry skin-induced itch.
Collapse
|
12
|
Kuehs S, Teege L, Hellberg AK, Stanke C, Haag N, Kurth I, Blum R, Nau C, Leipold E. Isolation and transfection of myenteric neurons from mice for patch-clamp applications. Front Mol Neurosci 2022; 15:1076187. [PMID: 36618826 PMCID: PMC9810798 DOI: 10.3389/fnmol.2022.1076187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
The enteric nervous system (ENS) is a complex neuronal network organized in ganglionated plexuses that extend along the entire length of the gastrointestinal tract. Largely independent of the central nervous system, the ENS coordinates motility and peristalsis of the digestive tract, regulates secretion and absorption, and is involved in immunological processes. Electrophysiological methods such as the patch-clamp technique are particularly suitable to study the function of neurons as well as the biophysical parameters of the underlying ion channels under both physiological and pathophysiological conditions. However, application of the patch-clamp method to ENS neurons remained difficult because they are embedded in substantial tissue layers that limit access to and targeted manipulation of these cells. Here, we present a robust step-by-step protocol that involves isolation of ENS neurons from adult mice, culturing of the cells, their transfection with plasmid DNA, and subsequent electrophysiological characterization of individual neurons in current-clamp and voltage-clamp recordings. With this protocol, ENS neurons can be prepared, transfected, and electrophysiologically characterized within 72 h. Using isolated ENS neurons, we demonstrate the feasibility of the approach by functional overexpression of recombinant voltage-gated NaV1.9 mutant channels associated with hereditary sensory and autonomic neuropathy type 7 (HSAN-7), a disorder characterized by congenital analgesia and severe constipation that can require parenteral nutrition. Although our focus is on the electrophysiological evaluation of isolated ENS neurons, the presented methodology is also useful to analyze molecules other than sodium channels or to apply alternative downstream assays including calcium imaging, proteomic and nucleic acid approaches, or immunochemistry.
Collapse
Affiliation(s)
- Samuel Kuehs
- Department of Anesthesiology and Intensive Care, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Laura Teege
- Department of Anesthesiology and Intensive Care, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Ann-Katrin Hellberg
- Department of Anesthesiology and Intensive Care, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Christina Stanke
- Department of Anesthesiology and Intensive Care, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Natja Haag
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, Rhine-Westphalia Technical University of Aachen, Aachen, Germany,Institute of Physiology, Medical Faculty, Rhine-Westphalia Technical University of Aachen, Aachen, Germany
| | - Ingo Kurth
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, Rhine-Westphalia Technical University of Aachen, Aachen, Germany
| | - Robert Blum
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Carla Nau
- Department of Anesthesiology and Intensive Care, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Enrico Leipold
- Department of Anesthesiology and Intensive Care, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany,*Correspondence: Enrico Leipold,
| |
Collapse
|
13
|
Effraim PR, Estacion M, Zhao P, Sosniak D, Waxman SG, Dib-Hajj SD. Fibroblast growth factor homologous factor 2 attenuates excitability of DRG neurons. J Neurophysiol 2022; 128:1258-1266. [PMID: 36222860 PMCID: PMC9909838 DOI: 10.1152/jn.00361.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Fibroblast growth factor homologous factors (FHFs) are cytosolic members of the superfamily of the FGF proteins. Four members of this subfamily (FHF1-4) are differentially expressed in multiple tissues in an isoform-dependent manner. Mutations in FHF proteins have been associated with multiple neurological disorders. FHF proteins bind to the COOH terminus of voltage-gated sodium (Nav) channels and regulate current amplitude and gating properties of these channels. FHF2, which is expressed in dorsal root ganglia (DRG) neurons, has two main splicing isoforms: FHF2A and FHF2B, which differ in the length and sequence of their NH2 termini, have been shown to differentially regulate gating properties of Nav1.7, a channel that is a major driver of DRG neuron firing. FHF2 expression levels are downregulated after peripheral nerve axotomy, which suggests that they may regulate neuronal excitability via an action on Nav channels after injury. We have previously shown that knockdown of FHF2 leads to gain-of-function changes in Nav1.7 gating properties: enhanced repriming, increased current density, and hyperpolarized activation. From this we posited that knockdown of FHF2 might also lead to DRG hyperexcitability. Here we show that knockdown of either FHF2A alone or all isoforms of FHF2 results in increased DRG neuron excitability. In addition, we demonstrate that supplementation of FHF2A and FHF2B reduces DRG neuron excitability. Overexpression of FHF2A or FHF2B also reduced excitability of DRG neurons treated with a cocktail of inflammatory mediators, a model of inflammatory pain. Our data suggest that increased neuronal excitability after nerve injury might be triggered, in part, via a loss of FHF2-Nav1.7 interaction.NEW & NOTEWORTHY FHF2 is known to bind to and modulate the function of Nav1.7. FHF2 expression is also reduced after nerve injury. We demonstrate that knockdown of FHF2 expression increases DRG neuronal excitability. More importantly, overexpression of FHF2 reduces DRG excitability in basal conditions and in the presence of inflammatory mediators (a model of inflammatory pain). These results suggest that FHF2 could potentially be used as a tool to reduce DRG neuronal excitability and to treat pain.
Collapse
Affiliation(s)
- Philip R. Effraim
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Mark Estacion
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Daniel Sosniak
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Sulayman D. Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
14
|
Gualdani R, Gailly P, Yuan JH, Yerna X, Di Stefano G, Truini A, Cruccu G, Dib-Hajj SD, Waxman SG. A TRPM7 mutation linked to familial trigeminal neuralgia: Omega current and hyperexcitability of trigeminal ganglion neurons. Proc Natl Acad Sci U S A 2022; 119:e2119630119. [PMID: 36095216 PMCID: PMC9499596 DOI: 10.1073/pnas.2119630119] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Trigeminal neuralgia (TN) is a unique pain disorder characterized by intense paroxysmal facial pain within areas innervated by the trigeminal nerve. Although most cases of TN are sporadic, familial clusters of TN suggest that genetic factors may contribute to this disorder. Whole-exome sequencing in patients with TN reporting positive family history demonstrated a spectrum of variants of ion channels including TRP channels. Here, we used patch-clamp analysis and Ca2+ and Na+ imaging to assess a rare variant in the TRPM7 channel, p.Ala931Thr, within transmembrane domain 3, identified in a man suffering from unilateral TN. We showed that A931T produced an abnormal inward current carried by Na+ and insensitive to the pore blocker Gd3+. Hypothesizing that replacement of the hydrophobic alanine at position 931 with the more polar threonine destabilizes a hydrophobic ring, near the voltage sensor domain, we performed alanine substitutions of F971 and W972 and obtained results suggesting a role of A931-W972 hydrophobic interaction in S3-S4 hydrophobic cleft stability. Finally, we transfected trigeminal ganglion neurons with A931T channels and observed that expression of this TRPM7 variant lowers current threshold and resting membrane potential, and increases evoked firing activity in TG neurons. Our results support the notion that the TRPM7-A931T mutation located in the S3 segment at the interface with the transmembrane region S4, generates an omega current that carries Na+ influx in physiological conditions. A931T produces hyperexcitability and a sustained Na+ influx in trigeminal ganglion neurons that may underlie pain in this kindred with trigeminal neuralgia.
Collapse
Affiliation(s)
- Roberta Gualdani
- aInstitute of Neuroscience, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Philippe Gailly
- aInstitute of Neuroscience, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Jun-Hui Yuan
- bDepartment of Neurology, Yale School of Medicine, New Haven, CT 06520
- cCenter for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT 06515
| | - Xavier Yerna
- aInstitute of Neuroscience, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Giulia Di Stefano
- dDepartment of Human Neuroscience, Sapienza University, 00185 Rome, Italy
| | - Andrea Truini
- dDepartment of Human Neuroscience, Sapienza University, 00185 Rome, Italy
| | - Giorgio Cruccu
- dDepartment of Human Neuroscience, Sapienza University, 00185 Rome, Italy
| | - Sulayman D. Dib-Hajj
- bDepartment of Neurology, Yale School of Medicine, New Haven, CT 06520
- cCenter for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT 06515
| | - Stephen G. Waxman
- bDepartment of Neurology, Yale School of Medicine, New Haven, CT 06520
- cCenter for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT 06515
- 2To whom correspondence may be addressed.
| |
Collapse
|
15
|
Labau JIR, Andelic M, Faber CG, Waxman SG, Lauria G, Dib-Hajj SD. Recent advances for using human induced-pluripotent stem cells as pain-in-a-dish models of neuropathic pain. Exp Neurol 2022; 358:114223. [PMID: 36100046 DOI: 10.1016/j.expneurol.2022.114223] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/15/2022] [Accepted: 09/05/2022] [Indexed: 11/28/2022]
Abstract
Neuropathic pain is amongst the most common non-communicable disorders and the poor effectiveness of current treatment is an unmet need. Although pain is a universal experience, there are significant inter-individual phenotypic differences. Developing models that can accurately recapitulate the clinical pain features is crucial to better understand underlying pathophysiological mechanisms and find innovative treatments. Current data from heterologous expression systems that investigate properties of specific molecules involved in pain signaling, and from animal models, show limited success with their translation into the development of novel treatments for pain. This is in part because they do not recapitulate the native environment in which a particular molecule functions, and due to species-specific differences in the properties of several key molecules that are involved in pain signaling. The limited availability of post-mortem tissue, in particular dorsal root ganglia (DRG), has hampered research using human cells in pre-clinical studies. Human induced-pluripotent stem cells (iPSCs) have emerged as an exciting alternative platform to study patient-specific diseases. Sensory neurons that are derived from iPSCs (iPSC-SNs) have provided new avenues towards elucidating peripheral pathophysiological mechanisms, the potential for development of personalized treatments, and as a cell-based system for high-throughput screening for discovering novel analgesics. Nevertheless, reprogramming and differentiation protocols to obtain nociceptors have mostly yielded immature homogenous cell populations that do not recapitulate the heterogeneity of native sensory neurons. To close the gap between native human tissue and iPSCs, alternative strategies have been developed. We will review here recent developments in differentiating iPSC-SNs and their use in pre-clinical translational studies. Direct conversion of stem cells into the cells of interest has provided a more cost- and time-saving method to improve reproducibility and diversity of sensory cell types. Furthermore, multi-cellular strategies that mimic in vivo microenvironments for cell maturation, by improving cell contact and communication (co-cultures), reproducing the organ complexity and architecture (three-dimensional organoid), and providing iPSCs with the full spatiotemporal context and nutrients needed for acquiring a mature phenotype (xenotransplantation), have led to functional sensory neuron-like systems. Finally, this review touches on novel prospective strategies, including fluorescent-tracking to select the differentiated neurons of relevance, and dynamic clamp, an electrophysiological method that allows direct manipulation of ionic conductances that are missing in iPSC-SNs.
Collapse
Affiliation(s)
- Julie I R Labau
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA; Department of Toxicogenomics, Clinical Genomics, Maastricht University Medical Centre+, Maastricht, the Netherlands; School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Mirna Andelic
- School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands; Department of Neurology, Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Catharina G Faber
- School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA
| | - Giuseppe Lauria
- Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Italy.
