1
|
Clément BF, Petrella L, Wallimann L, Duru J, Tringides CM, Vörös J, Ruff T. An in vitro platform for characterizing axonal electrophysiology of individual human iPSC-derived nociceptors. Biosens Bioelectron 2025; 281:117418. [PMID: 40215890 DOI: 10.1016/j.bios.2025.117418] [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: 12/20/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 05/04/2025]
Abstract
Neuropathic pain is characterized by aberrant activity of specific nociceptor populations, as demonstrated through functional assessments such as microneurography. Current treatments against severe forms of neuropathic pain demonstrate insufficient efficacy or lead to unwanted side effects as they fail to specifically target the affected nociceptors. Tools that can recapitulate aspects of microneurography in vitro would enable a more targeted compound screening. Therefore, we developed an in vitro platform combining a CMOS-based high-density microelectrode array with a polydimethylsiloxane (PDMS) guiding microstructure that captures the electrophysiological responses of individual axons. Human induced pluripotent stem cell-derived (hiPSC) sensory neurons were cultured in a way that allowed axons to be distributed through parallel 4 ×10μm microchannels exiting the seeding well before converging to a bigger axon-collecting channel. This configuration allowed the measurement of stimulation-induced responses of individual axons. Sensory neurons were found to exhibit a great diversity of electrophysiological response profiles that can be classified into different functional archetypes. Moreover, we show that some responses are affected by applying the TRPV1 agonist capsaicin. Overall, results using our platform demonstrate that we were able to distinguish individual axon responses, making the platform a promising tool for testing therapeutic candidates targeting particular sensory neuron subtypes.
Collapse
Affiliation(s)
- Blandine F Clément
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Gloriastrasse 37/39, Zurich, 8092, Switzerland
| | - Lorenzo Petrella
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Gloriastrasse 37/39, Zurich, 8092, Switzerland
| | - Lea Wallimann
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Gloriastrasse 37/39, Zurich, 8092, Switzerland
| | - Jens Duru
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Gloriastrasse 37/39, Zurich, 8092, Switzerland
| | - Christina M Tringides
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Gloriastrasse 37/39, Zurich, 8092, Switzerland
| | - János Vörös
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Gloriastrasse 37/39, Zurich, 8092, Switzerland.
| | - Tobias Ruff
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Gloriastrasse 37/39, Zurich, 8092, Switzerland.
| |
Collapse
|
2
|
Wood JN, Yan N, Huang J, Zhao J, Akopian A, Cox JJ, Woods CG, Nassar MA. Sensory neuron sodium channels as pain targets; from cocaine to Journavx (VX-548, suzetrigine). J Gen Physiol 2025; 157:e202513778. [PMID: 40294084 PMCID: PMC12036950 DOI: 10.1085/jgp.202513778] [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: 02/07/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Voltage-gated sodium channels underpin electrical signaling in sensory neurons. Their activity is an essential element in the vast majority of pain conditions, making them significant drug targets. Sensory neuron sodium channels play roles not only in afferent signaling but also in a range of efferent regulatory mechanisms. Side effects through actions on other cell types and efferent signaling are thus important issues to address during analgesic drug development. As an example, the human genetic evidence for NaV1.7 as an ideal pain target contrasts with the side effects of NaV1.7 antagonists. In this review, we describe the history and progress toward the development of useful analgesic drugs and the renewed focus on NaV1.8 as a key target in pain treatment. NaV1.8 antagonists alone or in combination with other analgesics are likely to provide new opportunities for pain relief for the vast number of people (about 33% of the population) impacted by chronic pain, particularly present in aging populations.
Collapse
Affiliation(s)
- John N. Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, UCL, London, UK
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation, Shenzhen, China
| | - Jian Huang
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation, Shenzhen, China
| | - Jing Zhao
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, UCL, London, UK
| | - Armen Akopian
- Department of Endodontics, The School of the Dentistry, UTHSCSA, San Antonio, TX, USA
| | - James J. Cox
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, UCL, London, UK
| | | | - Mohammed A. Nassar
- School of Biosciences, Firth Court, University of Sheffield, Sheffield, UK
| |
Collapse
|
3
|
Resch FJ, Heber S, Shahi F, Zauner M, Ciotu CI, Gleiss A, Sator S, Fischer MJM. Human cold pain: a randomized crossover trial. Pain 2025; 166:1406-1417. [PMID: 39693244 PMCID: PMC12067611 DOI: 10.1097/j.pain.0000000000003503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 12/20/2024]
Abstract
ABSTRACT The mechanism causing cold pain in humans is unresolved. Animal data suggest a nonredundant contribution to cold pain for transient receptor potential channels TRPM8 and TRPA1 for detection and voltage-gated sodium channels Na V 1.7 and Na V 1.8 for conduction at these temperatures. We established an intradermal injection-based cold pain model, which allows pharmacologically addressing molecular targets at the site of cooling. Lidocaine, added to the injection solution as positive control, largely reduced cold-induced pain in 36 volunteers. The 4 mentioned molecular targets were blocked by antagonists in a double-blinded crossover trial. Pain induced by 3°C intradermal fluid was not reduced to a relevant extent by any of the 4 antagonists alone or by the quadruple combination. However, the temperature threshold for cold pain appeared shifted by the inhibition of TRPA1, TRPM8, and Na V 1.7 and to a lesser extent by Na V 1.8 inhibition, 4-fold inhibition decreased the threshold by 5.8°C. Further mechanisms contributing to human cold pain need to be considered.
Collapse
Affiliation(s)
- Felix J. Resch
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Stefan Heber
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Farzin Shahi
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Manuel Zauner
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Cosmin I. Ciotu
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Andreas Gleiss
- Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Sabine Sator
- Division of Special Anesthesia and Pain Medicine, Department of Anesthesia, Intensive Care and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Michael J. M. Fischer
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
Santiago C, Siegrist J, Africawala N, Handler A, Tasnim A, Anjum R, Turecek J, Lehnert BP, Renauld S, Choi J, Nolan-Tamariz M, Iskols M, Magee AR, Paradis S, Sharma N, Ginty DD. Activity-dependent development of the body's touch receptors. Neuron 2025:S0896-6273(25)00298-3. [PMID: 40381613 DOI: 10.1016/j.neuron.2025.04.015] [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: 09/22/2023] [Revised: 02/12/2025] [Accepted: 04/16/2025] [Indexed: 05/20/2025]
Abstract
We report a role for activity in the development of the primary sensory neurons that detect touch. Genetic deletion of Piezo2, the principal mechanosensitive ion channel in somatosensory neurons, caused profound changes in the formation of mechanosensory end-organ structures. Peripheral-nervous-system-specific deletion of the voltage-gated sodium channel Nav1.6 (Scn8a), which resulted in altered electrophysiological responses to mechanical stimuli, also disrupted somatosensory neuron morphologies, supporting a role for neuronal activity in end-organ formation. Single-cell RNA sequencing of Piezo2 mutants revealed changes in gene expression in sensory neurons activated by light mechanical forces, whereas other neuronal classes were minimally affected, and genetic deletion of Piezo2-dependent genes partially reproduced the defects in mechanosensory neuron structures observed in Piezo2 mutants. These findings indicate that mechanically evoked neuronal activity acts early in life to shape the maturation of mechanosensory end-organs that underlie our sense of gentle touch.
Collapse
Affiliation(s)
- Celine Santiago
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Julianna Siegrist
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Nusrat Africawala
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Annie Handler
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Aniqa Tasnim
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Rabia Anjum
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, MA 02453, USA
| | - Josef Turecek
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Brendan P Lehnert
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Sophia Renauld
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jinheon Choi
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Nolan-Tamariz
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Iskols
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Alexandra R Magee
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Suzanne Paradis
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, MA 02453, USA
| | - Nikhil Sharma
- Department of Biochemistry and Molecular Biophysics and Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Medical Center, New York, NY 10032, USA
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
5
|
Xie YF. Nav1.8 and Chronic Pain: From Laboratory Animals to Clinical Patients. Biomolecules 2025; 15:694. [PMID: 40427587 PMCID: PMC12108746 DOI: 10.3390/biom15050694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
As a subtype of voltage-gated sodium channel and predominantly expressed in the sensory neurons located in the dorsal root ganglion (DRG), the Nav1.8 channel encoded by the SCN10A gene is found to have different variants in patients suffering chronic pain or insensitivity to pain due to the gain-of-function or loss-of-function of Nav1.8 channels. In animal models of chronic pain, Nav1.8 is also verified to be involved, suggesting that Nav1.8 may be a potential target for treatment of chronic pain. Another voltage-gated sodium channel, Nav1.7, is also proposed to be a target for chronic pain, supported by clinical findings in patients and laboratory animal models; however, there is no Nav1.7-specific drug that has passed clinical trials, although they demonstrated satisfactory effects in laboratory animals. This discrepancy between clinical and preclinical studies may be related to the differences between humans and laboratory animals or due to the degeneracy in different sodium channels governing the DRG neuronal excitability, which is thought of as the underlying machinery of chronic pain and mostly studied. This review summarizes recent findings of Nav1.8 in chronic pain from clinics and laboratories and discusses the difference, which may be helpful for future investigation of Nav1.8 in chronic pain, considering the dilemma of the Nav1.7 channel in chronic pain.
Collapse
Affiliation(s)
- Yu-Feng Xie
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| |
Collapse
|
6
|
Senior M. Precision neuroscience. Nat Biotechnol 2025; 43:659-664. [PMID: 40295786 DOI: 10.1038/s41587-025-02673-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
|
7
|
Yogi A, Banderali U, Moreno MJ, Martina M. Preclinical Animal Models to Investigate the Role of Na v1.7 Ion Channels in Pain. Life (Basel) 2025; 15:640. [PMID: 40283194 PMCID: PMC12028925 DOI: 10.3390/life15040640] [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: 01/22/2025] [Revised: 04/03/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Chronic pain is a maladaptive neurological disease that remains a major global healthcare problem. Voltage-gated sodium channels (Navs) are major drivers of the excitability of sensory neurons, and the Nav subtype 1.7 (Nav1.7) has been shown to be critical for the transmission of pain-related signaling. This is highlighted by demonstrations that gain-of-function mutations in the Nav1.7 gene SCN9A result in various pain pathologies, whereas loss-of-function mutations cause complete insensitivity to pain. A substantial body of evidence demonstrates that chronic neuropathy and inflammation result in an upregulation of Nav1.7, suggesting that this channel contributes to pain transmission and sensation. As such, Nav1.7 is an attractive human-validated target for the treatment of pain. Nonetheless, a lack of subtype selectivity, insufficient efficacy, and adverse reactions are some of the issues that have hindered Nav1.7-targeted drug development. This review summarizes the pain behavior profiles mediated by Nav1.7 reported in multiple preclinical models, outlining the current knowledge of the biophysical, physiological, and distribution properties required for a Nav1.7 inhibitor to produce analgesia.
Collapse
Affiliation(s)
- Alvaro Yogi
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON K1A 0R6, Canada; (U.B.); (M.J.M.); (M.M.)
| | | | | | | |
Collapse
|
8
|
Kim JI, Imaizumi K, Jurjuț O, Kelley KW, Wang D, Thete MV, Hudacova Z, Amin ND, Levy RJ, Scherrer G, Pașca SP. Human assembloid model of the ascending neural sensory pathway. Nature 2025:10.1038/s41586-025-08808-3. [PMID: 40205039 DOI: 10.1038/s41586-025-08808-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/19/2025] [Indexed: 04/11/2025]
Abstract
Somatosensory pathways convey crucial information about pain, touch, itch and body part movement from peripheral organs to the central nervous system1,2. Despite substantial needs to understand how these pathways assemble and to develop pain therapeutics, clinical translation remains challenging. This is probably related to species-specific features and the lack of in vitro models of the polysynaptic pathway. Here we established a human ascending somatosensory assembloid (hASA), a four-part assembloid generated from human pluripotent stem cells that integrates somatosensory, spinal, thalamic and cortical organoids to model the spinothalamic pathway. Transcriptomic profiling confirmed the presence of key cell types of this circuit. Rabies tracing and calcium imaging showed that sensory neurons connect to dorsal spinal cord neurons, which further connect to thalamic neurons. Following noxious chemical stimulation, calcium imaging of hASA demonstrated a coordinated response. In addition, extracellular recordings and imaging revealed synchronized activity across the assembloid. Notably, loss of the sodium channel NaV1.7, which causes pain insensitivity, disrupted synchrony across hASA. By contrast, a gain-of-function SCN9A variant associated with extreme pain disorder induced hypersynchrony. These experiments demonstrated the ability to functionally assemble the essential components of the human sensory pathway, which could accelerate our understanding of sensory circuits and facilitate therapeutic development.
Collapse
Affiliation(s)
- Ji-Il Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Kent Imaizumi
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Ovidiu Jurjuț
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Kevin W Kelley
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Dong Wang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Mayuri Vijay Thete
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Zuzana Hudacova
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Neal D Amin
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Rebecca J Levy
- Department of Neurology & Neurological Sciences, Division of Child Neurology, Stanford University, Stanford, CA, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA.
| |
Collapse
|
9
|
Chen R, Liu Y, Qian L, Yi M, Yin H, Wang S, Xiang B. Sodium channels as a new target for pain treatment. Front Pharmacol 2025; 16:1573254. [PMID: 40206072 PMCID: PMC11979154 DOI: 10.3389/fphar.2025.1573254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Voltage-gated sodium channels, especially the Nav1.7, Nav1.8, and Nav1.9 subtypes, play a crucial role in the transmission of pain signals. Nav1.7 is considered a threshold channel that regulates the generation of action potentials and is closely associated with various hereditary pain disorders. Nav1.8 primarily participates in inflammatory and neuropathic pain within the peripheral nervous system. Its characteristic of not involving the central nervous system makes it a potential target for non-addictive analgesics. Nav1.9 has shown significant involvement in cold pain sensing and small fiber neuropathy, although its mechanism of action is still under further investigation. Currently, despite promising results from preclinical studies, sodium channel inhibitors have not fully met expectations in clinical trials due to issues such as drug selectivity, dosing, and safety. The development of Nav1.7 and Nav1.8 inhibitors faces challenges such as drug intolerance, insufficient target occupancy, and off-target side effects. Future research may promote the development of non-opioid analgesics through combined inhibition strategies targeting multiple Nav subtypes, as well as improving drug selectivity and bioavailability. Overall, sodium channel inhibitors remain a key area of research in pain management, but their clinical application prospects still require further exploration.