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.
| |
Collapse
|
16
|
Transfection of Neuronal Cultures. Methods Mol Biol 2021. [PMID: 34033082 DOI: 10.1007/978-1-0716-1437-2_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Efficient transfection of genes into the neurons is a crucial step for the study of neuronal cell biology and functions. These include but not limited to investigating gene function by overexpression of target proteins via expression plasmids and knocking down the expression levels of neuronal genes by RNA interference (RNAi). In addition, reporter gene constructs are widely used to investigate the promoter activities of neuronal genes. Numerous transfection techniques have been established to deliver genes into the cells. However, efficient transfection of postmitotic cells, including neurons, still remains a challenging task. Here, we overview the advantages and disadvantages of various techniques for the transfection of primary neurons, and provide an optimized protocol for FuGENE-6 (Promega) which allows for a suitable transfection efficiency of primary neuronal cultures.
Collapse
|
17
|
Dusan M, Jastrow C, Alyce MM, Yingkai W, Shashikanth M, Andelain E, Christine BM, Stuart BM, Oliver BG, Michael MZ, Nicolas VH, Damien KJ, Rainer HV. Differentiation of the 50B11 dorsal ganglion cells into NGF and GDNF responsive nociceptor subtypes. Mol Pain 2021; 16:1744806920970368. [PMID: 33307981 PMCID: PMC7745567 DOI: 10.1177/1744806920970368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The embryonic rat dorsal root ganglion (DRG) neuron-derived 50B11 cell line is a promising sensory neuron model expressing markers characteristic of NGF and GDNF-dependent C-fibre nociceptors. Whether these cells have the capacity to develop into distinct nociceptive subtypes based on NGF- or GDNF-dependence has not been investigated. Here we show that by augmenting forskolin (FSK) and growth factor supplementation with NGF or GDNF, 50B11 cultures can be driven to acquire differential functional responses to common nociceptive agonists capsaicin and ATP respectively. In addition, to previous studies, we also demonstrate that a differentiated neuronal phenotype can be maintained for up to 7 days. Western blot analysis of nociceptive marker proteins further demonstrates that the 50B11 cells partially recapitulate the functional phenotypes of classical NGF-dependent (peptidergic) and GDNF-dependent (non-peptidergic) neuronal subtypes described in DRGs. Further, 50B11 cells differentiated with NGF/FSK, but not GDNF/FSK, show sensitization to acute prostaglandin E2 treatment. Finally, RNA-Seq analysis confirms that differentiation with NGF/FSK or GDNF/FSK produces two 50B11 cell subtypes with distinct transcriptome expression profiles. Gene ontology comparison of the two subtypes of differentiated 50B11 cells to rodent DRG neurons studies shows significant overlap in matching or partially matching categories. This transcriptomic analysis will aid future suitability assessment of the 50B11 cells as a high-throughput nociceptor model for a broad range of experimental applications. In conclusion, this study shows that the 50B11 cell line is capable of partially recapitulating features of two distinct types of embryonic NGF and GDNF-dependent nociceptor-like cells.
Collapse
Affiliation(s)
- Matusica Dusan
- Anatomy and Histology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Canlas Jastrow
- Anatomy and Histology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Martin M Alyce
- Human Physiology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Wei Yingkai
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University and Medical Centre, Bedford Park, Adelaide, South Australia, Australia
| | - Marri Shashikanth
- Visceral Pain Research Group, College of Medicine and Public Health, South Australian Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Erickson Andelain
- Flow Cytometry Facility, Department of Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Barry M Christine
- Anatomy and Histology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Brierley M Stuart
- Flow Cytometry Facility, Department of Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Best G Oliver
- Flow Cytometry Facility, Department of Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Michael Z Michael
- Visceral Pain Research Group, College of Medicine and Public Health, South Australian Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Voelcker H Nicolas
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University and Medical Centre, Bedford Park, Adelaide, South Australia, Australia
| | - Keating J Damien
- Human Physiology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Haberberger V Rainer
- Anatomy and Histology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| |
Collapse
|
18
|
Alsaloum M, Labau JIR, Sosniak D, Zhao P, Almomani R, Gerrits M, Hoeijmakers JGJ, Lauria G, Faber CG, Waxman SG, Dib-Hajj S. A novel gain-of-function sodium channel β2 subunit mutation in idiopathic small fiber neuropathy. J Neurophysiol 2021; 126:827-839. [PMID: 34320850 DOI: 10.1152/jn.00184.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Small fiber neuropathy (SFN) is a common condition affecting thinly myelinated Aδ and unmyelinated C fibers, often resulting in excruciating pain and dysautonomia. SFN has been associated with several conditions, but a significant number of cases have no discernible cause. Recent genetic studies have identified potentially pathogenic gain-of-function mutations in several the pore-forming voltage-gated sodium channel α subunits (NaVs) in a subset of patients with SFN, but the auxiliary sodium channel β subunits have been less implicated in the development of the disease. β subunits modulate NaV trafficking and gating, and several mutations have been linked to epilepsy and cardiac dysfunction. Recently, we provided the first evidence for the contribution of a mutation in the β2-subunit to pain in human painful diabetic neuropathy. Here, we provide the first evidence for the involvement of a sodium channel β subunit mutation in the pathogenesis of SFN with no other known causes. We show, through current-clamp analysis, that the newly-identified Y69H variant of the β2 subunit induces neuronal hyperexcitability in dorsal root ganglion neurons, lowering the threshold for action potential firing and allowing for increased repetitive action potential spiking. Underlying the hyperexcitability induced by the β2-Y69H variant, we demonstrate an upregulation in tetrodotoxin-sensitive, but not tetrodotoxin-resistant sodium currents. This provides the first evidence for the involvement of β2 subunits in SFN and strengthens the link between sodium channel β subunits and the development of neuropathic pain in humans.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, United States.,Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT, United States
| | - Julie I R Labau
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States.,Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, the Netherlands.,Department of Neurology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Daniel Sosniak
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Rowida Almomani
- Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, the Netherlands.,Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Monique Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | | | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, Milan, Italy.,Department of Biomedical and Clinical Sciences "Luigi Sacco," University of Milan, Milan, Italy
| | - Catherina G Faber
- Department of Neurology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Sulayman Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
19
|
Chen L, Wimalasena NK, Shim J, Han C, Lee SI, Gonzalez-Cano R, Estacion M, Faber CG, Lauria G, Dib-Hajj SD, Woolf CJ, Waxman SG. Two independent mouse lines carrying the Nav1.7 I228M gain-of-function variant display dorsal root ganglion neuron hyperexcitability but a minimal pain phenotype. Pain 2021; 162:1758-1770. [PMID: 33323889 PMCID: PMC8119301 DOI: 10.1097/j.pain.0000000000002171] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/23/2020] [Indexed: 01/26/2023]
Abstract
ABSTRACT Small-fiber neuropathy (SFN), characterized by distal unmyelinated or thinly myelinated fiber loss, produces a combination of sensory dysfunction and neuropathic pain. Gain-of-function variants in the sodium channel Nav1.7 that produce dorsal root ganglion (DRG) neuron hyperexcitability are present in 5% to 10% of patients with idiopathic painful SFN. We created 2 independent knock-in mouse lines carrying the Nav1.7 I228M gain-of-function variant, found in idiopathic SFN. Whole-cell patch-clamp and multielectrode array recordings show that Nav1.7 I228M knock-in DRG neurons are hyperexcitable compared with wild-type littermate-control neurons, but despite this, Nav1.7 I228M mice do not display mechanical or thermal hyperalgesia or intraepidermal nerve fiber loss in vivo. Therefore, although these 2 Nav1.7 I228M knock-in mouse lines recapitulate the DRG neuron hyperexcitability associated with gain-of-function mutations in Nav1.7, they do not recapitulate the pain or neuropathy phenotypes seen in patients. We suggest that the relationship between hyperexcitability in sensory neurons and the pain experienced by these patients may be more complex than previously appreciated and highlights the challenges in modelling channelopathy pain disorders in mice.
Collapse
Affiliation(s)
- Lubin Chen
- Department of Neurology, Regeneration Research, Yale
University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale
University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut
Healthcare System, West Haven, CT 06516, USA
| | - Nivanthika K. Wimalasena
- FM Kirby Neurobiology Center, Boston Children’s
Hospital, Harvard Medical school, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical school, Boston,
MA 02115, USA
| | - Jaehoon Shim
- FM Kirby Neurobiology Center, Boston Children’s
Hospital, Harvard Medical school, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical school, Boston,
MA 02115, USA
| | - Chongyang Han
- Department of Neurology, Regeneration Research, Yale
University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale
University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut
Healthcare System, West Haven, CT 06516, USA
| | - Seong-il Lee
- Department of Neurology, Regeneration Research, Yale
University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale
University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut
Healthcare System, West Haven, CT 06516, USA
| | - Rafael Gonzalez-Cano
- FM Kirby Neurobiology Center, Boston Children’s
Hospital, Harvard Medical school, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical school, Boston,
MA 02115, USA
| | - Mark Estacion
- Department of Neurology, Regeneration Research, Yale
University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale
University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut
Healthcare System, West Haven, CT 06516, USA
| | - Catharina G. Faber
- Department of Neurology, School of Mental Health and
Neuroscience, Maastricht University Medical Center, Maastricht, The
Netherlands
| | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS Foundation, “Carlo
Besta” Neurological Institute, Milan, Italy
- Department of Biomedical and Clinical Sciences
“Luigi Sacco”, University of Milan, Italy
| | - Sulayman D. Dib-Hajj
- Department of Neurology, Regeneration Research, Yale
University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale
University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut
Healthcare System, West Haven, CT 06516, USA
| | - Clifford J. Woolf
- FM Kirby Neurobiology Center, Boston Children’s
Hospital, Harvard Medical school, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical school, Boston,
MA 02115, USA
| | - Stephen G. Waxman
- Department of Neurology, Regeneration Research, Yale
University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale
University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut
Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
20
|
Yin W, Liu L, Zhou Y, Zhang Y, Kong D, Xu S, Tang D, Huang D, Wen D, Jiao Y, Fan Y, Gao P, Yu W. Complete Freund's adjuvant-induced decrement of pruriceptor-mediated suppression of itch. Acta Biochim Biophys Sin (Shanghai) 2021; 53:538-546. [PMID: 33693534 DOI: 10.1093/abbs/gmab027] [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: 09/28/2020] [Indexed: 11/13/2022] Open
Abstract
Peripheral inflammation is always accompanied by a noxious sensation, either pain or itch, providing a protective warning for the occurrence of pathological changes; however, the mechanisms determining whether pain, itch, or both will be elicited under certain inflammatory statuses are still far from clear. Complete Freund's adjuvant (CFA) contains heat killed and dried Mycobacterium tuberculosis widely used to induce inflammatory pain models, but how CFA treatment affects itch sensation and the possible mechanisms are still unclear. In this study, using itch behavior testing and calcium imaging, we showed that both the behaviors and calcium responses associated with Transient Receptor Potential Vanilloid 1 (TRPV1)-mediated histamine-dependent itch and Transient Receptor Potential Ankyrin 1 (TRPA1)-mediated histamine-independent itch were significantly suppressed by CFA treatment. Furthermore, to explore the possible cellular mechanisms, high-throughput single-cell RNA sequencing and real-time PCR were used to detect CFA-induced changes of itch-related genes in dorsal root ganglion (DRG) neurons. Our results revealed that although both nociceptive Trpv1+ and Trpa1+ DRG neurons were increased after CFA treatment, most known pruriceptors, including Hrh1+, Mrgpra3+, Mrgprd+, Htr3a+, Htr1f+, IL31ra+, Osmr+, and Lpar3+ DRG neurons, were significantly decreased, which may explain that CFA treatment caused itch suppression. This study indicated that itch sensation was affected after CFA treatment, although negatively, and comprehensive but not specific suppression of different pruriceptors was observed after CFA treatment, suggesting that a unified adaptive change of increased pain and decreased itch will occur simultaneously under CFA-induced inflammatory conditions.