Collapse
Affiliation(s)
- Rui Chen
- Department of Anesthesiology, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Yiran Liu
- Nursing Department, Cujin Community Health Service Center, Chengdu, China
| | - Liu Qian
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Mingliang Yi
- Department of Anesthesiology, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Hong Yin
- Department of Anesthesiology, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Shun Wang
- Department of Anesthesiology, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Bingbing Xiang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Köster PA, Leipold E, Tigerholm J, Maxion A, Namer B, Stiehl T, Lampert A. Nociceptor sodium channels shape subthreshold phase, upstroke, and shoulder of action potentials. J Gen Physiol 2025; 157:e202313526. [PMID: 39836077 PMCID: PMC11748974 DOI: 10.1085/jgp.202313526] [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: 12/13/2023] [Revised: 09/30/2024] [Accepted: 12/18/2024] [Indexed: 01/22/2025] Open
Abstract
Voltage-gated sodium channels (VGSCs) in the peripheral nervous system shape action potentials (APs) and thereby support the detection of sensory stimuli. Most of the nine mammalian VGSC subtypes are expressed in nociceptors, but predominantly, three are linked to several human pain syndromes: while Nav1.7 is suggested to be a (sub-)threshold channel, Nav1.8 is thought to support the fast AP upstroke. Nav1.9, as it produces large persistent currents, is attributed a role in determining the resting membrane potential. We characterized the gating of Nav1.1-Nav1.3 and Nav1.5-Nav1.9 in manual patch clamp with a focus on the AP subthreshold depolarization phase. Nav1.9 exhibited the most hyperpolarized activation, while its fast inactivation resembled the depolarized inactivation of Nav1.8. For some VGSCs (e.g., Nav1.1 and Nav1.2), a positive correlation between ramp current and window current was detected. Using a modified Hodgkin-Huxley model that accounts for the time needed for inactivation to occur, we used the acquired data to simulate two nociceptive nerve fiber types (an Aδ- and a mechano-insensitive C-nociceptor) containing VGSC conductances according to published human RNAseq data. Our simulations suggest that Nav1.9 is supporting both the AP upstroke and its shoulder. A reduced threshold for AP generation was induced by enhancing Nav1.7 conductivity or shifting its activation to more hyperpolarized potentials, as observed in Nav1.7-related pain disorders. Here, we provide a comprehensive, comparative functional characterization of VGSCs relevant in nociception and describe their gating with Hodgkin-Huxley-like models, which can serve as a tool to study their specific contributions to AP shape and sodium channel-related diseases.
Collapse
Affiliation(s)
- Phil Alexander Köster
- Institute for Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Enrico Leipold
- Department of Anesthesiology and Intensive Care and CBBM-Center of Brain, Behavior and Metabolism, University of Luebeck, Lübeck, Germany
| | - Jenny Tigerholm
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
- Joint Research Center for Computational Biomedicine (JRCC), Uniklinik RWTH Aachen University, Aachen, Germany
| | - Anna Maxion
- Institute for Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), Faculty of Medicine, Research Group Neurosciences, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Barbara Namer
- Institute for Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), Faculty of Medicine, Research Group Neurosciences, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Thomas Stiehl
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
- Joint Research Center for Computational Biomedicine (JRCC), Uniklinik RWTH Aachen University, Aachen, Germany
- Institute for Computational Biomedicine and Disease Modelling With Focus on Phase Transitions Between Phenotypes, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Angelika Lampert
- Institute for Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
| |
Collapse
|
11
|
Jahangiri Esfahani S, Ao X, Oveisi A, Diatchenko L. Rare variant association studies: Significance, methods, and applications in chronic pain studies. Osteoarthritis Cartilage 2025; 33:313-321. [PMID: 39725155 DOI: 10.1016/j.joca.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 11/27/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
Rare genetic variants, characterized by their low frequency in a population, have emerged as essential components in the study of complex disease genetics. The biology of rare variants underscores their significance, as they can exert profound effects on phenotypic variation and disease susceptibility. Recent advancements in sequencing technologies have yielded the availability of large-scale sequencing data such as the UK Biobank whole-exome sequencing (WES) cohort empowered researchers to conduct rare variant association studies (RVASs). This review paper discusses the significance of rare variants, available methodologies, and applications. We provide an overview of RVASs, emphasizing their relevance in unraveling the genetic architecture of complex diseases with special focus on chronic pain and Arthritis. Additionally, we discuss the strengths and limitations of various rare variant association testing methods, outlining a typical pipeline for conducting rare variant association. This pipeline encompasses crucial steps such as quality control of WES data, rare variant annotation, and association testing. It serves as a comprehensive guide for researchers in the field of chronic pain diseases interested in rare variant association studies in large-scale sequencing datasets like the UK Biobank WES cohort. Lastly, we discuss how the identified variants can be further investigated through detailed experimental studies in animal models to elucidate their functional impact and underlying mechanisms.
Collapse
Affiliation(s)
- Sahel Jahangiri Esfahani
- Faculty of Medicine and Health Sciences, Department of Human Genetics, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Xiang Ao
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Anahita Oveisi
- Department of Neuroscience, Faculty of Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Luda Diatchenko
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada.
| |
Collapse
|
12
|
Li Y, Lock A, Fedele L, Zebochin I, Sabate A, Siddle M, Cainarca S, Röderer P, Montag K, Tarroni P, Brüstle O, Shaw T, Taams L, Denk F. Modelling inflammation-induced peripheral sensitization in a dish-more complex than expected? Pain 2025:00006396-990000000-00838. [PMID: 40009350 DOI: 10.1097/j.pain.0000000000003512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/15/2024] [Indexed: 02/27/2025]
Abstract
ABSTRACT Peripheral sensitization of nociceptors is believed to be a key driver of chronic pain states. Here, we sought to study the effects of a modified version of inflammatory soup on the excitability of human stem cell-derived sensory neurons. For this, we used a preexisting and a novel stem cell line, modified to stably express the calcium sensor GCamP6f. Upon treatment with inflammatory soup, we observed no changes in neuronal transcription or functional responses upon calcium imaging and only a very minor increase in resting membrane potential (RMP) via whole cell patch clamping: control RMP (-71.31 ± 1.1 mV) vs inflammatory soup RMP (-67.74 ± 1.29 mV), uncorrected 2-tailed independent samples t test, P = 0.0383. Similarly, small changes were observed when treating mouse primary sensory neurons with inflammatory soup. A semi-systematic reexamination of past literature further indicated that observed effects of inflammatory mediators on dissociated sensory neuron cultures are generally small. We conclude that modelling inflammation-induced peripheral sensitization in vitro is nontrivial and will require careful selection of mediators and/or more complex, longitudinal multicellular setups. Especially in the latter, our novel GCamP6f-induced pluripotent stem cell line may be of value.
Collapse
Affiliation(s)
- Yuening Li
- Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Guy's Campus, King's College London, London, United Kingdom
- Centre for Inflammation Biology & Cancer Immunology, Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Amy Lock
- Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Guy's Campus, King's College London, London, United Kingdom
- Centre for Inflammation Biology & Cancer Immunology, Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Laura Fedele
- Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Guy's Campus, King's College London, London, United Kingdom
| | - Irene Zebochin
- Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Guy's Campus, King's College London, London, United Kingdom
| | - Alba Sabate
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Matthew Siddle
- Institute of Liver Studies, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | | | - Pascal Röderer
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
- LIFE&BRAIN GmbH, Cellomics Unit, Bonn, Germany
| | | | | | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
- LIFE&BRAIN GmbH, Cellomics Unit, Bonn, Germany
| | - Tanya Shaw
- Centre for Inflammation Biology & Cancer Immunology, Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Leonie Taams
- Centre for Inflammation Biology & Cancer Immunology, Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Franziska Denk
- Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Guy's Campus, King's College London, London, United Kingdom
| |
Collapse
|
13
|
Furia A, Liguori R, Donadio V. Small-Fiber Neuropathy: An Etiology-Oriented Review. Brain Sci 2025; 15:158. [PMID: 40002491 PMCID: PMC11853085 DOI: 10.3390/brainsci15020158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Small-fiber neuropathy (SFN), affecting Aδ or C nerve fibers, is characterized by alterations of pain and temperature sensation, as well as autonomic dysfunction. Its diagnosis may still remain challenging as methods specifically assessing small nerve fibers are not always readily available, and standard techniques for large-fiber neuropathies, such as electroneuromyography, yield negative results. Still, skin biopsy for epidermal innervation and quantitative sensory testing allow for diagnosis in the presence of a congruent clinical picture. OBJECTIVES Many different etiologies may underlie small-fiber neuropathy, of which metabolic (diabetes mellitus/impaired glucose tolerance) and idiopathic remain prevalent. The aim of this narrative review is to provide a general picture of SFN while focusing on the different etiologies described in the literature in order to raise awareness of the variegated set of different causes of SFN and promote adequate diagnostic investigation. METHODS The term "Small-Fiber Neuropathy" was searched on the PubMed database with its different recognized etiologies: the abstracts of the articles were reviewed and described in the article if relevant for a total of 40 studies. RESULTS Many different disorders have been associated with SFN, even though often in the form of case reports or small case series. CONCLUSIONS Idiopathic forms of SFN remain the most prevalent in the literature, but association with different disorders (e.g., infectious, autoimmune) should prompt investigation for SFN in the presence of a congruent clinical picture (e.g., pain with neuropathic features).
Collapse
Affiliation(s)
- Alessandro Furia
- Dipartimento di Scienze Biomediche e Neuromotorie, University of Bologna, 40138 Bologna, Italy
| | - Rocco Liguori
- Dipartimento di Scienze Biomediche e Neuromotorie, University of Bologna, 40138 Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche Di Bologna, UOC Clinica Neurologica, 40139 Bologna, Italy
| | - Vincenzo Donadio
- Dipartimento di Scienze Biomediche e Neuromotorie, University of Bologna, 40138 Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche Di Bologna, UOC Clinica Neurologica, 40139 Bologna, Italy
| |
Collapse
|
14
|
Su M, Ouyang X, Zhou P, Dong L, Shao L, Wang K, Liu Y. Inhibition of TTX-S Na + currents by a novel blocker QLS-278 for antinociception. J Pharmacol Exp Ther 2025; 392:100030. [PMID: 40023600 DOI: 10.1124/jpet.124.002273] [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: 04/15/2024] [Revised: 06/15/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024] Open
Abstract
Genetic loss-of-function mutations of the NaV1.7 channel, abundantly expressed in peripheral nociceptive neurons, cause congenital insensitivity to pain in humans, indicating that selective inhibition of the channel may lead to potential therapy for pain disorders. In this study, we investigated a novel compound, 5-chloro-N-(cyclopropylsulfonyl)-2-fluoro-4-(2-(8-(furan-2-ylmethyl)-8-azaspiro [4.5] decan-2-yl) ethoxy) benzamide (QLS-278) that inhibits NaV1.7 channels and exhibits antinociceptive activity. Compound QLS-278 exhibits inactivation- and concentration-dependent inhibition of macroscopic currents of NaV1.7 channels stably expressed in HEK293 cells with an IC50 of 1.2 ± 0.2 μM. QLS-278 causes a hyperpolarization shift of the channel inactivation and delays recovery from inactivation, without any noticeable effect on voltage-dependent activation. In mouse dorsal root ganglion neurons, QLS-278 suppresses native tetrodotoxin-sensitive NaV currents and also reduces neuronal firings. Moreover, QLS-278 dose-dependently relieves neuropathic pain induced by spared nerve injury and inflammatory pain induced by formalin without significantly altering spontaneous locomotor activity in mice. Therefore, our identification of the novel compound QLS-278 may hold developmental potential in chronic pain treatment. SIGNIFICANCE STATEMENT: QLS-278, a novel voltage-gated sodium NaV1.7 channel blocker, inhibits native tetrodotoxin-sensitive Na+ current and reduces action potential firings in dorsal root ganglion sensory neurons. QLS-278 also exhibits antinociceptive activity in mouse models of pain, demonstrating the potential for the development of a chronic pain treatment.
Collapse
Affiliation(s)
- Min Su
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
| | - Xiangshuo Ouyang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Ping Zhou
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
| | - Liying Dong
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
| | - Liming Shao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China.
| | - KeWei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China; Institute of Innovative Drugs, Qingdao University, Qingdao, China
| | - Yani Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China; Institute of Innovative Drugs, Qingdao University, Qingdao, China.
| |
Collapse
|
15
|
Chamessian A, Payne M, Gordon I, Zhou M, Gereau R. Small molecule-mediated targeted protein degradation of voltage-gated sodium channels involved in pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.21.634079. [PMID: 39896637 PMCID: PMC11785090 DOI: 10.1101/2025.01.21.634079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The voltage-gated sodium channels (VGSC) NaV1.8 and NaV1.7 (NaVs) have emerged as promising and high-value targets for the development of novel, non-addictive analgesics to combat the chronic pain epidemic. In recent years, many small molecule inhibitors against these channels have been developed. The recent successful clinical trial of VX-548, a NaV1.8-selective inhibitor, has spurred much interest in expanding the arsenal of subtype-selective voltage-gated sodium channel therapeutics. Toward that end, we sought to determine whether NaVs are amenable to targeted protein degradation with small molecule degraders, namely proteolysis-targeting chimeras (PROTACs) and molecular glues. Here, we report that degron-tagged NaVs are potently and rapidly degraded by small molecule degraders harnessing the E3 ubiquitin ligases cereblon (CRBN) and Von Hippel Lindau (VHL). Using LC/MS analysis, we demonstrate that PROTAC-mediated proximity between NaV1.8 and CRBN results in ubiquitination on the 2nd intracellular loop, pointing toward a potential mechanism of action and demonstrating the ability of CRBN to recognize a VGSC as a neosubstrate. Our foundational findings are an important first step toward realizing the immense potential of NaV-targeting degrader analgesics to combat chronic pain.
Collapse
|
16
|
Shehab S, Hamad MIK, Emerald BS. A novel approach to completely alleviate peripheral neuropathic pain in human patients: insights from preclinical data. Front Neuroanat 2025; 18:1523095. [PMID: 39839257 PMCID: PMC11747518 DOI: 10.3389/fnana.2024.1523095] [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: 11/05/2024] [Accepted: 12/23/2024] [Indexed: 01/23/2025] Open
Abstract
Neuropathic pain is a pervasive health concern worldwide, posing significant challenges to both clinicians and neuroscientists. While acute pain serves as a warning signal for potential tissue damage, neuropathic pain represents a chronic pathological condition resulting from injury or disease affecting sensory pathways of the nervous system. Neuropathic pain is characterized by long-lasting ipsilateral hyperalgesia (increased sensitivity to pain), allodynia (pain sensation in response to stimuli that are not normally painful), and spontaneous unprovoked pain. Current treatments for neuropathic pain are generally inadequate, and prevention remains elusive. In this review, we provide an overview of current treatments, their limitations, and a discussion on the potential of capsaicin and its analog, resiniferatoxin (RTX), for complete alleviation of nerve injury-induced neuropathic pain. In an animal model of neuropathic pain where the fifth lumbar (L5) spinal nerve is unilaterally ligated and cut, resulting in ipsilateral hyperalgesia, allodynia, and spontaneous pain akin to human neuropathic pain. The application of capsaicin or RTX to the adjacent uninjured L3 and L4 nerves completely alleviated and prevented mechanical and thermal hyperalgesia following the L5 nerve injury. The effects of this treatment were specific to unmyelinated fibers (responsible for pain sensation), while thick myelinated nerve fibers (responsible for touch and mechanoreceptor sensations) remained intact. Here, we propose to translate these promising preclinical results into effective therapeutic interventions in humans by direct application of capsaicin or RTX to adjacent uninjured nerves in patients who suffer from neuropathic pain due to peripheral nerve injury, following surgical interventions, diabetic neuropathy, trauma, vertebral disc herniation, nerve entrapment, ischemia, postherpetic lesion, and spinal cord injury.
Collapse
Affiliation(s)
- Safa Shehab
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | | | | |
Collapse
|
17
|
Zebochin I, Denk F, Nochi Z. Modeling neuropathic pain in a dish. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 179:233-278. [PMID: 39580214 DOI: 10.1016/bs.irn.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
The study of pain mechanisms has advanced significantly with the development of innovative in vitro models. This chapter explores those already used in or potentially useful for neuropathic pain research, emphasizing the complementary roles of animal and human cellular models to enhance translational success. Traditional animal models have provided foundational insights into the neurobiology of pain and remain invaluable for understanding complex pain pathways. However, integrating human cellular models addresses the need for better replication of human nociceptors. The chapter details methodologies for culturing rodent and human primary sensory neurons, including isolation and culture techniques, advantages, and limitations. It highlights the application of these models in neuropathic pain research, such as identifying pain-associated receptors and ion channels. Recent advancements in using induced pluripotent stem cell (iPSC)-derived sensory neurons are also discussed. Finally, the chapter explores advanced in vitro models, including 2D co-cultures and 3D organoids, and their implications for studying neuropathic pain. These models offer significant advantages for drug screening and ethical research practices, providing a more accurate representation of human pain pathways and paving the way for innovative therapeutic strategies. Despite challenges such as limited access to viable human tissue and variability between samples, these in vitro models, alongside traditional animal models, are indispensable for advancing our understanding of neuropathic pain and developing effective treatments.
Collapse
Affiliation(s)
- Irene Zebochin
- Wolfson Sensory Pain and Regeneration Centre (SPaRC), King's College London
| | - Franziska Denk
- Wolfson Sensory Pain and Regeneration Centre (SPaRC), King's College London
| | - Zahra Nochi
- Danish Pain Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
18
|
Zhao N, Bennett DL, Baskozos G, Barry AM. Predicting 'pain genes': multi-modal data integration using probabilistic classifiers and interaction networks. BIOINFORMATICS ADVANCES 2024; 4:vbae156. [PMID: 39526039 PMCID: PMC11549022 DOI: 10.1093/bioadv/vbae156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/16/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Motivation Accurate identification of pain-related genes remains challenging due to the complex nature of pain pathophysiology and the subjective nature of pain reporting in humans. Here, we use machine learning to identify possible 'pain genes'. Labelling was based on a gold-standard list with validated involvement across pain conditions, and was trained on a selection of -omics, protein-protein interaction network features, and biological function readouts for each gene. Results The top-performing model was selected to predict a 'pain score' per gene. The top-ranked genes were then validated against pain-related human SNPs. Functional analysis revealed JAK2/STAT3 signal, ErbB, and Rap1 signalling pathways as promising targets for further exploration, while network topological features contribute significantly to the identification of 'pain' genes. As such, a network based on top-ranked genes was constructed to reveal previously uncharacterized pain-related genes. Together, these novel insights into pain pathogenesis can indicate promising directions for future experimental research. Availability and implementation These analyses can be further explored using the linked open-source database at https://livedataoxford.shinyapps.io/drg-directory/, which is accompanied by a freely accessible code template and user guide for wider adoption across disciplines.
Collapse
Affiliation(s)
- Na Zhao
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - David L Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Georgios Baskozos
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Allison M Barry
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, United Kingdom
| |
Collapse
|
19
|
Sánchez-Carranza O, Chakrabarti S, Kühnemund J, Schwaller F, Bégay V, García-Contreras JA, Wang L, Lewin GR. Piezo2 voltage-block regulates mechanical pain sensitivity. Brain 2024; 147:3487-3500. [PMID: 38984717 PMCID: PMC11449130 DOI: 10.1093/brain/awae227] [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: 02/12/2024] [Revised: 05/19/2024] [Accepted: 06/13/2024] [Indexed: 07/11/2024] Open
Abstract
PIEZO2 is a trimeric mechanically-gated ion channel expressed by most sensory neurons in the dorsal root ganglia. Mechanosensitive PIEZO2 channels are also genetically required for normal touch sensation in both mice and humans. We previously showed that PIEZO2 channels are also strongly modulated by membrane voltage. Specifically, it is only at very positive voltages that all channels are available for opening by mechanical force. Conversely, most PIEZO2 channels are blocked at normal negative resting membrane potentials. The physiological function of this unusual biophysical property of PIEZO2 channels, however, remained unknown. We characterized the biophysical properties of three PIEZO2 ion channel mutations at an evolutionarily conserved arginine (R2756). Using genome engineering in mice we generated Piezo2R2756H/R2756H and Piezo2R2756K/R2756K knock-in mice to characterize the physiological consequences of altering PIEZO2 voltage sensitivity in vivo. We measured endogenous mechanosensitive currents in sensory neurons isolated from the dorsal root ganglia and characterized mechanoreceptor and nociceptor function using electrophysiology. Mice were also assessed behaviourally and morphologically. Mutations at the conserved Arginine (R2756) dramatically changed the biophysical properties of the channel relieving voltage block and lowering mechanical thresholds for channel activation. Piezo2R2756H/R2756H and Piezo2R2756K/R2756K knock-in mice that were homozygous for gain-of-function mutations were viable and were tested for sensory changes. Surprisingly, mechanosensitive currents in nociceptors, neurons that detect noxious mechanical stimuli, were substantially sensitized in Piezo2 knock-in mice, but mechanosensitive currents in most mechanoreceptors that underlie touch sensation were only mildly affected by the same mutations. Single-unit electrophysiological recordings from sensory neurons innervating the glabrous skin revealed that rapidly-adapting mechanoreceptors that innervate Meissner's corpuscles exhibited slightly decreased mechanical thresholds in Piezo2 knock-in mice. Consistent with measurements of mechanically activated currents in isolated sensory neurons essentially all cutaneous nociceptors, both fast conducting Aδ-mechanonociceptors and unmyelinated C-fibre nociceptors were substantially more sensitive to mechanical stimuli and indeed acquired receptor properties similar to ultrasensitive touch receptors in Piezo2 knock-in mice. Mechanical stimuli also induced enhanced ongoing activity in cutaneous nociceptors in Piezo2 knock-in mice and hyper-sensitive PIEZO2 channels were sufficient alone to drive ongoing activity, even in isolated nociceptive neurons. Consistently, Piezo2 knock-in mice showed substantial behavioural hypersensitivity to noxious mechanical stimuli. Our data indicate that ongoing activity and sensitization of nociceptors, phenomena commonly found in human chronic pain syndromes, can be driven by relieving the voltage-block of PIEZO2 ion channels. Indeed, membrane depolarization caused by multiple noxious stimuli may sensitize nociceptors by relieving voltage-block of PIEZO2 channels.
Collapse
Affiliation(s)
- Oscar Sánchez-Carranza
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 10409, Germany
| | - Sampurna Chakrabarti
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 10409, Germany
| | - Johannes Kühnemund
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 10409, Germany
| | - Fred Schwaller
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 10409, Germany
| | - Valérie Bégay
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 10409, Germany
| | - Jonathan Alexis García-Contreras
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 10409, Germany
| | - Lin Wang
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 10409, Germany
| | - Gary R Lewin
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 10409, Germany
- Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- German Center for Mental Health (DZPG), partner site Berlin, 10117 Berlin, Germany
| |
Collapse
|
20
|
Albani S, Eswaran VSB, Piergentili A, de Souza PCT, Lampert A, Rossetti G. Depletion of membrane cholesterol modifies structure, dynamic and activation of Na v1.7. Int J Biol Macromol 2024; 278:134219. [PMID: 39097041 DOI: 10.1016/j.ijbiomac.2024.134219] [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: 06/13/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024]
Abstract
Cholesterol is a major component of plasma membranes and plays a significant role in actively regulating the functioning of several membrane proteins in humans. In this study, we focus on the role of cholesterol depletion on the voltage-gated sodium channel Nav1.7, which is primarily expressed in the peripheral sensory neurons and linked to various chronic inherited pain syndromes. Coarse-grained molecular dynamics simulations revealed key dynamic changes of Nav1.7 upon membrane cholesterol depletion: A loss of rigidity in the structural motifs linked to activation and fast-inactivation is observed, suggesting an easier transition of the channel between different gating states. In-vitro whole-cell patch clamp experiments on HEK293t cells expressing Nav1.7 validated these predictions at the functional level: Hyperpolarizing shifts in the voltage-dependence of activation and fast-inactivation were observed along with an acceleration of the time to peak and onset kinetics of fast inactivation. These results underline the critical role of membrane composition, and of cholesterol in particular, in influencing Nav1.7 gating characteristics. Furthermore, our results also point to cholesterol-driven changes of the geometry of drug-binding regions, hinting to a key role of the membrane environment in the regulation of drug effects.
Collapse
Affiliation(s)
- Simone Albani
- Institute of Neuroscience and Medicine (INM-9)/Institute for Advanced Simulation (IAS-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52425 Jülich, Germany; Faculty of Biology, RWTH Aachen University, Aachen, Germany
| | | | - Alessia Piergentili
- Institute of Neuroscience and Medicine (INM-9)/Institute for Advanced Simulation (IAS-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52425 Jülich, Germany; Faculty of Biology, RWTH Aachen University, Aachen, Germany; Department of Neurology, University Hospital Aachen, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Paulo Cesar Telles de Souza
- Laboratoire de Biologie et Modélisation de la Cellule, CNRS, UMR 5239, Inserm, U1293, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, 69364 Lyon, France; Centre Blaise Pascal de Simulation et de Modélisation Numérique, Ecole Normale, Supérieure de Lyon, 46 All'ee d'Italie, 69364 Lyon, France
| | - Angelika Lampert
- Institute of Neurophysiology, Uniklinik RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | - Giulia Rossetti
- Institute of Neuroscience and Medicine (INM-9)/Institute for Advanced Simulation (IAS-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52425 Jülich, Germany; Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52425 Jülich, Germany; Department of Neurology, University Hospital Aachen, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany.
| |
Collapse
|
21
|
Mulcahy JV, Beckley JT, Klas SD, Odink DA, Delwig A, Pajouhesh H, Monteleone D, Zhou X, Du Bois J, Yeomans DC, Luu G, Hunter JC. ST-2560, a selective inhibitor of the Na V1.7 sodium channel, affects nocifensive and cardiovascular reflexes in non-human primates. Br J Pharmacol 2024; 181:3160-3171. [PMID: 38715413 DOI: 10.1111/bph.16398] [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: 10/27/2023] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 08/03/2024] Open
Abstract
BACKGROUND AND PURPOSE The voltage-gated sodium channel isoform NaV1.7 is a high-interest target for the development of non-opioid analgesics due to its preferential expression in pain-sensing neurons. NaV1.7 is also expressed in autonomic neurons, yet its contribution to involuntary visceral reflexes has received limited attention. The small molecule inhibitor ST-2560 was advanced into pain behaviour and cardiovascular models to understand the pharmacodynamic effects of selective inhibition of NaV1.7. EXPERIMENTAL APPROACH Potency of ST-2560 at NaV1.7 and off-target ion channels was evaluated by whole-cell patch-clamp electrophysiology. Effects on nocifensive reflexes were assessed in non-human primate (NHP) behavioural models, employing the chemical capsaicin and mechanical stimuli. Cardiovascular parameters were monitored continuously in freely-moving, telemetered NHPs following administration of vehicle and ST-2560. KEY RESULTS ST-2560 is a potent inhibitor (IC50 = 39 nM) of NaV1.7 in primates with ≥1000-fold selectivity over other isoforms of the human NaV1.x family. Following systemic administration, ST-2560 (0.1-0.3 mg·kg-1, s.c.) suppressed noxious mechanical- and chemical-evoked reflexes at free plasma concentrations threefold to fivefold above NaV1.7 IC50. ST-2560 (0.1-1.0 mg·kg-1, s.c.) also produced changes in haemodynamic parameters, most notably a 10- to 20-mmHg reduction in systolic and diastolic arterial blood pressure, at similar exposures. CONCLUSIONS AND IMPLICATIONS Acute pharmacological inhibition of NaV1.7 is antinociceptive, but also has the potential to impact the cardiovascular system. Further work is merited to understand the role of NaV1.7 in autonomic ganglia involved in the control of heart rate and blood pressure, and the effect of selective NaV1.7 inhibition on cardiovascular function.