Collapse
Affiliation(s)
- Wen Yin
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Li Liu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yuxi Zhou
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yunchun Zhang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Dexu Kong
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Saihong Xu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Dan Tang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Dan Huang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Daxiang Wen
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yingfu Jiao
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yinghui Fan
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Po Gao
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| |
Collapse
|
21
|
Akin EJ, Alsaloum M, Higerd GP, Liu S, Zhao P, Dib-Hajj FB, Waxman SG, Dib-Hajj SD. Paclitaxel increases axonal localization and vesicular trafficking of Nav1.7. Brain 2021; 144:1727-1737. [PMID: 33734317 PMCID: PMC8320304 DOI: 10.1093/brain/awab113] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/09/2021] [Accepted: 03/04/2021] [Indexed: 01/15/2023] Open
Abstract
The microtubule-stabilizing chemotherapy drug paclitaxel (PTX) causes dose-limiting chemotherapy-induced peripheral neuropathy (CIPN), which is often accompanied by pain. Among the multifaceted effects of PTX is an increased expression of sodium channel Nav1.7 in rat and human sensory neurons, enhancing their excitability. However, the mechanisms underlying this increased Nav1.7 expression have not been explored, and the effects of PTX treatment on the dynamics of trafficking and localization of Nav1.7 channels in sensory axons have not been possible to investigate to date. In this study we used a recently developed live imaging approach that allows visualization of Nav1.7 surface channels and long-distance axonal vesicular transport in sensory neurons to fill this basic knowledge gap. We demonstrate concentration and time-dependent effects of PTX on vesicular trafficking and membrane localization of Nav1.7 in real-time in sensory axons. Low concentrations of PTX increase surface channel expression and vesicular flux (number of vesicles per axon). By contrast, treatment with a higher concentration of PTX decreases vesicular flux. Interestingly, vesicular velocity is increased for both concentrations of PTX. Treatment with PTX increased levels of endogenous Nav1.7 mRNA and current density in dorsal root ganglion neurons. However, the current produced by transfection of dorsal root ganglion neurons with Halo-tag Nav1.7 was not increased after exposure to PTX. Taken together, this suggests that the increased trafficking and surface localization of Halo-Nav1.7 that we observed by live imaging in transfected dorsal root ganglion neurons after treatment with PTX might be independent of an increased pool of Nav1.7 channels. After exposure to inflammatory mediators to mimic the inflammatory condition seen during chemotherapy, both Nav1.7 surface levels and vesicular transport are increased for both low and high concentrations of PTX. Overall, our results show that PTX treatment increases levels of functional endogenous Nav1.7 channels in dorsal root ganglion neurons and enhances trafficking and surface distribution of Nav1.7 in sensory axons, with outcomes that depend on the presence of an inflammatory milieu, providing a mechanistic explanation for increased excitability of primary afferents and pain in CIPN.
Collapse
Affiliation(s)
- Elizabeth J Akin
- Department of Neurology, Yale University, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Matthew Alsaloum
- Department of Neurology, Yale University, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA.,MD/PhD Program, Yale University, New Haven, CT 06510, USA
| | - Grant P Higerd
- Department of Neurology, Yale University, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA.,MD/PhD Program, Yale University, New Haven, CT 06510, USA
| | - Shujun Liu
- Department of Neurology, Yale University, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Peng Zhao
- Department of Neurology, Yale University, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Fadia B Dib-Hajj
- Department of Neurology, Yale University, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
22
|
A Neanderthal Sodium Channel Increases Pain Sensitivity in Present-Day Humans. Curr Biol 2020; 30:3465-3469.e4. [DOI: 10.1016/j.cub.2020.06.045] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 03/20/2020] [Accepted: 06/12/2020] [Indexed: 01/10/2023]
|
23
|
Grubinska B, Chen L, Alsaloum M, Rampal N, Matson DJ, Yang C, Taborn K, Zhang M, Youngblood B, Liu D, Galbreath E, Allred S, Lepherd M, Ferrando R, Kornecook TJ, Lehto SG, Waxman SG, Moyer BD, Dib-Hajj S, Gingras J. Rat Na V1.7 loss-of-function genetic model: Deficient nociceptive and neuropathic pain behavior with retained olfactory function and intra-epidermal nerve fibers. Mol Pain 2020; 15:1744806919881846. [PMID: 31550995 PMCID: PMC6831982 DOI: 10.1177/1744806919881846] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recapitulating human disease pathophysiology using genetic animal models is a
powerful approach to enable mechanistic understanding of genotype–phenotype
relationships for drug development. NaV1.7 is a sodium channel
expressed in the peripheral nervous system with strong human genetic validation
as a pain target. Efforts to identify novel analgesics that are nonaddictive
resulted in industry exploration of a class of sulfonamide compounds that bind
to the fourth voltage-sensor domain of NaV1.7. Due to sequence
differences in this region, sulfonamide blockers generally are potent on human
but not rat NaV1.7 channels. To test sulfonamide-based chemical
matter in rat models of pain, we generated a humanized NaV1.7 rat
expressing a chimeric NaV1.7 protein containing the
sulfonamide-binding site of the human gene sequence as a replacement for the
equivalent rat sequence. Unexpectedly, upon transcription, the human insert was
spliced out, resulting in a premature stop codon. Using a validated antibody,
NaV1.7 protein was confirmed to be lost in the brainstem, dorsal
root ganglia, sciatic nerve, and gastrointestinal tissue but not in nasal
turbinates or olfactory bulb in rats homozygous for the knock-in allele
(HOM-KI). HOM-KI rats exhibited normal intraepidermal nerve fiber density with
reduced tetrodotoxin-sensitive current density and action potential firing in
small diameter dorsal root ganglia neurons. HOM-KI rats did not exhibit
nociceptive pain responses in hot plate or capsaicin-induced flinching assays
and did not exhibit neuropathic pain responses following spinal nerve ligation.
Consistent with expression of chimeric NaV1.7 in olfactory tissue,
HOM-KI rats retained olfactory function. This new genetic model highlights the
necessity of NaV1.7 for pain behavior in rats and indicates that
sufficient inhibition of NaV1.7 in humans may reduce pain in
neuropathic conditions. Due to preserved olfactory function, this rat model
represents an alternative to global NaV1.7 knockout mice that require
time-intensive hand feeding during early postnatal development.
Collapse
Affiliation(s)
- B Grubinska
- Neuroscience Department, Amgen Research, Cambridge, MA, USA.,Voyager Therapeutics, Cambridge, MA, USA
| | - L Chen
- Department of Neurology, Yale University, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA
| | - M Alsaloum
- Department of Neurology, Yale University, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.,Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA
| | - N Rampal
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - D J Matson
- Neuroscience Department, Amgen Research, Cambridge, MA, USA
| | - C Yang
- Neuroscience Department, Amgen Research, Cambridge, MA, USA
| | - K Taborn
- Neuroscience Department, Amgen Research, Cambridge, MA, USA.,Wave Life Sciences, Ltd, Cambridge, MA, USA
| | - M Zhang
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - B Youngblood
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - D Liu
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - E Galbreath
- Comparative Biology and Safety Sciences, Amgen Research, Cambridge, MA, USA.,Takeda Pharmaceutical Company Ltd, Cambridge, MA, USA
| | - S Allred
- Comparative Biology and Safety Sciences, Amgen Research, South San Francisco, CA, USA.,Seattle Genetics, Bothell, WA, USA
| | - M Lepherd
- Comparative Biology and Safety Sciences, Amgen Research, South San Francisco, CA, USA.,Genentech, Inc. South San Francisco, CA, USA
| | - R Ferrando
- Comparative Biology and Safety Sciences, Amgen Research, South San Francisco, CA, USA.,AbbVie Stemcentrx, Inc., South San Francisco, CA, USA
| | - T J Kornecook
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA.,Biogen Inc., Cambridge, MA, USA
| | - S G Lehto
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - S G Waxman
- Department of Neurology, Yale University, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA
| | - B D Moyer
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - S Dib-Hajj
- Department of Neurology, Yale University, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA
| | - J Gingras
- Neuroscience Department, Amgen Research, Cambridge, MA, USA.,Homology Medicine Inc., Bedford, MA, USA
| |
Collapse
|
24
|
Alsaloum M, Estacion M, Almomani R, Gerrits MM, Bönhof GJ, Ziegler D, Malik R, Ferdousi M, Lauria G, Merkies IS, Faber CG, Dib-Hajj S, Waxman SG. A gain-of-function sodium channel β2-subunit mutation in painful diabetic neuropathy. Mol Pain 2020; 15:1744806919849802. [PMID: 31041876 PMCID: PMC6510061 DOI: 10.1177/1744806919849802] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Diabetes mellitus is a global challenge with many diverse health sequelae, of which diabetic peripheral neuropathy is one of the most common. A substantial number of patients with diabetic peripheral neuropathy develop chronic pain, but the genetic and epigenetic factors that predispose diabetic peripheral neuropathy patients to develop neuropathic pain are poorly understood. Recent targeted genetic studies have identified mutations in α-subunits of voltage-gated sodium channels (Navs) in patients with painful diabetic peripheral neuropathy. Mutations in proteins that regulate trafficking or functional properties of Navs could expand the spectrum of patients with Nav-related peripheral neuropathies. The auxiliary sodium channel β-subunits (β1–4) have been reported to increase current density, alter inactivation kinetics, and modulate subcellular localization of Nav. Mutations in β-subunits have been associated with several diseases, including epilepsy, cancer, and diseases of the cardiac conducting system. However, mutations in β-subunits have never been shown previously to contribute to neuropathic pain. We report here a patient with painful diabetic peripheral neuropathy and negative genetic screening for mutations in SCN9A, SCN10A, and SCN11A—genes encoding sodium channel α-subunit that have been previously linked to the development of neuropathic pain. Genetic analysis revealed an aspartic acid to asparagine mutation, D109N, in the β2-subunit. Functional analysis using current-clamp revealed that the β2-D109N rendered dorsal root ganglion neurons hyperexcitable, especially in response to repetitive stimulation. Underlying the hyperexcitability induced by the β2-subunit mutation, as evidenced by voltage-clamp analysis, we found a depolarizing shift in the voltage dependence of Nav1.7 fast inactivation and reduced use-dependent inhibition of the Nav1.7 channel.
Collapse
Affiliation(s)
- Matthew Alsaloum
- 1 Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,2 Center for Neuroscience and Regeneration Research, Veterans Affairs Medical Center, West Haven, CT, USA.,3 Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
| | - Mark Estacion
- 1 Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,2 Center for Neuroscience and Regeneration Research, Veterans Affairs Medical Center, West Haven, CT, USA
| | - Rowida Almomani
- 4 Department of Clinical Genomics, University Medical Center Maastricht, Maastricht, the Netherlands.,5 Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Monique M Gerrits
- 4 Department of Clinical Genomics, University Medical Center Maastricht, Maastricht, the Netherlands.,6 Department of Neurology, University Medical Centre Maastricht, Maastricht, the Netherlands
| | - Gidon J Bönhof
- 7 Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Dan Ziegler
- 7 Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,1 8German Center for Diabetes Research, München-Neuherberg, Germany.,9 Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Rayaz Malik
- 10 Weill Cornell Medicine-Qatar, Doha, Qatar.,11 Division of Diabetes, Endocrinology and Gastroenterology, Institute of Human Development, University of Manchester, Manchester, UK
| | - Maryam Ferdousi
- 11 Division of Diabetes, Endocrinology and Gastroenterology, Institute of Human Development, University of Manchester, Manchester, UK
| | - Giuseppe Lauria
- 12 Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, Milan, Italy.,13 Department of Biomedical and Clinical Sciences "Luigi Sacco," University of Milan, Milan, Italy
| | - Ingemar Sj Merkies
- 6 Department of Neurology, University Medical Centre Maastricht, Maastricht, the Netherlands.,14 Department of Neurology, St. Elisabeth Hospital, Willemstad, Curaçao
| | - Catharina G Faber
- 6 Department of Neurology, University Medical Centre Maastricht, Maastricht, the Netherlands
| | - Sulayman Dib-Hajj
- 1 Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,2 Center for Neuroscience and Regeneration Research, Veterans Affairs Medical Center, West Haven, CT, USA
| | - Stephen G Waxman
- 1 Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,2 Center for Neuroscience and Regeneration Research, Veterans Affairs Medical Center, West Haven, CT, USA
| | | |
Collapse
|
25
|
Pharmacological characterization of a rat Nav1.7 loss-of-function model with insensitivity to pain. Pain 2020; 161:1350-1360. [DOI: 10.1097/j.pain.0000000000001807] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
26
|
Sizova DV, Huang J, Akin EJ, Estacion M, Gomis-Perez C, Waxman SG, Dib-Hajj SD. A 49-residue sequence motif in the C terminus of Nav1.9 regulates trafficking of the channel to the plasma membrane. J Biol Chem 2020. [DOI: 10.1016/s0021-9258(17)49917-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
27
|
Sizova DV, Huang J, Akin EJ, Estacion M, Gomis-Perez C, Waxman SG, Dib-Hajj SD. A 49-residue sequence motif in the C terminus of Nav1.9 regulates trafficking of the channel to the plasma membrane. J Biol Chem 2019; 295:1077-1090. [PMID: 31822564 DOI: 10.1074/jbc.ra119.011424] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/06/2019] [Indexed: 12/18/2022] Open
Abstract
Genetic and functional studies have confirmed an important role for the voltage-gated sodium channel Nav1.9 in human pain disorders. However, low functional expression of Nav1.9 in heterologous systems (e.g. in human embryonic kidney 293 (HEK293) cells) has hampered studies of its biophysical and pharmacological properties and the development of high-throughput assays for drug development targeting this channel. The mechanistic basis for the low level of Nav1.9 currents in heterologous expression systems is not understood. Here, we implemented a multidisciplinary approach to investigate the mechanisms that govern functional Nav1.9 expression. Recombinant expression of a series of Nav1.9-Nav1.7 C-terminal chimeras in HEK293 cells identified a 49-amino-acid-long motif in the C terminus of the two channels that regulates expression levels of these chimeras. We confirmed the critical role of this motif in the context of a full-length channel chimera, Nav1.9-Ct49aaNav1.7, which displayed significantly increased current density in HEK293 cells while largely retaining the characteristic Nav1.9-gating properties. High-resolution live microscopy indicated that the newly identified C-terminal motif dramatically increases the number of channels on the plasma membrane of HEK293 cells. Molecular modeling results suggested that this motif is exposed on the cytoplasmic face of the folded C terminus, where it might interact with other channel partners. These findings reveal that a 49-residue-long motif in Nav1.9 regulates channel trafficking to the plasma membrane.
Collapse
Affiliation(s)
- Daria V Sizova
- Department of Neurology, Yale University, New Haven, Connecticut 06510.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, Connecticut 06510.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516
| | - Jianying Huang
- Department of Neurology, Yale University, New Haven, Connecticut 06510.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, Connecticut 06510.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516
| | - Elizabeth J Akin
- Department of Neurology, Yale University, New Haven, Connecticut 06510.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, Connecticut 06510.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516
| | - Mark Estacion
- Department of Neurology, Yale University, New Haven, Connecticut 06510.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, Connecticut 06510.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516
| | - Carolina Gomis-Perez
- Department of Neurology, Yale University, New Haven, Connecticut 06510.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, Connecticut 06510.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516
| | - Stephen G Waxman
- Department of Neurology, Yale University, New Haven, Connecticut 06510 .,Center for Neuroscience and Regeneration Research, Yale University, New Haven, Connecticut 06510.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University, New Haven, Connecticut 06510 .,Center for Neuroscience and Regeneration Research, Yale University, New Haven, Connecticut 06510.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516
| |
Collapse
|
28
|
Akin EJ, Higerd GP, Mis MA, Tanaka BS, Adi T, Liu S, Dib-Hajj FB, Waxman SG, Dib-Hajj SD. Building sensory axons: Delivery and distribution of Na V1.7 channels and effects of inflammatory mediators. SCIENCE ADVANCES 2019; 5:eaax4755. [PMID: 31681845 PMCID: PMC6810356 DOI: 10.1126/sciadv.aax4755] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 09/13/2019] [Indexed: 05/12/2023]
Abstract
Sodium channel NaV1.7 controls firing of nociceptors, and its role in human pain has been validated by genetic and functional studies. However, little is known about NaV1.7 trafficking or membrane distribution along sensory axons, which can be a meter or more in length. We show here with single-molecule resolution the first live visualization of NaV1.7 channels in dorsal root ganglia neurons, including long-distance microtubule-dependent vesicular transport in Rab6A-containing vesicles. We demonstrate nanoclusters that contain a median of 12.5 channels at the plasma membrane on axon termini. We also demonstrate that inflammatory mediators trigger an increase in the number of NaV1.7-carrying vesicles per axon, a threefold increase in the median number of NaV1.7 channels per vesicle and a ~50% increase in forward velocity. This remarkable enhancement of NaV1.7 vesicular trafficking and surface delivery under conditions that mimic a disease state provides new insights into the contribution of NaV1.7 to inflammatory pain.
Collapse
Affiliation(s)
- Elizabeth J. Akin
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Grant P. Higerd
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
- MD-PhD Program, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Malgorzata A. Mis
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Brian S. Tanaka
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Talia Adi
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Shujun Liu
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Fadia B. Dib-Hajj
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G. Waxman
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
- Corresponding author. (S.D.D.-H.); (S.G.W.)
| | - Sulayman D. Dib-Hajj
- Department of Neurology, Yale University, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
- Corresponding author. (S.D.D.-H.); (S.G.W.)
| |
Collapse
|
29
|
Huang J, Estacion M, Zhao P, Dib-Hajj FB, Schulman B, Abicht A, Kurth I, Brockmann K, Waxman SG, Dib-Hajj SD. A Novel Gain-of-Function Nav1.9 Mutation in a Child With Episodic Pain. Front Neurosci 2019; 13:918. [PMID: 31551682 PMCID: PMC6733892 DOI: 10.3389/fnins.2019.00918] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/16/2019] [Indexed: 12/22/2022] Open
Abstract
Voltage-gated sodium channel Nav1.9 is a threshold channel that regulates action potential firing. Nav1.9 is preferentially expressed in myenteric neurons, and small-diameter dorsal root ganglion (DRG) and trigeminal ganglion neurons including nociceptors. Recent studies have demonstrated a monogenic Mendelian link of Nav1.9 to human pain disorders. Gain-of-function variants in Nav1.9, which cause smaller depolarizations of RMP, have been identified in patients with familial episodic pain type 3 (FEPS3) and the more common pain disorder small fiber neuropathy. To explore the phenotypic spectrum of Nav1.9 channelopathy, here we report a new Nav1.9 mutation, N816K, in a child with early-onset episodic pain in both legs, episodic abdominal pain, and chronic constipation. Sequencing of further selected pain genes was normal. N816K alters a residue at the N-terminus of loop 2, proximal to the cytoplasmic terminus of transmembrane segment 6 in domain II. Voltage-clamp recordings demonstrate that Nav1.9-N816K significantly increases current density and hyperpolarizes voltage-dependence of activation by 10 mV, enabling a larger window current. Current-clamp recordings in DRG neurons shows that N816K channels depolarize RMP of small DRG neurons by 7 mV, reduce current threshold of firing an action potential and render DRG neurons hyperexcitable. Taken together these data demonstrate gain-of-function attributes of the newly described N816K mutation at the channel and cellular levels, which are consistent with a pain phenotype in the carrier of this mutation.
Collapse
Affiliation(s)
- Jianying Huang
- Department of Neurology, Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, United States.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Mark Estacion
- Department of Neurology, Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, United States.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Peng Zhao
- Department of Neurology, Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, United States.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Fadia B Dib-Hajj
- Department of Neurology, Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, United States.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Betsy Schulman
- Department of Neurology, Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, United States.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Angela Abicht
- Medizinisch Genetisches Zentrum, Munich, Germany.,Department of Neurology, Friedrich-Baur-Institute, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Ingo Kurth
- Medical Faculty, Institute of Human Genetics, RWTH Aachen University, Aachen, Germany
| | - Knut Brockmann
- Department of Pediatrics and Pediatric Neurology, Georg August University, Göttingen, Germany
| | - Stephen G Waxman
- Department of Neurology, Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, United States.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Sulayman D Dib-Hajj
- Department of Neurology, Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, United States.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
30
|
Effraim PR, Huang J, Lampert A, Stamboulian S, Zhao P, Black JA, Dib-Hajj SD, Waxman SG. Fibroblast growth factor homologous factor 2 (FGF-13) associates with Nav1.7 in DRG neurons and alters its current properties in an isoform-dependent manner. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2019; 6:100029. [PMID: 31223136 PMCID: PMC6565799 DOI: 10.1016/j.ynpai.2019.100029] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 03/22/2019] [Accepted: 03/22/2019] [Indexed: 11/29/2022]
Abstract
Fibroblast Growth Factor Homologous Factors (FHF) constitute a subfamily of FGF proteins with four prototypes (FHF1-4; also known as FGF11-14). FHF proteins have been shown to bind directly to the membrane-proximal segment of the C-terminus in voltage-gated sodium channels (Nav), and regulate current density, availability, and frequency-dependent inhibition of sodium currents. Members of the FHF2 subfamily, FHF2A and FHF2B, differ in the length and sequence of their N-termini, and, importantly, differentially regulate Nav1.6 gating properties. Using immunohistochemistry, we show that FHF2 isoforms are expressed in adult dorsal root ganglion (DRG) neurons where they co-localize with Nav1.6 and Nav1.7. FHF2A and FHF2B show differential localization in neuronal compartments in DRG neurons, and levels of expression of FHF2 factors are down-regulated following sciatic nerve axotomy. Because Nav1.7 in nociceptors plays a critical role in pain, we reasoned that its interaction with FHF2 isoforms might regulate its current properties. Using whole-cell patch clamp in heterologous expression systems, we show that the expression of FHF2A in HEK293 cell line stably expressing Nav1.7 channels causes no change in activation, whereas FHF2B depolarizes activation. Both FHF2 isoforms depolarize fast-inactivation. Additionally, FHF2A causes an accumulation of inactivated channels at all frequencies tested due to a slowing of recovery from inactivation, whereas FHF2B has little effect on these properties of Nav1.7. Measurements of the Nav1.7 current in DRG neurons in which FHF2 levels are knocked down confirmed the effects of FHF2A on repriming, and FHF2B on activation, however FHF2A and B did not have an effect on fast inactivation. Our data demonstrates that FHF2 does indeed regulate the current properties of Nav1.7 and does so in an isoform and cell-specific manner.