Collapse
Affiliation(s)
- John V Mulcahy
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| | - Jacob T Beckley
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| | - Sheri D Klas
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| | - Debra A Odink
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| | - Anton Delwig
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| | - Hassan Pajouhesh
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| | | | - Xiang Zhou
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| | - Justin Du Bois
- Department of Chemistry, Stanford University, Stanford, California, USA
| | - David C Yeomans
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - George Luu
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| | - John C Hunter
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| |
Collapse
|
22
|
LeBlang CJ, Pazyra-Murphy MF, Silagi ES, Dasgupta S, Tsolias M, Miller T, Petrova V, Zhen S, Jovanovic V, Castellano D, Gerrish K, Ormanoglu P, Tristan C, Singeç I, Woolf CJ, Tasdemir-Yilmaz O, Segal RA. Satellite glial contact enhances differentiation and maturation of human iPSC-derived sensory neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604966. [PMID: 39211268 PMCID: PMC11361066 DOI: 10.1101/2024.07.24.604966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Sensory neurons generated from induced pluripotent stem cells (iSNs) are used to model human peripheral neuropathies, however current differentiation protocols produce sensory neurons with an embryonic phenotype. Peripheral glial cells contact sensory neurons early in development and contribute to formation of the canonical pseudounipolar morphology, but these signals are not encompassed in current iSN differentiation protocols. Here, we show that terminal differentiation of iSNs in co-culture with rodent Dorsal Root Ganglion satellite glia (rSG) advances their differentiation and maturation. Co-cultured iSNs develop a pseudounipolar morphology through contact with rSGs. This transition depends on semaphorin-plexin guidance cues and on glial gap junction signaling. In addition to morphological changes, iSNs terminally differentiated in co-culture exhibit enhanced spontaneous action potential firing, more mature gene expression, and increased susceptibility to paclitaxel induced axonal degeneration. Thus, iSNs differentiated in coculture with rSGs provide a better model for investigating human peripheral neuropathies.
Collapse
|
23
|
Ogawa T, Yamada S, Fukushi S, Imai Y, Kawada J, Ikeda K, Ohka S, Kaneda S. Formation and Long-Term Culture of hiPSC-Derived Sensory Nerve Organoids Using Microfluidic Devices. Bioengineering (Basel) 2024; 11:794. [PMID: 39199753 PMCID: PMC11352057 DOI: 10.3390/bioengineering11080794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/20/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024] Open
Abstract
Although methods for generating human induced pluripotent stem cell (hiPSC)-derived motor nerve organoids are well established, those for sensory nerve organoids are not. Therefore, this study investigated the feasibility of generating sensory nerve organoids composed of hiPSC-derived sensory neurons using a microfluidic approach. Notably, sensory neuronal axons from neurospheres containing 100,000 cells were unidirectionally elongated to form sensory nerve organoids over 6 mm long axon bundles within 14 days using I-shaped microchannels in microfluidic devices composed of polydimethylsiloxane (PDMS) chips and glass substrates. Additionally, the organoids were successfully cultured for more than 60 days by exchanging the culture medium. The percentage of nuclei located in the distal part of the axon bundles (the region 3-6 mm from the entrance of the microchannel) compared to the total number of cells in the neurosphere was 0.005% for live cells and 0.008% for dead cells. Molecular characterization confirmed the presence of the sensory neuron marker ISL LIM homeobox 1 (ISL1) and the capsaicin receptor transient receptor potential vanilloid 1 (TRPV1). Moreover, capsaicin stimulation activated TRPV1 in organoids, as evidenced by significant calcium ion influx. Conclusively, this study demonstrated the feasibility of long-term organoid culture and the potential applications of sensory nerve organoids in bioengineered nociceptive sensors.
Collapse
Affiliation(s)
- Takuma Ogawa
- Mechanical Engineering Program, Graduate School of Engineering, Kogakuin University, 1-24-2 Nishishinjuku, Shinjuku-ku, Tokyo 163-8677, Japan
| | - Souichi Yamada
- Department of Virology I, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Shuetsu Fukushi
- Department of Virology I, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Yuya Imai
- Mechanical Engineering Program, Graduate School of Engineering, Kogakuin University, 1-24-2 Nishishinjuku, Shinjuku-ku, Tokyo 163-8677, Japan
| | - Jiro Kawada
- Jiksak Bioengineering, Inc., 3-25-16 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Kanagawa, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan (S.O.)
- Department of Neuropsychopharmacology, National Institute of Mental Health, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi-cho, Kodaira, Tokyo 187-8553, Japan
| | - Seii Ohka
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan (S.O.)
| | - Shohei Kaneda
- Mechanical Engineering Program, Graduate School of Engineering, Kogakuin University, 1-24-2 Nishishinjuku, Shinjuku-ku, Tokyo 163-8677, Japan
| |
Collapse
|
24
|
Le Franc A, Da Silva A, Lepetre-Mouelhi S. Nanomedicine and voltage-gated sodium channel blockers in pain management: a game changer or a lost cause? Drug Deliv Transl Res 2024; 14:2112-2145. [PMID: 38861139 DOI: 10.1007/s13346-024-01615-9] [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] [Accepted: 04/25/2024] [Indexed: 06/12/2024]
Abstract
Pain, a complex and debilitating condition affecting millions globally, is a significant concern, especially in the context of post-operative recovery. This comprehensive review explores the complexity of pain and its global impact, emphasizing the modulation of voltage-gated sodium channels (VGSC or NaV channels) as a promising avenue for pain management with the aim of reducing reliance on opioids. The article delves into the role of specific NaV isoforms, particularly NaV 1.7, NaV 1.8, and NaV 1.9, in pain process and discusses the development of sodium channel blockers to target these isoforms precisely. Traditional local anesthetics and selective NaV isoform inhibitors, despite showing varying efficacy in pain management, face challenges in systemic distribution and potential side effects. The review highlights the potential of nanomedicine in improving the delivery of local anesthetics, toxins and selective NaV isoform inhibitors for a targeted and sustained release at the site of pain. This innovative strategy seeks to improve drug bioavailability, minimize systemic exposure, and optimize therapeutic outcomes, holding significant promise for secure pain management and enhancing the quality of life for individuals recovering from surgical procedures or suffering from chronic pain.
Collapse
Affiliation(s)
- Adélaïde Le Franc
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | - Alexandre Da Silva
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | | |
Collapse
|
25
|
Zurek NA, Ehsanian R, Goins AE, Adams IM, Petersen T, Goyal S, Shilling M, Westlund KN, Alles SRA. Electrophysiological Analyses of Human Dorsal Root Ganglia and Human Induced Pluripotent Stem Cell-derived Sensory Neurons From Male and Female Donors. THE JOURNAL OF PAIN 2024; 25:104451. [PMID: 38154622 PMCID: PMC11128351 DOI: 10.1016/j.jpain.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023]
Abstract
Human induced pluripotent stem cell-derived sensory neurons (hiPSC-SNs) and human dorsal root ganglia neurons (hDRG-N) are popular tools in the field of pain research; however, few groups make use of both approaches. For screening and analgesic validation purposes, important characterizations can be determined of the similarities and differences between hDRG-N and hiPSC-SNs. This study focuses specifically on the electrophysiology properties of hDRG-N in comparison to hiPSC-SNs. We also compared hDRG-N and hiPSC-SNs from both male and female donors to evaluate potential sex differences. We recorded neuronal size, rheobase, resting membrane potential, input resistance, and action potential waveform properties from 83 hiPSCs-SNs (2 donors) and 108 hDRG-N neurons (8 donors). We observed several statistically significant electrophysiological differences between hDRG-N and hiPSC-SNs, such as size, rheobase, input resistance, and several action potential waveform properties. Correlation analysis also revealed many properties that were positively or negatively correlated, some of which were differentially correlated between hDRG-N and hiPSC-SNs. This study shows several differences between hDRG-N and hiPSC-SNs and allows a better understanding of the advantages and disadvantages of both for use in pain research. We hope this study will be a valuable resource for pain researchers considering the use of these human in vitro systems for mechanistic studies and/or drug development projects. PERSPECTIVE: hiPSC-SNs and hDRG-N are popular tools in the field of pain research. This study allows for a better functional understanding of the pros and cons of both tools.
Collapse
Affiliation(s)
- Nesia A Zurek
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Reza Ehsanian
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Aleyah E Goins
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Ian M Adams
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Timothy Petersen
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Sachin Goyal
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Mark Shilling
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Karin N Westlund
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Sascha R A Alles
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico.
| |
Collapse
|
26
|
Van Lent J, Prior R, Pérez Siles G, Cutrupi AN, Kennerson ML, Vangansewinkel T, Wolfs E, Mukherjee-Clavin B, Nevin Z, Judge L, Conklin B, Tyynismaa H, Clark AJ, Bennett DL, Van Den Bosch L, Saporta M, Timmerman V. Advances and challenges in modeling inherited peripheral neuropathies using iPSCs. Exp Mol Med 2024; 56:1348-1364. [PMID: 38825644 PMCID: PMC11263568 DOI: 10.1038/s12276-024-01250-x] [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: 11/10/2023] [Revised: 02/21/2024] [Accepted: 03/18/2024] [Indexed: 06/04/2024] Open
Abstract
Inherited peripheral neuropathies (IPNs) are a group of diseases associated with mutations in various genes with fundamental roles in the development and function of peripheral nerves. Over the past 10 years, significant advances in identifying molecular disease mechanisms underlying axonal and myelin degeneration, acquired from cellular biology studies and transgenic fly and rodent models, have facilitated the development of promising treatment strategies. However, no clinical treatment has emerged to date. This lack of treatment highlights the urgent need for more biologically and clinically relevant models recapitulating IPNs. For both neurodevelopmental and neurodegenerative diseases, patient-specific induced pluripotent stem cells (iPSCs) are a particularly powerful platform for disease modeling and preclinical studies. In this review, we provide an update on different in vitro human cellular IPN models, including traditional two-dimensional monoculture iPSC derivatives, and recent advances in more complex human iPSC-based systems using microfluidic chips, organoids, and assembloids.
Collapse
Grants
- R01 NS119678 NINDS NIH HHS
- U01 ES032673 NIEHS NIH HHS
- Wellcome Trust
- R01 AG072052 NIA NIH HHS
- DOC-PRO4 Universiteit Antwerpen (University of Antwerp)
- RF1 AG072052 NIA NIH HHS
- This work was supported in part by the University of Antwerp (DOC-PRO4 PhD fellowship to J.V.L. and TOP-BOF research grant no. 38694 to V.T.), the Association Française contre les Myopathies (AFM research grant no. 24063 to V.T.), Association Belge contre les Maladies Neuromusculaires (ABMM research grant no. 1 to J.V.L and V.T), the interuniversity research fund (iBOF project to. L.V.D.B, E.W. and V.T.). V.T. is part of the μNEURO Research Centre of Excellence of the University of Antwerp and is an active member of the European Network for Stem Cell Core Facilities (CorEUStem, COST Action CA20140). Work in the M.L.K group was supported by the NHMRC Ideas Grant (APP1186867), CMT Australia Grant awarded to M.L.K and G.P.-S and the Australian Medical Research Future Fund (MRFF) Genomics Health Futures Mission Grant 2007681. B.M.C. is supported by the American Academy of Neurology and the Passano Foundation. L.M.J. and B.R.C. are supported by the Charcot-Marie-Tooth Association, NINDS R01 NS119678, NIEHS U01 ES032673. H.T. is supported by Academy of Finland Centre of Excellence in Stem Cell Metabolism and Sigrid Juselius Foundation. Work in the D.L.B. group is supported by a Wellcome Investigator Grant (223149/Z/21/Z), the MRC (MR/T020113/1), and with funding from the MRC and Versus Arthritis to the PAINSTORM consortium as part of the Advanced Pain Discovery Platform (MR/W002388/1).