Collapse
Affiliation(s)
- Philip R. Effraim
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Jianying Huang
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Angelika Lampert
- Institute of Physiology, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Severine Stamboulian
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Joel A. Black
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Sulayman D. Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G. Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
31
|
Abstract
Acute pain is adaptive, but chronic pain is a global challenge. Many chronic pain syndromes are peripheral in origin and reflect hyperactivity of peripheral pain-signaling neurons. Current treatments are ineffective or only partially effective and in some cases can be addictive, underscoring the need for better therapies. Molecular genetic studies have now linked multiple human pain disorders to voltage-gated sodium channels, including disorders characterized by insensitivity or reduced sensitivity to pain and others characterized by exaggerated pain in response to normally innocuous stimuli. Here, we review recent developments that have enhanced our understanding of pathophysiological mechanisms in human pain and advances in targeting sodium channels in peripheral neurons for the treatment of pain using novel and existing sodium channel blockers.
Collapse
Affiliation(s)
- Sulayman D Dib-Hajj
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut 06510, USA; .,Rehabilitation Research Center, Veterans Affairs, Connecticut Healthcare System, West Haven, Connecticut 06516, USA
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut 06510, USA; .,Rehabilitation Research Center, Veterans Affairs, Connecticut Healthcare System, West Haven, Connecticut 06516, USA
| |
Collapse
|
32
|
Pollard KJ, Sharma AD, Moore MJ. Neural microphysiological systems for in vitro modeling of peripheral nervous system disorders. ACTA ACUST UNITED AC 2019. [DOI: 10.2217/bem-2019-0018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PNS disease pathology is diverse and underappreciated. Peripheral neuropathy may result in sensory, motor or autonomic nerve dysfunction and can be induced by metabolic dysfunction, inflammatory dysfunction, cytotoxic pharmaceuticals, rare hereditary disorders or may be idiopathic. Current preclinical PNS disease research relies heavily on the use of rodent models. In vivo methods are effective but too time-consuming and expensive for high-throughput experimentation. Conventional in vitro methods can be performed with high throughput but lack the biological complexity necessary to directly model in vivo nerve structure and function. In this review, we survey in vitro PNS model systems and propose that 3D-bioengineered microphysiological nerve tissue can improve in vitro–in vivo extrapolation and expand the capabilities of in vitro PNS disease modeling.
Collapse
Affiliation(s)
- Kevin J Pollard
- Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA
| | | | - Michael J Moore
- Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA
- AxoSim, Inc., New Orleans, LA 70112, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| |
Collapse
|
33
|
Israel MR, Tanaka BS, Castro J, Thongyoo P, Robinson SD, Zhao P, Deuis JR, Craik DJ, Durek T, Brierley SM, Waxman SG, Dib-Hajj SD, Vetter I. Na V 1.6 regulates excitability of mechanosensitive sensory neurons. J Physiol 2019; 597:3751-3768. [PMID: 31087362 DOI: 10.1113/jp278148] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 05/08/2019] [Indexed: 12/20/2022] Open
Abstract
KEY POINTS Voltage-gated sodium channels are critical for peripheral sensory neuron transduction and have been implicated in a number of painful and painless disorders. The β-scorpion toxin, Cn2, is selective for NaV 1.6 in dorsal root ganglion neurons. NaV 1.6 plays an essential role in peripheral sensory neurons, specifically at the distal terminals of mechanosensing fibres innervating the skin and colon. NaV 1.6 activation also leads to enhanced response to mechanical stimulus in vivo. This works highlights the use of toxins in elucidating pain pathways moreover the importance of non-peripherally restricted NaV isoforms in pain generation. ABSTRACT Peripheral sensory neurons express multiple voltage-gated sodium channels (NaV ) critical for the initiation and propagation of action potentials and transmission of sensory input. Three pore-forming sodium channel isoforms are primarily expressed in the peripheral nervous system (PNS): NaV 1.7, NaV 1.8 and NaV 1.9. These sodium channels have been implicated in painful and painless channelopathies and there has been intense interest in them as potential therapeutic targets in human pain. Emerging evidence suggests NaV 1.6 channels are an important isoform in pain sensing. This study aimed to assess, using pharmacological approaches, the function of NaV 1.6 channels in peripheral sensory neurons. The potent and NaV 1.6 selective β-scorpion toxin Cn2 was used to assess the effect of NaV 1.6 channel activation in the PNS. The multidisciplinary approach included Ca2+ imaging, whole-cell patch-clamp recordings, skin-nerve and gut-nerve preparations and in vivo behavioural assessment of pain. Cn2 facilitates NaV 1.6 early channel opening, and increased persistent and resurgent currents in large-diameter dorsal root ganglion (DRG) neurons. This promotes enhanced excitatory drive and tonic action potential firing in these neurons. In addition, NaV 1.6 channel activation in the skin and gut leads to increased response to mechanical stimuli. Finally, intra-plantar injection of Cn2 causes mechanical but not thermal allodynia. This study confirms selectivity of Cn2 on NaV 1.6 channels in sensory neurons. Activation of NaV 1.6 channels, in terminals of the skin and viscera, leads to profound changes in neuronal responses to mechanical stimuli. In conclusion, sensory neurons expressing NaV 1.6 are important for the transduction of mechanical information in sensory afferents innervating the skin and viscera.
Collapse
Affiliation(s)
- Mathilde R Israel
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Brian S Tanaka
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT, 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Joel Castro
- Visceral Pain Research Group, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, 5042, Australia.,Hopwood Centre for Neurobiology and Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, 5000, Australia
| | - Panumart Thongyoo
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Samuel D Robinson
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Peng Zhao
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT, 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Jennifer R Deuis
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - David J Craik
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Thomas Durek
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, 5042, Australia.,Hopwood Centre for Neurobiology and Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, 5000, Australia
| | - Stephen G Waxman
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT, 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Sulayman D Dib-Hajj
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT, 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Irina Vetter
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia.,School of Pharmacy, Pharmacy Australia Centre of Excellence, 20 Cornwall St, Woolloongabba, Queensland, 4102, Australia
| |
Collapse
|
34
|
Zhao F, Tang Q, Xu J, Wang S, Li S, Zou X, Cao Z. Dehydrocrenatidine Inhibits Voltage-Gated Sodium Channels and Ameliorates Mechanic Allodia in a Rat Model of Neuropathic Pain. Toxins (Basel) 2019; 11:E229. [PMID: 31003411 PMCID: PMC6521113 DOI: 10.3390/toxins11040229] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 12/27/2022] Open
Abstract
Picrasma quassioides (D. Don) Benn, a medical plant, is used in clinic to treat inflammation, pain, sore throat, and eczema. The alkaloids are the main active components in P. quassioides. In this study, we examined the analgesic effect of dehydrocrenatidine (DHCT), a β-carboline alkaloid abundantly found in P. quassioides in a neuropathic pain rat model of a sciatic nerve chronic constriction injury. DHCT dose-dependently attenuated the mechanic allodynia. In acutely isolated dorsal root ganglion, DHCT completely suppressed the action potential firing. Further electrophysiological characterization demonstrated that DHCT suppressed both tetrodotoxin-resistant (TTX-R) and sensitive (TTX-S) voltage-gated sodium channel (VGSC) currents with IC50 values of 12.36 μM and 4.87 µM, respectively. DHCT shifted half-maximal voltage (V1/2) of inactivation to hyperpolarizing direction by ~16.7 mV in TTX-S VGSCs. In TTX-R VGSCs, DHCT shifted V1/2 of inactivation voltage to hyperpolarizing direction and V1/2 of activation voltage to more depolarizing potential by ~23.9 mV and ~12.2 mV, respectively. DHCT preferred to interact with an inactivated state of VGSCs and prolonged the repriming time in both TTX-S and TTX-R VGSCs, transiting the channels into a slow inactivated state from a fast inactivated state. Considered together, these data demonstrated that the analgesic effect of DHCT was likely though the inhibition of neuronal excitability.
Collapse
Affiliation(s)
- Fang Zhao
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Qinglian Tang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Jian Xu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Shuangyan Wang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Shaoheng Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Xiaohan Zou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Zhengyu Cao
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
35
|
Increased Resurgent Sodium Currents in Nav1.8 Contribute to Nociceptive Sensory Neuron Hyperexcitability Associated with Peripheral Neuropathies. J Neurosci 2019; 39:1539-1550. [PMID: 30617209 DOI: 10.1523/jneurosci.0468-18.2018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 10/22/2018] [Accepted: 11/25/2018] [Indexed: 11/21/2022] Open
Abstract
Neuropathic pain is a significant public health challenge, yet the underlying mechanisms remain poorly understood. Painful small fiber neuropathy (SFN) may be caused by gain-of-function mutations in Nav1.8, a sodium channel subtype predominantly expressed in peripheral nociceptive neurons. However, it is not clear how Nav1.8 disease mutations induce sensory neuron hyperexcitability. Here we studied two mutations in Nav1.8 associated with hypersensitive sensory neurons: G1662S reported in painful SFN; and T790A, which underlies increased pain behaviors in the Possum transgenic mouse strain. We show that, in male DRG neurons, these mutations, which impair inactivation, significantly increase TTX-resistant resurgent sodium currents mediated by Nav1.8. The G1662S mutation doubled resurgent currents, and the T790A mutation increased them fourfold. These unusual currents are typically evoked during the repolarization phase of action potentials. We show that the T790A mutation greatly enhances DRG neuron excitability by reducing current threshold and increasing firing frequency. Interestingly, the mutation endows DRG neurons with multiple early afterdepolarizations and leads to substantial prolongation of action potential duration. In DRG neurons, siRNA knockdown of sodium channel β4 subunits fails to significantly alter T790A current density but reduces TTX-resistant resurgent currents by 56%. Furthermore, DRG neurons expressing T790A channels exhibited reduced excitability with fewer early afterdepolarizations and narrower action potentials after β4 knockdown. Together, our data demonstrate that open-channel block of TTX-resistant currents, enhanced by gain-of-function mutations in Nav1.8, can make major contributions to the hyperexcitability of nociceptive neurons, likely leading to altered sensory phenotypes including neuropathic pain in SFN.SIGNIFICANCE STATEMENT This work demonstrates that two disease mutations in the voltage-gated sodium channel Nav1.8 that induce nociceptor hyperexcitability increase resurgent currents. Nav1.8 is crucial for pain sensations. Because resurgent currents are evoked during action potential repolarization, they can be crucial regulators of action potential activity. Our data indicate that increased Nav1.8 resurgent currents in DRG neurons greatly prolong action potential duration and enhance repetitive firing. We propose that Nav1.8 open-channel block is a major factor in Nav1.8-associated pain mechanisms and that targeting the molecular mechanism underlying these unique resurgent currents represents a novel therapeutic target for the treatment of aberrant pain sensations.