- Australian Medical Association (Australian Medical Association Limited)
- Universiteit Hasselt (UHasselt)
- American Academy of Neurology (AAN)
- Gladstone Institutes (J. David Gladstone Institutes)
- Academy of Finland (Suomen Akatemia)
- Academy of Medical Royal Colleges (AoMRC)
- Wellcome Trust (Wellcome)
- Oxford University Hospitals NHS Trust (Oxford University Hospitals National Health Service Trust)
- KU Leuven (Katholieke Universiteit Leuven)
- Vlaams Instituut voor Biotechnologie (Flanders Institute for Biotechnology)
- Miami University | Leonard M. Miller School of Medicine (Miller School of Medicine)
Collapse
Affiliation(s)
- Jonas Van Lent
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, 2610, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, 2610, Antwerp, Belgium
- Institute of Oncology Research (IOR), BIOS+, 6500, Bellinzona, Switzerland
- Università della Svizzera Italiana, 6900, Lugano, Switzerland
| | - Robert Prior
- Universitätsklinikum Bonn (UKB), University of Bonn, Bonn, Germany
| | - Gonzalo Pérez Siles
- Northcott Neuroscience Laboratory, ANZAC Research Institute Sydney Local Health District and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Anthony N Cutrupi
- Northcott Neuroscience Laboratory, ANZAC Research Institute Sydney Local Health District and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Marina L Kennerson
- Northcott Neuroscience Laboratory, ANZAC Research Institute Sydney Local Health District and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Molecular Medicine Laboratory, Concord Hospital, Sydney, NSW, Australia
| | - Tim Vangansewinkel
- UHasselt - Hasselt University, BIOMED, Laboratory for Functional Imaging and Research on Stem Cells (FIERCE Lab), Agoralaan, 3590, Diepenbeek, Belgium
- VIB-Center for Brain and Disease Research, Laboratory of Neurobiology, 3000, Leuven, Belgium
| | - Esther Wolfs
- UHasselt - Hasselt University, BIOMED, Laboratory for Functional Imaging and Research on Stem Cells (FIERCE Lab), Agoralaan, 3590, Diepenbeek, Belgium
| | | | | | - Luke Judge
- Gladstone Institutes, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce Conklin
- Gladstone Institutes, San Francisco, CA, USA
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Henna Tyynismaa
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00290, Helsinki, Finland
| | - Alex J Clark
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - David L Bennett
- Nuffield Department of Clinical Neuroscience, Oxford University, Oxford, UK
| | - Ludo Van Den Bosch
- VIB-Center for Brain and Disease Research, Laboratory of Neurobiology, 3000, Leuven, Belgium
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute, KU Leuven-University of Leuven, 3000, Leuven, Belgium
| | - Mario Saporta
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Vincent Timmerman
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, 2610, Antwerp, Belgium.
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, 2610, Antwerp, Belgium.
| |
Collapse
|
27
|
Allison RL, Welby E, Ehlers V, Burand A, Isaeva O, Nieves Torres D, Highland J, Brandow AM, Stucky CL, Ebert AD. Sickle cell disease iPSC-derived sensory neurons exhibit increased excitability and sensitization to patient plasma. Blood 2024; 143:2037-2052. [PMID: 38427938 PMCID: PMC11143522 DOI: 10.1182/blood.2023022591] [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: 09/18/2023] [Revised: 01/25/2024] [Accepted: 02/14/2024] [Indexed: 03/03/2024] Open
Abstract
ABSTRACT Individuals living with sickle cell disease (SCD) experience severe recurrent acute and chronic pain. Challenges to gaining mechanistic insight into pathogenic SCD pain processes include differential gene expression and function of sensory neurons between humans and mice with SCD, and extremely limited availability of neuronal tissues from patients with SCD. Here, we used induced pluripotent stem cells (iPSCs), derived from patients with SCD, differentiated into sensory neurons (SCD iSNs) to begin to overcome these challenges. We characterize key gene expression and function of SCD iSNs to establish a model to investigate intrinsic and extrinsic factors that may contribute to SCD pain. Despite similarities in receptor gene expression, SCD iSNs show pronounced excitability using patch clamp electrophysiology. Furthermore, we find that plasma taken from patients with SCD during acute pain associated with a vaso-occlusive event increases the calcium responses to the nociceptive stimulus capsaicin in SCD iSNs compared with those treated with paired plasma from patients with SCD at steady state baseline or healthy control plasma samples. We identified high levels of the polyamine spermine in baseline and acute pain states of plasma from patients with SCD, which sensitizes SCD iSNs to subthreshold concentrations of capsaicin. Together, these data identify potential intrinsic mechanisms within SCD iSNs that may extend beyond a blood-based pathology.
Collapse
Affiliation(s)
- Reilly L. Allison
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Emily Welby
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Vanessa Ehlers
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Anthony Burand
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Olena Isaeva
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Damaris Nieves Torres
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI
| | - Janelle Highland
- Department of Pediatrics, Section of Hematology/Oncology/Bone Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI
| | - Amanda M. Brandow
- Department of Pediatrics, Section of Hematology/Oncology/Bone Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI
| | - Cheryl L. Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Allison D. Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
28
|
Xie YF, Yang J, Ratté S, Prescott SA. Similar excitability through different sodium channels and implications for the analgesic efficacy of selective drugs. eLife 2024; 12:RP90960. [PMID: 38687187 PMCID: PMC11060714 DOI: 10.7554/elife.90960] [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] [Indexed: 05/02/2024] Open
Abstract
Nociceptive sensory neurons convey pain-related signals to the CNS using action potentials. Loss-of-function mutations in the voltage-gated sodium channel NaV1.7 cause insensitivity to pain (presumably by reducing nociceptor excitability) but clinical trials seeking to treat pain by inhibiting NaV1.7 pharmacologically have struggled. This may reflect the variable contribution of NaV1.7 to nociceptor excitability. Contrary to claims that NaV1.7 is necessary for nociceptors to initiate action potentials, we show that nociceptors can achieve similar excitability using different combinations of NaV1.3, NaV1.7, and NaV1.8. Selectively blocking one of those NaV subtypes reduces nociceptor excitability only if the other subtypes are weakly expressed. For example, excitability relies on NaV1.8 in acutely dissociated nociceptors but responsibility shifts to NaV1.7 and NaV1.3 by the fourth day in culture. A similar shift in NaV dependence occurs in vivo after inflammation, impacting ability of the NaV1.7-selective inhibitor PF-05089771 to reduce pain in behavioral tests. Flexible use of different NaV subtypes exemplifies degeneracy - achieving similar function using different components - and compromises reliable modulation of nociceptor excitability by subtype-selective inhibitors. Identifying the dominant NaV subtype to predict drug efficacy is not trivial. Degeneracy at the cellular level must be considered when choosing drug targets at the molecular level.
Collapse
Affiliation(s)
- Yu-Feng Xie
- Neurosciences and Mental Health, The Hospital for Sick ChildrenTorontoCanada
| | - Jane Yang
- Neurosciences and Mental Health, The Hospital for Sick ChildrenTorontoCanada
- Institute of Biomedical Engineering, University of TorontoTorontoCanada
| | - Stéphanie Ratté
- Neurosciences and Mental Health, The Hospital for Sick ChildrenTorontoCanada
| | - Steven A Prescott
- Neurosciences and Mental Health, The Hospital for Sick ChildrenTorontoCanada
- Institute of Biomedical Engineering, University of TorontoTorontoCanada
- Department of Physiology, University of TorontoTorontoCanada
| |
Collapse
|
29
|
Kalia AK, Rösseler C, Granja-Vazquez R, Ahmad A, Pancrazio JJ, Neureiter A, Zhang M, Sauter D, Vetter I, Andersson A, Dussor G, Price TJ, Kolber BJ, Truong V, Walsh P, Lampert A. How to differentiate induced pluripotent stem cells into sensory neurons for disease modelling: a functional assessment. Stem Cell Res Ther 2024; 15:99. [PMID: 38581069 PMCID: PMC10998320 DOI: 10.1186/s13287-024-03696-2] [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: 07/07/2023] [Accepted: 03/13/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Human induced pluripotent stem cell (iPSC)-derived peripheral sensory neurons present a valuable tool to model human diseases and are a source for applications in drug discovery and regenerative medicine. Clinically, peripheral sensory neuropathies can result in maladies ranging from a complete loss of pain to severe painful neuropathic disorders. Sensory neurons are located in the dorsal root ganglion and are comprised of functionally diverse neuronal types. Low efficiency, reproducibility concerns, variations arising due to genetic factors and time needed to generate functionally mature neuronal populations from iPSCs remain key challenges to study human nociception in vitro. Here, we report a detailed functional characterization of iPSC-derived sensory neurons with an accelerated differentiation protocol ("Anatomic" protocol) compared to the most commonly used small molecule approach ("Chambers" protocol). Anatomic's commercially available RealDRG™ were further characterized for both functional and expression phenotyping of key nociceptor markers. METHODS Multiple iPSC clones derived from different reprogramming methods, genetics, age, and somatic cell sources were used to generate sensory neurons. Manual patch clamp was used to functionally characterize both control and patient-derived neurons. High throughput techniques were further used to demonstrate that RealDRGs™ derived from the Anatomic protocol are amenable to high throughput technologies for disease modelling. RESULTS The Anatomic protocol rendered a purer culture without the use of mitomycin C to suppress non-neuronal outgrowth, while Chambers differentiations yielded a mix of cell types. Chambers protocol results in predominantly tonic firing when compared to Anatomic protocol. Patient-derived nociceptors displayed higher frequency firing compared to control subject with both, Chambers and Anatomic differentiation approaches, underlining their potential use for clinical phenotyping as a disease-in-a-dish model. RealDRG™ sensory neurons show heterogeneity of nociceptive markers indicating that the cells may be useful as a humanized model system for translational studies. CONCLUSIONS We validated the efficiency of two differentiation protocols and their potential application for functional assessment and thus understanding the disease mechanisms from patients suffering from pain disorders. We propose that both differentiation methods can be further exploited for understanding mechanisms and development of novel treatments in pain disorders.
Collapse
Affiliation(s)
- Anil Kumar Kalia
- Institute of Neurophysiology, Uniklinik RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
- Research Training Group 2416 MultiSenses-MultiScales, RWTH Aachen University, Aachen, Germany
| | - Corinna Rösseler
- Institute of Neurophysiology, Uniklinik RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Rafael Granja-Vazquez
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Ayesha Ahmad
- Department of Neuroscience, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Joseph J Pancrazio
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Anika Neureiter
- Institute of Neurophysiology, Uniklinik RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Mei Zhang
- Sophion Bioscience Inc., Bedford, MA, 01730, USA
| | | | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Asa Andersson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Gregory Dussor
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
- Department of Neuroscience, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Theodore J Price
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
- Department of Neuroscience, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Benedict J Kolber
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
- Department of Neuroscience, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Vincent Truong
- Anatomic Incorporated, 2112 Broadway Street NE #135, Minneapolis, MN, 55413, USA
| | - Patrick Walsh
- Anatomic Incorporated, 2112 Broadway Street NE #135, Minneapolis, MN, 55413, USA
| | - Angelika Lampert
- Institute of Neurophysiology, Uniklinik RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany.
- Research Training Group 2416 MultiSenses-MultiScales, RWTH Aachen University, Aachen, Germany.
- Scientific Center for Neuropathic Pain Aachen - SCN-Aachen, Uniklinik RWTH Aachen University, 52074, Aachen, Germany.
| |
Collapse
|
30
|
Klein T, Grüner J, Breyer M, Schlegel J, Schottmann NM, Hofmann L, Gauss K, Mease R, Erbacher C, Finke L, Klein A, Klug K, Karl-Schöller F, Vignolo B, Reinhard S, Schneider T, Günther K, Fink J, Dudek J, Maack C, Klopocki E, Seibel J, Edenhofer F, Wischmeyer E, Sauer M, Üçeyler N. Small fibre neuropathy in Fabry disease: a human-derived neuronal in vitro disease model and pilot data. Brain Commun 2024; 6:fcae095. [PMID: 38638148 PMCID: PMC11024803 DOI: 10.1093/braincomms/fcae095] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/24/2024] [Accepted: 03/15/2024] [Indexed: 04/20/2024] Open
Abstract
Acral burning pain triggered by fever, thermal hyposensitivity and skin denervation are hallmarks of small fibre neuropathy in Fabry disease, a life-threatening X-linked lysosomal storage disorder. Variants in the gene encoding alpha-galactosidase A may lead to impaired enzyme activity with cellular accumulation of globotriaosylceramide. To study the underlying pathomechanism of Fabry-associated small fibre neuropathy, we generated a neuronal in vitro disease model using patient-derived induced pluripotent stem cells from three Fabry patients and one healthy control. We further generated an isogenic control line via gene editing. We subjected induced pluripotent stem cells to targeted peripheral neuronal differentiation and observed intra-lysosomal globotriaosylceramide accumulations in somas and neurites of Fabry sensory neurons using super-resolution microscopy. At functional level, patch-clamp analysis revealed a hyperpolarizing shift of voltage-gated sodium channel steady-state inactivation kinetics in isogenic control neurons compared with healthy control neurons (P < 0.001). Moreover, we demonstrate a drastic increase in Fabry sensory neuron calcium levels at 39°C mimicking clinical fever (P < 0.001). This pathophysiological phenotype was accompanied by thinning of neurite calibres in sensory neurons differentiated from induced pluripotent stem cells derived from Fabry patients compared with healthy control cells (P < 0.001). Linear-nonlinear cascade models fit to spiking responses revealed that Fabry cell lines exhibit altered single neuron encoding properties relative to control. We further observed mitochondrial aggregation at sphingolipid accumulations within Fabry sensory neurites utilizing a click chemistry approach together with mitochondrial dysmorphism compared with healthy control cells. We pioneer pilot insights into the cellular mechanisms contributing to pain, thermal hyposensitivity and denervation in Fabry small fibre neuropathy and pave the way for further mechanistic in vitro studies in Fabry disease and the development of novel treatment approaches.