Collapse
|
36
|
|
37
|
Zhang N, Chin JS, Chew SY. Localised non-viral delivery of nucleic acids for nerve regeneration in injured nervous systems. Exp Neurol 2018; 319:112820. [PMID: 30195695 DOI: 10.1016/j.expneurol.2018.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/31/2018] [Accepted: 09/05/2018] [Indexed: 02/07/2023]
Abstract
Axons damaged by traumatic injuries are often unable to spontaneously regenerate in the adult central nervous system (CNS). Although the peripheral nervous system (PNS) has some regenerative capacity, its ability to regrow remains limited across large lesion gaps due to scar tissue formation. Nucleic acid therapy holds the potential of improving regeneration by enhancing the intrinsic growth ability of neurons and overcoming the inhibitory environment that prevents neurite outgrowth. Nucleic acids modulate gene expression by over-expression of neuronal growth factor or silencing growth-inhibitory molecules. Although in vitro outcomes appear promising, the lack of efficient non-viral nucleic acid delivery methods to the nervous system has limited the application of nucleic acid therapeutics to patients. Here, we review the recent development of efficient non-viral nucleic acid delivery platforms, as applied to the nervous system, including the transfection vectors and carriers used, as well as matrices and scaffolds that are currently used. Additionally, we will discuss possible improvements for localised nucleic acid delivery.
Collapse
Affiliation(s)
- Na Zhang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore
| | - Jiah Shin Chin
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore; NTU Institute of Health Technologies, Interdisciplinary Graduate School, Nanyang Technological University, 639798, Singapore
| | - Sing Yian Chew
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore.
| |
Collapse
|
38
|
Involvement of advillin in somatosensory neuron subtype-specific axon regeneration and neuropathic pain. Proc Natl Acad Sci U S A 2018; 115:E8557-E8566. [PMID: 30126982 PMCID: PMC6130359 DOI: 10.1073/pnas.1716470115] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
An estimated 20 million people in the United States have chronic neuropathic pain, but current analgesics are nonspecific or insufficiently effective. Here we show that advillin, a sensory neuron-specific protein, modulates axonal regeneration of a specific subset of pain-sensing afferent neurons (nociceptors) that binds with isolectin B4 and neuropathic pain. In addition, we identify the cell behavior of advillin shed-off from the growth cone in the context of axonal regeneration and thus detected advillin protein in the cerebrospinal fluid in mice with painful peripheral neuropathy. Advillin is a potential biosignature to diagnose the lesion cause of neuropathic pain associated with isolectin B4+ nociceptors. Advillin is a sensory neuron-specific actin-binding protein expressed at high levels in all types of somatosensory neurons in early development. However, the precise role of advillin in adulthood is largely unknown. Here we reveal advillin expression restricted to isolectin B4-positive (IB4+) neurons in the adult dorsal root ganglia (DRG). Advillin knockout (KO) specifically impaired axonal regeneration in adult IB4+ DRG neurons. During axon regeneration, advillin was expressed at the very tips of filopodia and modulated growth cone formation by interacting with and regulating focal-adhesion–related proteins. The advillin-containing focal-adhesion protein complex was shed from neurite tips during neurite retraction and was detectable in cerebrospinal fluid in experimental autoimmune encephalomyelitis, oxaliplatin-induced peripheral neuropathy, and chronic constriction injury of the sciatic nerve. In addition, advillin KO disturbed experimental autoimmune encephalomyelitis-induced neural plasticity in the spinal-cord dorsal horn and aggravated neuropathic pain. Our study highlights a role for advillin in growth cone formation, axon regeneration, and neuropathic pain associated with IB4+ DRG neurons in adulthood.
Collapse
|
39
|
Stenström P, Manzanares D, Zhang Y, Ceña V, Malkoch M. Evaluation of Amino-Functional Polyester Dendrimers Based on Bis-MPA as Nonviral Vectors for siRNA Delivery. Molecules 2018; 23:E2028. [PMID: 30110914 PMCID: PMC6222295 DOI: 10.3390/molecules23082028] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 11/23/2022] Open
Abstract
Herein, we present the first evaluation of cationic dendrimers based on 2,2-bis(methylol)propionic acid (bis-MPA) as nonviral vectors for transfection of short interfering RNA (siRNA) in cell cultures. The study encompassed dendrimers of generation one to four (G1⁻G4), modified to bear 6⁻48 amino end-groups, where the G2⁻G4 proved to be capable of siRNA complexation and protection against RNase-mediated degradation. The dendrimers were nontoxic to astrocytes, glioma (C6), and glioblastoma (U87), while G3 and G4 exhibited concentration dependent toxicity towards primary neurons. The G2 showed no toxicity to primary neurons at any of the tested concentrations. Fluorescence microscopy experiments suggested that the dendrimers are highly efficient at endo-lysosomal escape since fluorescently labeled dendrimers were localized specifically in mitochondria, and diffuse cytosolic distribution of fluorescent siRNA complexed by dendrimers was observed. This is a desired feature for intracellular drug delivery, since the endocytic pathway otherwise transfers the drugs into lysosomes where they can be degraded without reaching their intended target. siRNA-transfection was successful in C6 and U87 cell lines using the G3 and G4 dendrimers followed by a decrease of approximately 20% of target protein p42-MAPK expression.
Collapse
Affiliation(s)
- Patrik Stenström
- Fiber and Polymer Technology, KTH Royal Institute of Technology, 10044 Stockholm, Sweden.
| | - Dario Manzanares
- Unidad Asociada Neurodeath, Universidad de Castilla-La Mancha, 02006 Albacete, Spain.
- CIBERNED, Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Yuning Zhang
- Fiber and Polymer Technology, KTH Royal Institute of Technology, 10044 Stockholm, Sweden.
| | - Valentin Ceña
- Unidad Asociada Neurodeath, Universidad de Castilla-La Mancha, 02006 Albacete, Spain.
- CIBERNED, Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Michael Malkoch
- Fiber and Polymer Technology, KTH Royal Institute of Technology, 10044 Stockholm, Sweden.
| |
Collapse
|
40
|
Viventi S, Dottori M. Modelling the dorsal root ganglia using human pluripotent stem cells: A platform to study peripheral neuropathies. Int J Biochem Cell Biol 2018; 100:61-68. [PMID: 29772357 DOI: 10.1016/j.biocel.2018.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/08/2018] [Accepted: 05/13/2018] [Indexed: 12/28/2022]
Abstract
Sensory neurons of the dorsal root ganglia (DRG) are the primary responders to stimuli inducing feelings of touch, pain, temperature, vibration, pressure and muscle tension. They consist of multiple subpopulations based on their morphology, molecular and functional properties. Our understanding of DRG sensory neurons has been predominantly driven by rodent studies and using transformed cell lines, whereas less is known about human sensory DRG neurons simply because of limited availability of human tissue. Although these previous studies have been fundamental for our understanding of the sensory system, it is imperative to profile human DRG subpopulations as it is becoming evident that human sensory neurons do not share the identical molecular and functional properties found in other species. Furthermore, there are wide range of diseases and disorders that directly/indirectly cause sensory neuronal degeneration or dysfunctionality. Having an in vitro source of human DRG sensory neurons is paramount for studying their development, unique neuronal properties and for accelerating regenerative therapies to treat sensory neuropathies. Here we review the major studies describing generation of DRG sensory neurons from human pluripotent stem cells and fibroblasts and the gaps that need to be addressed for using in vitro-generated human DRG neurons to model human DRG tissue.
Collapse
Affiliation(s)
- Serena Viventi
- Department of Biomedical Engineering, University of Melbourne, Australia
| | - Mirella Dottori
- Department of Biomedical Engineering, University of Melbourne, Australia; Illawarra Health and Medical Research Institute, Centre for Molecular and Medical Bioscience, University of Wollongong, Australia.
| |
Collapse
|
41
|
Yang Y, Adi T, Effraim PR, Chen L, Dib‐Hajj SD, Waxman SG. Reverse pharmacogenomics: carbamazepine normalizes activation and attenuates thermal hyperexcitability of sensory neurons due to Na v 1.7 mutation I234T. Br J Pharmacol 2018; 175:2261-2271. [PMID: 28658526 PMCID: PMC5980548 DOI: 10.1111/bph.13935] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 04/17/2017] [Accepted: 06/05/2017] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND PURPOSE Pharmacotherapy for pain currently involves trial and error. A previous study on inherited erythromelalgia (a genetic model of neuropathic pain due to mutations in the sodium channel, Nav 1.7) used genomics, structural modelling and biophysical and pharmacological analyses to guide pharmacotherapy and showed that carbamazepine normalizes voltage dependence of activation of the Nav 1.7-S241T mutant channel, reducing pain in patients carrying this mutation. However, whether this approach is applicable to other Nav channel mutants is still unknown. EXPERIMENTAL APPROACH We used structural modelling, patch clamp and multi-electrode array (MEA) recording to assess the effects of carbamazepine on Nav 1.7-I234T mutant channels and on the firing of dorsal root ganglion (DRG) sensory neurons expressing these mutant channels. KEY RESULTS In a reverse engineering approach, structural modelling showed that the I234T mutation is located in atomic proximity to the carbamazepine-responsive S241T mutation and that activation of Nav 1.7-I234T mutant channels, from patients who are known to respond to carbamazepine, is partly normalized with a clinically relevant concentration (30 μM) of carbamazepine. There was significantly higher firing in intact sensory neurons expressing Nav 1.7-I234T channels, compared with neurons expressing the normal channels (Nav 1.7-WT). Pre-incubation with 30 μM carbamazepine also significantly reduced the firing of intact DRG sensory neurons expressing Nav 1.7-I234T channels. Although the expected use-dependent inhibition of Nav 1.7-WT channels by carbamazepine was confirmed, carbamazepine did not enhance use-dependent inhibition of Nav 1.7-I234T mutant channels. CONCLUSION AND IMPLICATIONS These results support the utility of a pharmacogenomic approach to treatment of pain in patients carrying sodium channel variants. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.