Collapse
Affiliation(s)
- Thomas Klein
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Julia Grüner
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Maximilian Breyer
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Jan Schlegel
- Department of Biotechnology and Biophysics, University of Würzburg, 97074 Würzburg, Germany
| | | | - Lukas Hofmann
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Kevin Gauss
- Medical Biophysics, Institute for Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Rebecca Mease
- Medical Biophysics, Institute for Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Christoph Erbacher
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Laura Finke
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Alexandra Klein
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Katharina Klug
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany
| | | | - Bettina Vignolo
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Sebastian Reinhard
- Department of Biotechnology and Biophysics, University of Würzburg, 97074 Würzburg, Germany
| | - Tamara Schneider
- Institute for Human Genetics, University of Würzburg, 97074 Würzburg, Germany
| | - Katharina Günther
- Institute of Anatomy and Cell Biology, University of Würzburg, 97070 Würzburg, Germany
| | - Julian Fink
- Institute of Organic Chemistry, University of Würzburg, 97074 Würzburg, Germany
| | - Jan Dudek
- Comprehensive Heart Failure Center CHFC, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center CHFC, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Eva Klopocki
- Institute for Human Genetics, University of Würzburg, 97074 Würzburg, Germany
| | - Jürgen Seibel
- Institute of Organic Chemistry, University of Würzburg, 97074 Würzburg, Germany
| | - Frank Edenhofer
- Institute of Anatomy and Cell Biology, University of Würzburg, 97070 Würzburg, Germany
| | - Erhard Wischmeyer
- Institute of Physiology, University of Würzburg, 97070 Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, University of Würzburg, 97074 Würzburg, Germany
| | - Nurcan Üçeyler
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany
- Würzburg Fabry Center for Interdisciplinary Therapy (FAZIT), University Hospital Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
31
|
Kim JI, Imaizumi K, Thete MV, Hudacova Z, Jurjuţ O, Amin ND, Scherrer G, Paşca SP. Human assembloid model of the ascending neural sensory pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584539. [PMID: 38559133 PMCID: PMC10979925 DOI: 10.1101/2024.03.11.584539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The ascending somatosensory pathways convey crucial information about pain, touch, itch, and body part movement from peripheral organs to the central nervous system. Despite a significant need for effective therapeutics modulating pain and other somatosensory modalities, clinical translation remains challenging, which is likely related to species-specific features and the lack of in vitro models to directly probe and manipulate this polysynaptic pathway. Here, we established human ascending somatosensory assembloids (hASA)- a four-part assembloid completely generated from human pluripotent stem cells that integrates somatosensory, spinal, diencephalic, and cortical organoids to model the human ascending spinothalamic pathway. Transcriptomic profiling confirmed the presence of key cell types in this circuit. Rabies tracing and calcium imaging showed that sensory neurons connected with dorsal spinal cord projection neurons, which ascending axons further connected to thalamic neurons. Following noxious chemical stimulation, single neuron calcium imaging of intact hASA demonstrated coordinated response, while four-part concomitant extracellular recordings and calcium imaging revealed synchronized activity across the assembloid. Loss of the sodium channel SCN9A, which causes pain insensitivity in humans, disrupted synchrony across the four-part hASA. Taken together, these experiments demonstrate the ability to functionally assemble the essential components of the human sensory pathway. These findings could both accelerate our understanding of human sensory circuits and facilitate therapeutic development.
Collapse
|
32
|
Ullah Z, Zafar A, Ishaq H, Umar Z, Khan A, Badar Y, Din N, Khan MF, McCombe P, Khan N. Transient binocular vision loss and pain insensitivity in Klippel-Feil syndrome: a case report. J Med Case Rep 2024; 18:137. [PMID: 38444009 PMCID: PMC10916052 DOI: 10.1186/s13256-024-04374-w] [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: 12/25/2023] [Accepted: 01/11/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Klippel-Feil syndrome is a rare congenital bone disorder characterized by an abnormal fusion of two or more cervical spine vertebrae. Individuals with Klippel-Feil syndrome exhibit diverse clinical manifestations, including skeletal irregularities, visual and hearing impairments, orofacial anomalies, and anomalies in various internal organs, such as the heart, kidneys, genitourinary system, and nervous system. CASE PRESENTATION This case report describes a 12-year-old Pashtun female patient who presented with acute bilateral visual loss. The patient had Klippel-Feil syndrome, with the typical clinical triad symptoms of Klippel-Feil syndrome, along with Sprengel's deformity. She also exhibited generalized hypoalgesia, which had previously resulted in widespread burn-related injuries. Upon examination, bilateral optic disc swelling was observed, but intracranial pressure was found to be normal. Extensive investigations yielded normal results, except for hypocalcemia and low vitamin D levels, while parathyroid function remained within the normal range. Visual acuity improved following 2 months of calcium and vitamin D supplementation, suggesting that the visual loss and optic nerve swelling were attributed to hypocalcemia. Given the normal parathyroid function, it is possible that hypocalcemia resulted from low vitamin D levels, which can occur after severe burn scarring. Furthermore, the patient received a provisional diagnosis of congenital insensitivity to pain on the basis of the detailed medical history and the findings of severe and widespread loss of the ability to perceive painful stimuli, as well as impaired temperature sensation. However, due to limitations in genetic testing, confirmation of the congenital insensitivity to pain diagnosis could not be obtained. CONCLUSION This case highlights a rare presentation of transient binocular vision loss and pain insensitivity in a patient with Klippel-Feil syndrome, emphasizing the importance of considering unusual associations in symptom interpretation.
Collapse
Affiliation(s)
- Zeeshan Ullah
- Department of Neurology, Lady Reading Hospital, Peshawar, Pakistan
| | - Ayesha Zafar
- Department of Neurology, Lady Reading Hospital, Peshawar, Pakistan
| | - Hira Ishaq
- Department of Neurology, Lady Reading Hospital, Peshawar, Pakistan
| | - Zainab Umar
- Department of Neonatology, Pakistan Institute of Medical Sciences, Islamabad, Pakistan
| | - Amir Khan
- Department of Neurology, Lady Reading Hospital, Peshawar, Pakistan
| | - Yaseen Badar
- Department of Neurology, Lady Reading Hospital, Peshawar, Pakistan
| | - Nizamud Din
- Department of Neurology, Lady Reading Hospital, Peshawar, Pakistan
| | | | - Pamela McCombe
- Faculty of Medicine, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Herston, Brisbane, QLD, 4029, Australia.
| | - Nemat Khan
- College of Medicine and Health Science, Khalifa University of Science and Technology, 127788, Abu Dhabi, United Arab Emirates.
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St. Lucia Campus, Brisbane, Australia.
| |
Collapse
|
33
|
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
|
34
|
Makau CM, Towett PK, Kanui TI, Abelson KSP. Effects of inhibition of Nav1.3, Nav1.7, and Nav1.8 channels on pain-related behavior in Speke's hinge-back tortoise (Kinixys spekii). J Neurosci Res 2024; 102:e25274. [PMID: 38284848 DOI: 10.1002/jnr.25274] [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: 06/01/2023] [Revised: 10/10/2023] [Accepted: 10/25/2023] [Indexed: 01/30/2024]
Abstract
Comparative studies using reptiles as experimental animals in pain research could expand our knowledge on the evolution and adaptation of pain mechanisms. Currently, there are no data reported on the involvement of voltage-gated sodium ion channels on nociception in reptiles. The aim of this study was to investigate the involvement of Nav1.3, Nav1.7, and Nav1.8 ion channels in nociception in Speke's hinge-back tortoise. ICA 121341 (selective blocker for Nav1.1/Nav1.3), NAV 26 (selective blocker for Nav1.7), and A803467 (selective blocker for Nav1.8) were used to investigate the involvement of Nav1.3, Nav1.7, and Nav1.8, respectively. The chemicals were administered intracoelomically thirty minutes before the start of nociceptive tests. ICA 121341 did not cause a significant decrease in the time spent in pain-related behavior in all the nociceptive tests. NAV 26 and A8034667 caused a statistically significant decrease in the mean time spent in pain-related behavior in the formalin and capsaicin tests. Only A803467 caused a statistically significant increase in the mean latency to pain-related behavior in the hot plate test. NAV 26 and A803467 had no observable side effects. In conclusion, Nav1.7 and Nav1.8 are involved in the processing of chemically induced inflammatory pain in Speke's hinge back tortoise. In addition, Nav1.8 are also significantly involved in the development of thermal-induced pain-related behavior in this species of reptile. However, our results do not support the involvement of Nav1.3 on the development of chemical or thermal induced pain-related behavior in the Speke's hinge back tortoise.
Collapse
Affiliation(s)
- Christopher M Makau
- Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| | - Philemon K Towett
- Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| | - Titus I Kanui
- School of Agriculture and Veterinary Sciences, South Eastern Kenya University, Kitui, Kenya
| | - Klas S P Abelson
- Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
35
|
Harraz OF, Delpire E. Recent insights into channelopathies. Physiol Rev 2024; 104:23-31. [PMID: 37561136 DOI: 10.1152/physrev.00022.2023] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/12/2023] [Accepted: 08/05/2023] [Indexed: 08/11/2023] Open
Affiliation(s)
- Osama F Harraz
- Department of Pharmacology, Larner College of Medicine, Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont, United States
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical School, Nashville, Tennessee, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical School, Nashville, Tennessee, United States
| |
Collapse
|
36
|
Shehab S, Javed H, Johnson AM, Tariq S, Kumar CA, Emerald BS. Unveiling the mechanisms of neuropathic pain suppression: perineural resiniferatoxin targets Trpv1 and beyond. Front Neuroanat 2023; 17:1306180. [PMID: 38099210 PMCID: PMC10720729 DOI: 10.3389/fnana.2023.1306180] [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: 10/03/2023] [Accepted: 11/10/2023] [Indexed: 12/17/2023] Open
Abstract
Neuropathic pain arises from damage or disorders affecting the somatosensory system. In rats, L5 nerve injury induces thermal and mechanical hypersensitivity/hyperalgesia. Recently, we demonstrated that applying resiniferatoxin (RTX) directly on uninjured L3 and L4 nerves alleviated thermal and mechanical hypersensitivity resulting from L5 nerve injury. Herein, using immunohistochemistry, Western blot, and qRT-PCR techniques, we reveal that perineural application of RTX (0.002%) on the L4 nerve substantially downregulated the expression of its receptor (Trpv1) and three different voltage-gated ion channels (Nav1.9, Kv4.3, and Cav2.2). These channels are found primarily in small-sized neurons and show significant colocalization with Trpv1 in the dorsal root ganglion (DRG). However, RTX treatment did not affect the expression of Kv1.1, Piezo2 (found in large-sized neurons without colocalization with Trpv1), and Kir4.1 (localized in satellite cells) in the ipsilateral DRGs. Furthermore, RTX application on L3 and L4 nerves reduced the activation of c-fos in the spinal neurons induced by heat stimulation. Subsequently, we investigated whether applying RTX to the L3 and L4 nerves 3 weeks before the L5 nerve injury could prevent the onset of neuropathic pain. Both 0.002 and 0.004% concentrations of RTX produced significant analgesic effects, while complete prevention of thermal and mechanical hypersensitivity required a concentration of 0.008%. Importantly, this preventive effect on neuropathic manifestations was not associated with nerve degeneration, as microscopic examination revealed no morphological changes. Overall, this study underscores the mechanisms and the significance of perineural RTX treatment applied to adjacent uninjured nerves in entirely preventing nerve injury-induced neuropathic pain in humans and animals.
Collapse
Affiliation(s)
- Safa Shehab
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | | | | | | | | | | |
Collapse
|
37
|
Fetell M, Sendel M, Li T, Marinelli L, Vollert J, Ruggerio E, Houk G, Dockum M, Albrecht PJ, Rice FL, Baron R. Cutaneous nerve fiber and peripheral Nav1.7 assessment in a large cohort of patients with postherpetic neuralgia. Pain 2023; 164:2435-2446. [PMID: 37366590 PMCID: PMC10578423 DOI: 10.1097/j.pain.0000000000002950] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/06/2023] [Accepted: 04/25/2023] [Indexed: 06/28/2023]
Abstract
ABSTRACT The mechanisms of pain in postherpetic neuralgia (PHN) are still unclear, with some studies showing loss of cutaneous sensory nerve fibers that seemed to correlate with pain level. We report results of skin biopsies and correlations with baseline pain scores, mechanical hyperalgesia, and the Neuropathic Pain Symptom Inventory (NPSI) in 294 patients who participated in a clinical trial of TV-45070, a topical semiselective sodium 1.7 channel (Nav1.7) blocker. Intraepidermal nerve fibers and subepidermal Nav1.7 immunolabeled fibers were quantified in skin punch biopsies from the area of maximal PHN pain, as well as from the contralateral, homologous (mirror image) region. Across the entire study population, a 20% reduction in nerve fibers on the PHN-affected side compared with that in the contralateral side was noted; however, the reduction was much higher in older individuals, approaching 40% in those aged 70 years or older. There was a decrease in contralateral fiber counts as well, also noted in prior biopsy studies, the mechanism of which is not fully clear. Nav1.7-positive immunolabeling was present in approximately one-third of subepidermal nerve fibers and did not differ on the PHN-affected vs contralateral sides. Using cluster analysis, 2 groups could be identified, with the first cluster showing higher baseline pain, higher NPSI scores for squeezing and cold-induced pain, higher nerve fiber density, and higher Nav1.7 expression. While Nav1.7 varies from patient to patient, it does not seem to be a key pathophysiological driver of PHN pain. Individual differences in Nav1.7 expression, however, may determine the intensity and sensory aspects of pain.
Collapse
Affiliation(s)
| | - Manon Sendel
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital of Schleswig-Holstein, Campus Kiel, Germany
| | - Thomas Li
- Teva Pharmaceuticals, West Chester, PA, United States
| | | | - Jan Vollert
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital of Schleswig-Holstein, Campus Kiel, Germany
- Pain Research, MSk Lab, Department of Surgery and Cancer, Imperial College, London, United Kingdom
- Department of Neurophysiology, Mannheim Center for Translational Neuroscience MCTN, Medical Faculty Mannheim, Ruprecht Karls University, Heidelberg, Germany
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | | | - George Houk
- Integrated Tissue Dynamics LLC, Rensselaer, NY, United States
| | - Marilyn Dockum
- Integrated Tissue Dynamics LLC, Rensselaer, NY, United States
| | | | - Frank L. Rice
- Integrated Tissue Dynamics LLC, Rensselaer, NY, United States
| | - Ralf Baron
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital of Schleswig-Holstein, Campus Kiel, Germany
| |
Collapse
|
38
|
Kalia AK, Rösseler C, Granja-Vazquez R, Ahmad A, Pancrazio JJ, Neureiter A, Zhang M, Sauter D, Vetter I, Andersson A, Dussor G, Price TJ, Kolber BJ, Truong V, Walsh P, Lampert A. How to differentiate induced pluripotent stem cells into sensory neurons for disease modelling: a comparison of two protocols. RESEARCH SQUARE 2023:rs.3.rs-3127017. [PMID: 37961300 PMCID: PMC10635298 DOI: 10.21203/rs.3.rs-3127017/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Human induced pluripotent stem cell (iPSC)-derived peripheral sensory neurons present a valuable tool to model human diseases and are a source for applications in drug discovery and regenerative medicine. Clinically, peripheral sensory neuropathies can result in maladies ranging from a complete loss of pain to severe painful neuropathic symptoms. Sensory neurons are located in the dorsal root ganglion and are comprised of functionally diverse neuronal types. Low efficiency, reproducibility concerns, variations arising due to genetic factors and time needed to generate functionally mature neuronal populations from iPSCs for disease modelling remain key challenges to study human nociception in vitro. Here, we report a detailed characterization of iPSC-derived sensory neurons with an accelerated differentiation protocol ("Anatomic" protocol) compared to the most commonly used small molecule approach ("Chambers" protocol). Methods Multiple iPSC clones derived from different reprogramming methods, genetics, age, and somatic cell sources were used to generate sensory neurons. Expression profiling of sensory neurons was performed with Immunocytochemistry and in situ hybridization techniques. Manual patch clamp and high throughput cellular screening systems (Fluorescence imaging plate reader, automated patch clamp and multi-well microelectrode arrays recordings) were applied to functionally characterize the generated sensory neurons. Results The Anatomic protocol rendered a purer culture without the use of mitomycin C to suppress non-neuronal outgrowth, while Chambers differentiations yielded a mix of cell types. High throughput systems confirmed functional expression of Na+ and K+ ion channels. Multi-well microelectrode recordings display spontaneously active neurons with sensitivity to increased temperature indicating expression of heat sensitive ion channels. Patient-derived nociceptors displayed higher frequency firing compared to control subject with both, Chambers and Anatomic differentiation approaches, underlining their potential use for clinical phenotyping as a disease-in-a-dish model. Conclusions We validated the efficiency of two differentiation protocols and their potential application for understanding the disease mechanisms from patients suffering from pain disorders. We propose that both differentiation methods can be further exploited for understanding mechanisms and development of novel treatments in pain disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mei Zhang
- Sophion Bioscience A/S: Biolin Scientific AB
| | | | - Irina Vetter
- The University of Queensland Institute for Molecular Bioscience
| | - Asa Andersson
- The University of Queensland Institute for Molecular Bioscience
| | | | | | | | | | | | | |
Collapse
|
39
|
Zhao Q, Zhang X, Long S, Wang S, Yu H, Zhou Y, Li Y, Xue L, Hu Y, Yin S. Licochalcone Mediates the Pain Relief by Targeting the Voltage-Gated Sodium Channel. Mol Pharmacol 2023; 104:133-143. [PMID: 37419692 DOI: 10.1124/molpharm.122.000658] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 06/04/2023] [Accepted: 06/08/2023] [Indexed: 07/09/2023] Open
Abstract
Licorice is a traditional Chinese medicine and recorded to have pain relief effects in national pharmacopoeia, but the mechanisms behind these effects have not been fully explored. Among the hundreds of compounds in licorice, licochalcone A (LCA) and licochalcone B (LCB) are two important components belonging to the chalcone family. In this study, we compared the analgesic effects of these two licochalcones and the molecular mechanisms. LCA and LCB were applied in cultured dorsal root ganglion (DRG) neurons, and the voltage-gated sodium (NaV) currents and action potentials were recorded. The electrophysiological experiments showed that LCA can inhibit NaV currents and dampen excitabilities of DRG neurons, whereas LCB did not show inhibition effect on NaV currents. Because the NaV1.7 channel can modulate Subthreshold membrane potential oscillations in DRG neuron, which can palliate neuropathic pain, HEK293T cells were transfected with NaV1.7 channel and recorded with whole-cell patch clamp. LCA can also inhibit NaV1.7 channels exogenously expressed in HEK293T cells. We further explored the analgesic effects of LCA and LCB on formalin-induced pain animal models. The animal behavior tests revealed that LCA can inhibit the pain responses during phase 1 and phase 2 of formalin test, and LCB can inhibit the pain responses during phase 2. The differences of the effects on NaV currents between LCA and LCB provide us with the basis for developing NaV channel inhibitors, and the novel findings of analgesic effects indicate that licochalcones can be developed into effective analgesic medicines. SIGNIFICANCE STATEMENT: This study found that licochalcone A (LCA) can inhibit voltage-gated sodium (NaV) currents, dampen excitabilities of dorsal root ganglion neurons, and inhibit the NaV1.7 channels exogenously expressed in HEK293T cells. Animal behavior tests showed that LCA can inhibit the pain responses during phase 1 and phase 2 of formalin test, whereas licochalcone B can inhibit the pain responses during phase 2. These findings indicate that licochalcones could be the leading compounds for developing NaV channel inhibitors and effective analgesic medicines.
Collapse
Affiliation(s)
- Qianru Zhao
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Q.Z., X.Z., S.L., S.W., H.Y., Y.Z., Y.H., S.Y.) and Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Y.L., L.X.)
| | - Xu Zhang
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Q.Z., X.Z., S.L., S.W., H.Y., Y.Z., Y.H., S.Y.) and Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Y.L., L.X.)
| | - Siru Long
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Q.Z., X.Z., S.L., S.W., H.Y., Y.Z., Y.H., S.Y.) and Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Y.L., L.X.)
| | - Shaobing Wang
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Q.Z., X.Z., S.L., S.W., H.Y., Y.Z., Y.H., S.Y.) and Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Y.L., L.X.)
| | - Hui Yu
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Q.Z., X.Z., S.L., S.W., H.Y., Y.Z., Y.H., S.Y.) and Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Y.L., L.X.)
| | - Yongsheng Zhou
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Q.Z., X.Z., S.L., S.W., H.Y., Y.Z., Y.H., S.Y.) and Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Y.L., L.X.)
| | - Yi Li
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Q.Z., X.Z., S.L., S.W., H.Y., Y.Z., Y.H., S.Y.) and Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Y.L., L.X.)
| | - Lu Xue
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Q.Z., X.Z., S.L., S.W., H.Y., Y.Z., Y.H., S.Y.) and Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Y.L., L.X.)
| | - Yan Hu
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Q.Z., X.Z., S.L., S.W., H.Y., Y.Z., Y.H., S.Y.) and Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Y.L., L.X.)
| | - Shijin Yin
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Q.Z., X.Z., S.L., S.W., H.Y., Y.Z., Y.H., S.Y.) and Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central Minzu University, Wuhan, People's Republic of China (Y.L., L.X.)
| |
Collapse
|
40
|
Santiago C, Sharma N, Africawala N, Siegrist J, Handler A, Tasnim A, Anjum R, Turecek J, Lehnert BP, Renauld S, Nolan-Tamariz M, Iskols M, Magee AR, Paradis S, Ginty DD. Activity-dependent development of the body's touch receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.23.559109. [PMID: 37790437 PMCID: PMC10542488 DOI: 10.1101/2023.09.23.559109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
We report a role for activity in the development of the primary sensory neurons that detect touch. Genetic deletion of Piezo2, the principal mechanosensitive ion channel in somatosensory neurons, caused profound changes in the formation of mechanosensory end organ structures and altered somatosensory neuron central targeting. Single cell RNA sequencing of Piezo2 conditional mutants revealed changes in gene expression in the sensory neurons activated by light mechanical forces, whereas other neuronal classes were less affected. To further test the role of activity in mechanosensory end organ development, we genetically deleted the voltage-gated sodium channel Nav1.6 (Scn8a) in somatosensory neurons throughout development and found that Scn8a mutants also have disrupted somatosensory neuron morphologies and altered electrophysiological responses to mechanical stimuli. Together, these findings indicate that mechanically evoked neuronal activity acts early in life to shape the maturation of the mechanosensory end organs that underlie our sense of gentle touch.
Collapse
Affiliation(s)
- Celine Santiago
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Nikhil Sharma
- Department of Molecular Pharmacology and Therapeutics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Nusrat Africawala
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Julianna Siegrist
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Annie Handler
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Aniqa Tasnim
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Rabia Anjum
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, MA, 02453, USA
| | - Josef Turecek
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Brendan P. Lehnert
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Sophia Renauld
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Michael Nolan-Tamariz
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Michael Iskols
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Alexandra R. Magee
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Suzanne Paradis
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, MA, 02453, USA
| | - David D. Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
- Lead Contact
| |
Collapse
|
41
|
Smith PA. Neuropathic pain; what we know and what we should do about it. FRONTIERS IN PAIN RESEARCH 2023; 4:1220034. [PMID: 37810432 PMCID: PMC10559888 DOI: 10.3389/fpain.2023.1220034] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Neuropathic pain can result from injury to, or disease of the nervous system. It is notoriously difficult to treat. Peripheral nerve injury promotes Schwann cell activation and invasion of immunocompetent cells into the site of injury, spinal cord and higher sensory structures such as thalamus and cingulate and sensory cortices. Various cytokines, chemokines, growth factors, monoamines and neuropeptides effect two-way signalling between neurons, glia and immune cells. This promotes sustained hyperexcitability and spontaneous activity in primary afferents that is crucial for onset and persistence of pain as well as misprocessing of sensory information in the spinal cord and supraspinal structures. Much of the current understanding of pain aetiology and identification of drug targets derives from studies of the consequences of peripheral nerve injury in rodent models. Although a vast amount of information has been forthcoming, the translation of this information into the clinical arena has been minimal. Few, if any, major therapeutic approaches have appeared since the mid 1990's. This may reflect failure to recognise differences in pain processing in males vs. females, differences in cellular responses to different types of injury and differences in pain processing in humans vs. animals. Basic science and clinical approaches which seek to bridge this knowledge gap include better assessment of pain in animal models, use of pain models which better emulate human disease, and stratification of human pain phenotypes according to quantitative assessment of signs and symptoms of disease. This can lead to more personalized and effective treatments for individual patients. Significance statement: There is an urgent need to find new treatments for neuropathic pain. Although classical animal models have revealed essential features of pain aetiology such as peripheral and central sensitization and some of the molecular and cellular mechanisms involved, they do not adequately model the multiplicity of disease states or injuries that may bring forth neuropathic pain in the clinic. This review seeks to integrate information from the multiplicity of disciplines that seek to understand neuropathic pain; including immunology, cell biology, electrophysiology and biophysics, anatomy, cell biology, neurology, molecular biology, pharmacology and behavioral science. Beyond this, it underlines ongoing refinements in basic science and clinical practice that will engender improved approaches to pain management.
Collapse
Affiliation(s)
- Peter A. Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
42
|
Röderer P, Belu A, Heidrich L, Siobal M, Isensee J, Prolingheuer J, Janocha E, Valdor M, Hagendorf S, Bahrenberg G, Opitz T, Segschneider M, Haupt S, Nitzsche A, Brüstle O, Hucho T. Emergence of nociceptive functionality and opioid signaling in human induced pluripotent stem cell-derived sensory neurons. Pain 2023; 164:1718-1733. [PMID: 36727909 PMCID: PMC10348637 DOI: 10.1097/j.pain.0000000000002860] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 10/31/2022] [Accepted: 11/15/2022] [Indexed: 02/03/2023]
Abstract
ABSTRACT Induced pluripotent stem cells (iPSCs) have enabled the generation of various difficult-to-access cell types such as human nociceptors. A key challenge associated with human iPSC-derived nociceptors (hiPSCdNs) is their prolonged functional maturation. While numerous studies have addressed the expression of classic neuronal markers and ion channels in hiPSCdNs, the temporal development of key signaling cascades regulating nociceptor activity has remained largely unexplored. In this study, we used an immunocytochemical high-content imaging approach alongside electrophysiological staging to assess metabotropic and ionotropic signaling of large scale-generated hiPSCdNs across 70 days of in vitro differentiation. During this period, the resting membrane potential became more hyperpolarized, while rheobase, action potential peak amplitude, and membrane capacitance increased. After 70 days, hiPSCdNs exhibited robust physiological responses induced by GABA, pH shift, ATP, and capsaicin. Direct activation of protein kinase A type II (PKA-II) through adenylyl cyclase stimulation with forskolin resulted in PKA-II activation at all time points. Depolarization-induced activation of PKA-II emerged after 35 days of differentiation. However, effective inhibition of forskolin-induced PKA-II activation by opioid receptor agonists required 70 days of in vitro differentiation. Our results identify a pronounced time difference between early expression of functionally important ion channels and emergence of regulatory metabotropic sensitizing and desensitizing signaling only at advanced stages of in vitro cultivation, suggesting an independent regulation of ionotropic and metabotropic signaling. These data are relevant for devising future studies into the development and regulation of human nociceptor function and for defining time windows suitable for hiPSCdN-based drug discovery.