Collapse
Affiliation(s)
- Yang Yang
- Department of NeurologyYale University School of MedicineNew HavenCTUSA
- Center for Neuroscience and Regeneration ResearchYale University School of MedicineNew HavenCTUSA
- Rehabilitation Research CenterVA Connecticut Healthcare SystemWest HavenCTUSA
| | - Talia Adi
- Department of NeurologyYale University School of MedicineNew HavenCTUSA
- Center for Neuroscience and Regeneration ResearchYale University School of MedicineNew HavenCTUSA
- Rehabilitation Research CenterVA Connecticut Healthcare SystemWest HavenCTUSA
| | - Philip R Effraim
- Department of NeurologyYale University School of MedicineNew HavenCTUSA
- Center for Neuroscience and Regeneration ResearchYale University School of MedicineNew HavenCTUSA
- Rehabilitation Research CenterVA Connecticut Healthcare SystemWest HavenCTUSA
- Department of AnesthesiologyYale University School of MedicineNew HavenCTUSA
| | - Lubin Chen
- Department of NeurologyYale University School of MedicineNew HavenCTUSA
- Center for Neuroscience and Regeneration ResearchYale University School of MedicineNew HavenCTUSA
- Rehabilitation Research CenterVA Connecticut Healthcare SystemWest HavenCTUSA
| | - Sulayman D Dib‐Hajj
- Department of NeurologyYale University School of MedicineNew HavenCTUSA
- Center for Neuroscience and Regeneration ResearchYale University School of MedicineNew HavenCTUSA
- Rehabilitation Research CenterVA Connecticut Healthcare SystemWest HavenCTUSA
| | - Stephen G Waxman
- Department of NeurologyYale University School of MedicineNew HavenCTUSA
- Center for Neuroscience and Regeneration ResearchYale University School of MedicineNew HavenCTUSA
- Rehabilitation Research CenterVA Connecticut Healthcare SystemWest HavenCTUSA
| |
Collapse
|
42
|
Chen L, Huang J, Zhao P, Persson AK, Dib-Hajj FB, Cheng X, Tan A, Waxman SG, Dib-Hajj SD. Conditional knockout of Na V1.6 in adult mice ameliorates neuropathic pain. Sci Rep 2018; 8:3845. [PMID: 29497094 PMCID: PMC5832877 DOI: 10.1038/s41598-018-22216-w] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/19/2018] [Indexed: 01/09/2023] Open
Abstract
Voltage-gated sodium channels NaV1.7, NaV1.8 and NaV1.9 have been the focus for pain studies because their mutations are associated with human pain disorders, but the role of NaV1.6 in pain is less understood. In this study, we selectively knocked out NaV1.6 in dorsal root ganglion (DRG) neurons, using NaV1.8-Cre directed or adeno-associated virus (AAV)-Cre mediated approaches, and examined the specific contribution of NaV1.6 to the tetrodotoxin-sensitive (TTX-S) current in these neurons and its role in neuropathic pain. We report here that NaV1.6 contributes up to 60% of the TTX-S current in large, and 34% in small DRG neurons. We also show NaV1.6 accumulates at nodes of Ranvier within the neuroma following spared nerve injury (SNI). Although NaV1.8-Cre driven NaV1.6 knockout does not alter acute, inflammatory or neuropathic pain behaviors, AAV-Cre mediated NaV1.6 knockout in adult mice partially attenuates SNI-induced mechanical allodynia. Additionally, AAV-Cre mediated NaV1.6 knockout, mostly in large DRG neurons, significantly attenuates excitability of these neurons after SNI and reduces NaV1.6 accumulation at nodes of Ranvier at the neuroma. Together, NaV1.6 in NaV1.8-positive neurons does not influence pain thresholds under normal or pathological conditions, but NaV1.6 in large NaV1.8-negative DRG neurons plays an important role in neuropathic pain.
Collapse
Affiliation(s)
- Lubin Chen
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Jianying Huang
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Anna-Karin Persson
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Fadia B Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Xiaoyang Cheng
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Andrew Tan
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA. .,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA. .,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA.
| |
Collapse
|
43
|
Huang J, Mis MA, Tanaka B, Adi T, Estacion M, Liu S, Walker S, Dib-Hajj SD, Waxman SG. Atypical changes in DRG neuron excitability and complex pain phenotype associated with a Na v1.7 mutation that massively hyperpolarizes activation. Sci Rep 2018; 8:1811. [PMID: 29379075 PMCID: PMC5788866 DOI: 10.1038/s41598-018-20221-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/16/2018] [Indexed: 02/06/2023] Open
Abstract
Sodium channel Nav1.7 plays a central role in pain-signaling: gain-of-function Nav1.7 mutations usually cause severe pain and loss-of-function mutations produce insensitivity to pain. The Nav1.7 I234T gain-of-function mutation, however, is linked to a dual clinical presentation of episodic pain, together with absence of pain following fractures, and corneal anesthesia. How a Nav1.7 mutation that produces gain-of-function at the channel level causes clinical loss-of-function has remained enigmatic. We show by current-clamp that expression of I234T in dorsal root ganglion (DRG) neurons produces a range of membrane depolarizations including a massive shift to >−40 mV that reduces excitability in a small number of neurons. Dynamic-clamp permitted us to mimic the heterozygous condition via replacement of 50% endogenous wild-type Nav1.7 channels by I234T, and confirmed that the I234T conductance could drastically depolarize DRG neurons, resulting in loss of excitability. We conclude that attenuation of pain sensation by I234T is caused by massively depolarized membrane potential of some DRG neurons which is partly due to enhanced overlap between activation and fast-inactivation, impairing their ability to fire. Our results demonstrate how a Nav1.7 mutation that produces channel gain-of-function can contribute to a dual clinical presentation that includes loss of pain sensation at the clinical level.
Collapse
Affiliation(s)
- Jianying Huang
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA, 06510.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA, 06516
| | - Malgorzata A Mis
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA, 06510.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA, 06516
| | - Brian Tanaka
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA, 06510.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA, 06516
| | - Talia Adi
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA, 06510.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA, 06516
| | - Mark Estacion
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA, 06510.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA, 06516
| | - Shujun Liu
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA, 06510.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA, 06516
| | - Suellen Walker
- Developmental Neurosciences Program, Department of Anaesthesia and Pain Medicine, UCL Great Ormond Street Hospital, London, WC1N 1EH, UK
| | - Sulayman D Dib-Hajj
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA, 06510.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA, 06516
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA, 06510. .,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA, 06516.
| |
Collapse
|
44
|
Martin C, Stoffer C, Mohammadi M, Hugo J, Leipold E, Oehler B, Rittner HL, Blum R. Na V1.9 Potentiates Oxidized Phospholipid-Induced TRP Responses Only under Inflammatory Conditions. Front Mol Neurosci 2018; 11:7. [PMID: 29410612 PMCID: PMC5787077 DOI: 10.3389/fnmol.2018.00007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 01/08/2018] [Indexed: 12/13/2022] Open
Abstract
Oxidized phospholipids (OxPL) like oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (OxPAPC) were recently identified as novel proalgesic targets in acute and chronic inflammatory pain. These endogenous chemical irritants are generated in inflamed tissue and mediate their pain-inducing function by activating the transient receptor potential channels TRPA1 and TRPV1 expressed in sensory neurons. Notably, prototypical therapeutics interfering with OxPL were shown to inhibit TRP channel activation and pain behavior. Here, we asked how OxPL excite primary sensory neurons of dorsal root ganglia (DRG neurons from mice of either sex). Acute stimulation of sensory neurons with the prototypical OxPL 1-palmitoyl-2-glutaryl-sn-glycero-3-phosphocholine (PGPC) evoked repetitive calcium spikes in small-diameter neurons. As NaV1.9, a voltage-gated sodium channel involved in nociceptor excitability, was previously shown to be essential for the generation of calcium spikes in motoneurons, we asked if this channel is also important for OxPL mediated calcium spike and action potential generation in nociceptors. In wild-type and NaV1.9-deficient neurons, the action potential firing rate and the calcium spike frequency to an acute PGPC stimulus was similar. When preincubated with inflammatory mediators, both, the action potential firing rate and the calcium spike frequency were markedly increased in response to an acute PGPC stimulus. However, this potentiating effect was completely lost in NaV1.9-deficient small-diameter neurons. After treatment with inflammatory mediators, the resting membrane potential of NaV1.9 KO neurons was slightly more negative than that of wild-type control neurons. This suggests that NaV1.9 channels are active under this condition and therefore increases the ease with which action potentials are elicited after OxPL stimulation. In summary, our data suggest that NaV1.9 has a switch function to potentiate the receptor potentials induced by OxPL under inflammatory conditions. Since human NaV1.9 has been shown to mediate painful and painless channelopathies, this study provides new insights into the mechanism by which NaV1.9 amplifies stimuli of endogenous irritants under inflammatory conditions.
Collapse
Affiliation(s)
- Corinna Martin
- Institute of Clinical Neurobiology, University Hospital Würzburg, University of Würzburg, Würzburg, Germany.,Department of Anesthesiology, University Hospital Würzburg, Würzburg, Germany
| | - Carolin Stoffer
- Institute of Clinical Neurobiology, University Hospital Würzburg, University of Würzburg, Würzburg, Germany
| | - Milad Mohammadi
- Institute of Clinical Neurobiology, University Hospital Würzburg, University of Würzburg, Würzburg, Germany.,Department of Anesthesiology, University Hospital Würzburg, Würzburg, Germany
| | - Julian Hugo
- Institute of Clinical Neurobiology, University Hospital Würzburg, University of Würzburg, Würzburg, Germany.,Department of Anesthesiology, University Hospital Würzburg, Würzburg, Germany
| | - Enrico Leipold
- Department of Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena and Jena University Hospital, Jena, Germany
| | - Beatrice Oehler
- Department of Anesthesiology, University Hospital Würzburg, Würzburg, Germany
| | - Heike L Rittner
- Department of Anesthesiology, University Hospital Würzburg, Würzburg, Germany
| | - Robert Blum
- Institute of Clinical Neurobiology, University Hospital Würzburg, University of Würzburg, Würzburg, Germany
| |
Collapse
|
45
|
Adi T, Estacion M, Schulman BR, Vernino S, Dib-Hajj SD, Waxman SG. A novel gain-of-function Na v1.7 mutation in a carbamazepine-responsive patient with adult-onset painful peripheral neuropathy. Mol Pain 2018; 14:1744806918815007. [PMID: 30392441 PMCID: PMC6856981 DOI: 10.1177/1744806918815007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/12/2018] [Indexed: 12/13/2022] Open
Abstract
Voltage-gated sodium channel Nav1.7 is a threshold channel in peripheral dorsal root ganglion (DRG), trigeminal ganglion, and sympathetic ganglion neurons. Gain-of-function mutations in Nav1.7 have been shown to increase excitability in DRG neurons and have been linked to rare Mendelian and more common pain disorders. Discovery of Nav1.7 variants in patients with pain disorders may expand the spectrum of painful peripheral neuropathies associated with a well-defined molecular target, thereby providing a basis for more targeted approaches for treatment. We screened the genome of a patient with adult-onset painful peripheral neuropathy characterized by severe burning pain and report here the new Nav1.7-V810M variant. Voltage-clamp recordings were used to assess the effects of the mutation on biophysical properties of Nav1.7 and the response of the mutant channel to treatment with carbamazepine (CBZ), and multi-electrode array (MEA) recordings were used to assess the effects of the mutation on the excitability of neonatal rat pup DRG neurons. The V810M variant increases current density, shifts activation in a hyperpolarizing direction, and slows kinetics of deactivation, all gain-of-function attributes. We also show that DRG neurons that express the V810M variant become hyperexcitable. The patient responded to treatment with CBZ. Although CBZ did not depolarize activation of the mutant channel, it enhanced use-dependent inhibition. Our results demonstrate the presence of a novel gain-of-function variant of Nav1.7 in a patient with adult-onset painful peripheral neuropathy and the responsiveness of that patient to treatment with CBZ, which is likely due to the classical mechanism of use-dependent inhibition.
Collapse
Affiliation(s)
- Talia Adi
- Department of Neurology, Yale University School of Medicine, New
Haven, CT, USA
- Center for Neuroscience and Regeneration Research, Veterans
Affairs Medical Center, West Haven, CT, USA
| | - Mark Estacion
- Department of Neurology, Yale University School of Medicine, New
Haven, CT, USA
- Center for Neuroscience and Regeneration Research, Veterans
Affairs Medical Center, West Haven, CT, USA
| | - Betsy R Schulman
- Department of Neurology, Yale University School of Medicine, New
Haven, CT, USA
- Center for Neuroscience and Regeneration Research, Veterans
Affairs Medical Center, West Haven, CT, USA
| | - Steven Vernino
- Department of Neurology and Neurotherapeutics, UT Southwestern
Medical Center, Dallas, TX, USA
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New
Haven, CT, USA
- Center for Neuroscience and Regeneration Research, Veterans
Affairs Medical Center, West Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New
Haven, CT, USA
- Center for Neuroscience and Regeneration Research, Veterans
Affairs Medical Center, West Haven, CT, USA
| |
Collapse
|
46
|
Nav1.7-A1632G Mutation from a Family with Inherited Erythromelalgia: Enhanced Firing of Dorsal Root Ganglia Neurons Evoked by Thermal Stimuli. J Neurosci 2017; 36:7511-22. [PMID: 27413160 DOI: 10.1523/jneurosci.0462-16.2016] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 06/06/2016] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Voltage-gated sodium channel Nav1.7 is a central player in human pain. Mutations in Nav1.7 produce several pain syndromes, including inherited erythromelalgia (IEM), a disorder in which gain-of-function mutations render dorsal root ganglia (DRG) neurons hyperexcitable. Although patients with IEM suffer from episodes of intense burning pain triggered by warmth, the effects of increased temperature on DRG neurons expressing mutant Nav1.7 channels have not been well documented. Here, using structural modeling, voltage-clamp, current-clamp, and multielectrode array recordings, we have studied a newly identified Nav1.7 mutation, Ala1632Gly, from a multigeneration family with IEM. Structural modeling suggests that Ala1632 is a molecular hinge and that the Ala1632Gly mutation may affect channel gating. Voltage-clamp recordings revealed that the Nav1.7-A1632G mutation hyperpolarizes activation and depolarizes fast-inactivation, both gain-of-function attributes at the channel level. Whole-cell current-clamp recordings demonstrated increased spontaneous firing, lower current threshold, and enhanced evoked firing in rat DRG neurons expressing Nav1.7-A1632G mutant channels. Multielectrode array recordings further revealed that intact rat DRG neurons expressing Nav1.7-A1632G mutant channels are more active than those expressing Nav1.7 WT channels. We also showed that physiologically relevant thermal stimuli markedly increase the mean firing frequencies and the number of active rat DRG neurons expressing Nav1.7-A1632G mutant channels, whereas the same thermal stimuli only increase these parameters slightly in rat DRG neurons expressing Nav1.7 WT channels. The response of DRG neurons expressing Nav1.7-A1632G mutant channels upon increase in temperature suggests a cellular basis for warmth-triggered pain in IEM. SIGNIFICANCE STATEMENT Inherited erythromelalgia (IEM), a severe pain syndrome characterized by episodes of intense burning pain triggered by warmth, is caused by mutations in sodium channel Nav1.7, which are preferentially expressed in sensory and sympathetic neurons. More than 20 gain-of-function Nav1.7 mutations have been identified from IEM patients, but the question of how warmth triggers episodes of pain in IEM has not been well addressed. Combining multielectrode array, voltage-clamp, and current-clamp recordings, we assessed a newly identified IEM mutation (Nav1.7-A1632G) from a multigeneration family. Our data demonstrate gain-of-function attributes at the channel level and differential effects of physiologically relevant thermal stimuli on the excitability of DRG neurons expressing mutant and WT Nav1.7 channels, suggesting a cellular mechanism for warmth-triggered pain episodes in IEM patients.
Collapse
|
47
|
Huang J, Vanoye CG, Cutts A, Goldberg YP, Dib-Hajj SD, Cohen CJ, Waxman SG, George AL. Sodium channel NaV1.9 mutations associated with insensitivity to pain dampen neuronal excitability. J Clin Invest 2017; 127:2805-2814. [PMID: 28530638 DOI: 10.1172/jci92373] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/23/2017] [Indexed: 02/05/2023] Open
Abstract
Voltage-gated sodium channel (NaV) mutations cause genetic pain disorders that range from severe paroxysmal pain to a congenital inability to sense pain. Previous studies on NaV1.7 and NaV1.8 established clear relationships between perturbations in channel function and divergent clinical phenotypes. By contrast, studies of NaV1.9 mutations have not revealed a clear relationship of channel dysfunction with the associated and contrasting clinical phenotypes. Here, we have elucidated the functional consequences of a NaV1.9 mutation (L1302F) that is associated with insensitivity to pain. We investigated the effects of L1302F and a previously reported mutation (L811P) on neuronal excitability. In transfected heterologous cells, the L1302F mutation caused a large hyperpolarizing shift in the voltage-dependence of activation, leading to substantially enhanced overlap between activation and steady-state inactivation relationships. In transfected small rat dorsal root ganglion neurons, expression of L1302F and L811P evoked large depolarizations of the resting membrane potential and impaired action potential generation. Therefore, our findings implicate a cellular loss of function as the basis for impaired pain sensation. We further demonstrated that a U-shaped relationship between the resting potential and the neuronal action potential threshold explains why NaV1.9 mutations that evoke small degrees of membrane depolarization cause hyperexcitability and familial episodic pain disorder or painful neuropathy, while mutations evoking larger membrane depolarizations cause hypoexcitability and insensitivity to pain.
Collapse
Affiliation(s)
- Jianying Huang
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine; and Rehabilitation Research Center, Veterans Administration Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Carlos G Vanoye
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Alison Cutts
- Xenon Pharmaceuticals, Burnaby, British Columbia, Canada
| | | | - Sulayman D Dib-Hajj
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine; and Rehabilitation Research Center, Veterans Administration Connecticut Healthcare System, West Haven, Connecticut, USA
| | | | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine; and Rehabilitation Research Center, Veterans Administration Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Alfred L George
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
48
|
Han C, Yang Y, Te Morsche RH, Drenth JPH, Politei JM, Waxman SG, Dib-Hajj SD. Familial gain-of-function Na v1.9 mutation in a painful channelopathy. J Neurol Neurosurg Psychiatry 2017; 88:233-240. [PMID: 27503742 DOI: 10.1136/jnnp-2016-313804] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/23/2016] [Accepted: 07/19/2016] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Gain-of-function mutations in Nav1.9 have been identified in three families with rare heritable pain disorders, and in patients with painful small-fibre neuropathy. Identification and functional assessment of new Nav1.9 mutations will help to elucidate the phenotypic spectrum of Nav1.9 channelopathies. METHODS Patients from a large family with early-onset pain symptoms were evaluated by clinical examination and genomic screening for mutations in SCN9A and SCN11A. Electrophysiological recordings and multistate modelling analysis were implemented for functional analyses. RESULTS A novel Nav1.9 mutation, p.Arg222His, was identified in patients with early-onset pain in distal extremities including joints and gastrointestinal disturbances, but was absent from an asymptomatic blood relative. This mutation alters channel structure by substituting the highly conserved first arginine residue in transmembrane segment 4 (domain 1), the voltage sensor, with histidine. Voltage-clamp recordings demonstrate a hyperpolarising shift and acceleration of activation of the p.Arg222His mutant channel, which make it easier to open the channel. When expressed in dorsal root ganglion neurons, mutant p.Arg222His channels increase excitability via a depolarisation of resting potential and increased evoked firing. CONCLUSIONS This study expands the spectrum of heritable pain disorders linked to gain-of-function mutations in Nav1.9, strengthening human validation of this channel as a potential therapeutic target for pain.
Collapse
Affiliation(s)
- Chongyang Han
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA.,Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA.,Center for Restoration of Nervous System Function, Veterans Affairs Medical Center, West Haven, Connecticut, USA
| | - Yang Yang
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA.,Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA.,Center for Restoration of Nervous System Function, Veterans Affairs Medical Center, West Haven, Connecticut, USA
| | - Rene H Te Morsche
- Department of Gastroenterology and Hepatology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Joost P H Drenth
- Department of Gastroenterology and Hepatology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Juan M Politei
- Department of Neurology, Fundación para el Estudio de las Enfermedades Neurometabólicas, Buenos Aires, Argentina
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA.,Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA.,Center for Restoration of Nervous System Function, Veterans Affairs Medical Center, West Haven, Connecticut, USA
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA.,Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA.,Center for Restoration of Nervous System Function, Veterans Affairs Medical Center, West Haven, Connecticut, USA
| |
Collapse
|
49
|
Estacion M, Waxman SG. Nonlinear effects of hyperpolarizing shifts in activation of mutant Na v1.7 channels on resting membrane potential. J Neurophysiol 2017; 117:1702-1712. [PMID: 28148645 DOI: 10.1152/jn.00898.2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/11/2017] [Accepted: 01/26/2017] [Indexed: 01/16/2023] Open
Abstract
The Nav1.7 sodium channel is preferentially expressed within dorsal root ganglion (DRG) and sympathetic ganglion neurons. Gain-of-function mutations that cause the painful disorder inherited erythromelalgia (IEM) shift channel activation in a hyperpolarizing direction. When expressed within DRG neurons, these mutations produce a depolarization of resting membrane potential (RMP). The biophysical basis for the depolarized RMP has to date not been established. To explore the effect on RMP of the shift in activation associated with a prototypical IEM mutation (L858H), we used dynamic-clamp models that represent graded shifts that fractionate the effect of the mutation on activation voltage dependence. Dynamic-clamp recording from DRG neurons using a before-and-after protocol for each cell made it possible, even in the presence of cell-to-cell variation in starting RMP, to assess the effects of these graded mutant models. Our results demonstrate a nonlinear, progressively larger effect on RMP as the shift in activation voltage dependence becomes more hyperpolarized. The observed differences in RMP were predicted by the "late" current of each mutant model. Since the depolarization of RMP imposed by IEM mutant channels is known, in itself, to produce hyperexcitability of DRG neurons, the development of pharmacological agents that normalize or partially normalize activation voltage dependence of IEM mutant channels merits further study.NEW & NOTEWORTHY Inherited erythromelalgia (IEM), the first human pain disorder linked to a sodium channel, is widely regarded as a genetic model of neuropathic pain. IEM is produced by Nav1.7 mutations that hyperpolarize activation. These mutations produce a depolarization of resting membrane potential (RMP) in dorsal root ganglion neurons. Using dynamic clamp to explore the effect on RMP of the shift in activation, we demonstrate a nonlinear effect on RMP as the shift in activation voltage dependence becomes more hyperpolarized.
Collapse
Affiliation(s)
- Mark Estacion
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and.,Rehabilitation Research Center, Department of Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and .,Rehabilitation Research Center, Department of Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
50
|
Shahbazi R, Ozcicek I, Ozturk G, Ulubayram K. Functionalized gold nanoparticles manifested as potent carriers for nucleolar targeting. NANOTECHNOLOGY 2017; 28:025103. [PMID: 27924783 DOI: 10.1088/1361-6528/28/2/025103] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
It is generally known that gold nanoparticles are localised in the cytoplasm and, if synthesised in small sizes or functionalized with specific proteins, they enter the cell nucleus. However, there is no report emphasising the importance of surface functionalization in their accumulation in the nucleolus. Here, for the first time in the literature, it is proposed that functionalization of gold nanoparticles with a thin layer of polyethyleneimine (PEI) spearheads them to the nucleolus of hard-to-transfect post-mitotic dorsal root ganglion neurones in a size-independent manner. As a potential for theranostic applications, it was found that functionalization with a thin layer of PEI affected the emission signal intensity of gold nanoparticles so that the cellular biodistribution of nanoparticles was visualised clearly under both confocal and two-photon microscopes.
Collapse
Affiliation(s)
- Reza Shahbazi
- Department of Nanotechnology and Nanomedicine, Hacettepe University, Ankara, Turkey
| | | | | | | |
Collapse
|