Collapse
Affiliation(s)
- Pascal Röderer
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Andreea Belu
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Luzia Heidrich
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Maike Siobal
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jörg Isensee
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jonathan Prolingheuer
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | | | | | | | - Thoralf Opitz
- Institute of Experimental Epileptology and Cognition Research, University of Bonn, Bonn, Germany
| | - Michaela Segschneider
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
| | - Simone Haupt
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Anja Nitzsche
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Tim Hucho
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
43
|
Hung CH, Chin Y, Fong YO, Lee CH, Han DS, Lin JH, Sun WH, Chen CC. Acidosis-related pain and its receptors as targets for chronic pain. Pharmacol Ther 2023; 247:108444. [PMID: 37210007 DOI: 10.1016/j.pharmthera.2023.108444] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/24/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Sensing acidosis is an important somatosensory function in responses to ischemia, inflammation, and metabolic alteration. Accumulating evidence has shown that acidosis is an effective factor for pain induction and that many intractable chronic pain diseases are associated with acidosis signaling. Various receptors have been known to detect extracellular acidosis and all express in the somatosensory neurons, such as acid sensing ion channels (ASIC), transient receptor potential (TRP) channels and proton-sensing G-protein coupled receptors. In addition to sense noxious acidic stimulation, these proton-sensing receptors also play a vital role in pain processing. For example, ASICs and TRPs are involved in not only nociceptive activation but also anti-nociceptive effects as well as some other non-nociceptive pathways. Herein, we review recent progress in probing the roles of proton-sensing receptors in preclinical pain research and their clinical relevance. We also propose a new concept of sngception to address the specific somatosensory function of acid sensation. This review aims to connect these acid-sensing receptors with basic pain research and clinical pain diseases, thus helping with better understanding the acid-related pain pathogenesis and their potential therapeutic roles via the mechanism of acid-mediated antinociception.
Collapse
Affiliation(s)
- Chih-Hsien Hung
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yin Chin
- Department of Life Science & Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-On Fong
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Cheng-Han Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Der-Shen Han
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan
| | - Jiann-Her Lin
- Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan; Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Wei-Hsin Sun
- Department of Life Science & Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Cheng Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan; Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan; Neuroscience Program of Academia Sinica, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
44
|
Deng T, Jovanovic VM, Tristan CA, Weber C, Chu PH, Inman J, Ryu S, Jethmalani Y, Ferreira de Sousa J, Ormanoglu P, Twumasi P, Sen C, Shim J, Jayakar S, Bear Zhang HX, Jo S, Yu W, Voss TC, Simeonov A, Bean BP, Woolf CJ, Singeç I. Scalable generation of sensory neurons from human pluripotent stem cells. Stem Cell Reports 2023; 18:1030-1047. [PMID: 37044067 PMCID: PMC10147831 DOI: 10.1016/j.stemcr.2023.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 04/14/2023] Open
Abstract
Development of new non-addictive analgesics requires advanced strategies to differentiate human pluripotent stem cells (hPSCs) into relevant cell types. Following principles of developmental biology and translational applicability, here we developed an efficient stepwise differentiation method for peptidergic and non-peptidergic nociceptors. By modulating specific cell signaling pathways, hPSCs were first converted into SOX10+ neural crest, followed by differentiation into sensory neurons. Detailed characterization, including ultrastructural analysis, confirmed that the hPSC-derived nociceptors displayed cellular and molecular features comparable to native dorsal root ganglion (DRG) neurons, and expressed high-threshold primary sensory neuron markers, transcription factors, neuropeptides, and over 150 ion channels and receptors relevant for pain research and axonal growth/regeneration studies (e.g., TRPV1, NAV1.7, NAV1.8, TAC1, CALCA, GAP43, DPYSL2, NMNAT2). Moreover, after confirming robust functional activities and differential response to noxious stimuli and specific drugs, a robotic cell culture system was employed to produce large quantities of human sensory neurons, which can be used to develop nociceptor-selective analgesics.
Collapse
Affiliation(s)
- Tao Deng
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Vukasin M Jovanovic
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Carlos A Tristan
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Claire Weber
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Pei-Hsuan Chu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Jason Inman
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Seungmi Ryu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Yogita Jethmalani
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Juliana Ferreira de Sousa
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Pinar Ormanoglu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Prisca Twumasi
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Chaitali Sen
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Jaehoon Shim
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Selwyn Jayakar
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Sooyeon Jo
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Weifeng Yu
- Sophion Bioscience, North Brunswick, NJ 08902, USA
| | - Ty C Voss
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Anton Simeonov
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Bruce P Bean
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Ilyas Singeç
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA.
| |
Collapse
|
45
|
Liu M, He F, Shao M, Li T, Wang L, Wang Y, Xu W. PACAP inhibition alleviates neuropathic pain by modulating Nav1.7 through the MAPK/ERK signaling pathway in a rat model of chronic constriction injury. Neuropeptides 2023; 99:102327. [PMID: 36842389 DOI: 10.1016/j.npep.2023.102327] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 02/08/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023]
Abstract
BACKGROUND Trigeminal neuralgia is a common chronic maxillofacial neuropathic pain disorder, and voltage-gated sodium channels (VSGCs) are likely involved in its pathology. Prior studies report that pituitary adenylate cyclase-activating polypeptide (PACAP), a neuropeptide highly expressed in the trigeminal ganglion, may contribute to dorsal root ganglion neuron excitability by modulating the Nav1.7. OBJECTIVE We investigated whether PACAP can regulate Nav1.7 through the mitogen-activated protein kinase/ERK kinase/extracellular-signal-regulated kinase (MEK/ERK) pathway in the trigeminal ganglion after chronic constriction injury of the infraorbital nerve (ION-CCI) in rats. STUDY DESIGN Sprague-Dawley rats underwent ION-CCI, followed by intrathecal injection of PACAP 6-38 (PAC1 receptor antagonist) and PD98059 (MEK/ERK antagonist). Quantitative real-time PCR and western blot were used to quantify ATF3, PACAP, ERK, p-ERK, and Nav1.7 expression. RESULTS The mechanical pain threshold decreased from day 3 to day 21 after ION-CCI and reached the lowest testing value by day 14; however, it increased after PACAP 6-38 and PD98059 injections. Additionally, ION-CCI surgery increased ATF3, PACAP, and p-ERK expression in the rat trigeminal ganglion and decreased Nav1.7 and PAC1 receptor expression; however, there was no difference in ERK expression. PACAP 6-38 injection significantly decreased PACAP, p-ERK, and Nav1.7 expression and increased the PAC1 receptor expression, with no change in ERK expression. Moreover, PD98059 injection decreased PACAP, p-ERK, and Nav1.7 expression and increased the expression of PAC1 receptor. CONCLUSION After ION-CCI, PACAP in the rat trigeminal ganglion can modulate Nav1.7 through the MEK/ERK pathway via the PAC1 receptor. Further, PACAP inhibition alleviates allodynia in ION-CCI rats.
Collapse
Affiliation(s)
- Mingzheng Liu
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Fan He
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Mengci Shao
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Tianyuan Li
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Liecheng Wang
- School of Basic Medical Sciences, Department of Physiology, Anhui Medical University, Hefei 230032, China
| | - Yuanyin Wang
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China.
| | - Wenhua Xu
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China.
| |
Collapse
|
46
|
Shiers S, Funk G, Cervantes A, Horton P, Dussor G, Hennen S, Price TJ. Na V1.7 mRNA and protein expression in putative projection neurons of the human spinal dorsal horn. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.04.527110. [PMID: 36778234 PMCID: PMC9915702 DOI: 10.1101/2023.02.04.527110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
NaV1.7, a membrane-bound voltage-gated sodium channel, is preferentially expressed along primary sensory neurons, including their peripheral & central nerve endings, axons, and soma within the dorsal root ganglia and plays an integral role in amplifying membrane depolarization and pain neurotransmission. Loss- and gain-of-function mutations in the gene encoding NaV1.7, SCN9A, are associated with a complete loss of pain sensation or exacerbated pain in humans, respectively. As an enticing pain target supported by human genetic validation, many compounds have been developed to inhibit NaV1.7 but have disappointed in clinical trials. The underlying reasons are still unclear, but recent reports suggest that inhibiting NaV1.7 in central terminals of nociceptor afferents is critical for achieving pain relief by pharmacological inhibition of NaV1.7. We report for the first time that NaV1.7 mRNA is expressed in putative projection neurons (NK1R+) in the human spinal dorsal horn, predominantly in lamina 1 and 2, as well as in deep dorsal horn neurons and motor neurons in the ventral horn. NaV1.7 protein was found in the central axons of sensory neurons terminating in lamina 1-2, but also was detected in the axon initial segment of resident spinal dorsal horn neurons and in axons entering the anterior commissure. Given that projection neurons are critical for conveying nociceptive information from the dorsal horn to the brain, these data support that dorsal horn NaV1.7 expression may play an unappreciated role in pain phenotypes observed in humans with genetic SCN9A mutations, and in achieving analgesic efficacy in clinical trials.
Collapse
Affiliation(s)
- Stephanie Shiers
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | | | | | | | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | | | - Theodore J. Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| |
Collapse
|
47
|
Modulating the activity of human nociceptors with a SCN10A promoter-specific viral vector tool. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2023; 13:100120. [PMID: 36816616 PMCID: PMC9932673 DOI: 10.1016/j.ynpai.2023.100120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023]
Abstract
Despite the high prevalence of chronic pain as a disease in our society, there is a lack of effective treatment options for patients living with this condition. Gene therapies using recombinant AAVs are a direct method to selectively express genes of interest in target cells with the potential of, in the case of nociceptors, reducing neuronal firing in pain conditions. We designed a recombinant AAV vector expressing cargos whose expression was driven by a portion of the SCN10A (NaV1.8) promoter, which is predominantly active in nociceptors. We validated its specificity for nociceptors in mouse and human dorsal root ganglia and showed that it can drive the expression of functional proteins. Our viral vector and promoter package drove the expression of both excitatory or inhibitory DREADDs in primary human DRG cultures and in whole cell electrophysiology experiments, increased or decreased neuronal firing, respectively. Taken together, we present a novel viral tool that drives expression of cargo specifically in human nociceptors. This will allow for future specific studies of human nociceptor properties as well as pave the way for potential future gene therapies for chronic pain.
Collapse
|
48
|
Allison RL, Burand A, Torres DN, Brandow AM, Stucky CL, Ebert AD. Sickle cell disease patient plasma sensitizes iPSC-derived sensory neurons from sickle cell disease patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.10.523446. [PMID: 36711992 PMCID: PMC9882050 DOI: 10.1101/2023.01.10.523446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Individuals living with sickle cell disease (SCD) experience severe recurrent acute and chronic pain. In order to develop novel therapies, it is necessary to better understand the neurobiological mechanisms underlying SCD pain. There are many barriers to gaining mechanistic insight into pathogenic SCD pain processes, such as differential gene expression and function of sensory neurons between humans and mice with SCD, as well as the limited availability of patient samples. These can be overcome by utilizing SCD patient-derived induced pluripotent stem cells (iPSCs) differentiated into sensory neurons (SCD iSNs). Here, we characterize the key gene expression and function of SCD iSNs to establish a model for higher-throughput investigation of intrinsic and extrinsic factors that may contribute to increased SCD patient pain. Importantly, identified roles for C-C Motif Chemokine Ligand 2 (CCL2) and endothelin 1 (ET1) in SCD pain can be recapitulated in SCD iSNs. Further, we find that plasma taken from SCD patients during acute pain increases SCD iSN calcium response to the nociceptive stimulus capsaicin compared to those treated with paired SCD patient plasma at baseline or healthy control plasma samples. Together, these data provide the framework necessary to utilize iSNs as a powerful tool to investigate the neurobiology of SCD and identify potential intrinsic mechanisms of SCD pain which may extend beyond a blood-based pathology.
Collapse
Affiliation(s)
- Reilly L. Allison
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Anthony Burand
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Damaris Nieves Torres
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI
| | - Amanda M. Brandow
- Department of Pediatrics, Section of Hematology/Oncology/Bone Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI
| | - Cheryl L. Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Allison D. Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
49
|
Yang J, Ding H, Shuai B, Zhang Y, Zhang Y. Mechanism and effects of STING-IFN-I pathway on nociception: A narrative review. Front Mol Neurosci 2023; 15:1081288. [PMID: 36683857 PMCID: PMC9846240 DOI: 10.3389/fnmol.2022.1081288] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/05/2022] [Indexed: 01/05/2023] Open
Abstract
Since the discovery of STING in 2008, numerous studies have investigated its functions in immunity, inflammation, and cancer. STING activates downstream molecules including IFN-I, NLRP3, and NF-κB. The STING-IFN-I pathway plays a vital role in nociception. After receiving the upstream signal, STING is activated and induces the expression of IFN-I, and after paracrine and autocrine signaling, IFN-I binds to IFN receptors. Subsequently, the activity of ion channels is inhibited by TYK2, which induces an acute antinociceptive effect. JAK activates PIK3 and MAPK-MNK-eIF4E pathways, which sensitize nociceptors in the peripheral nervous system. In the mid-late stage, the STING-IFN-I pathway activates STAT, increases pro-inflammatory and anti-inflammatory cytokines, inhibits ER-phagy, and promotes microglial M1-polarization in the central nervous system, leading to central sensitization. Thus, the STING-IFN-I pathway may exert complex effects on nociception at various stages, and these effects require further comprehensive elucidation. Therefore, in this review, we systematically summarized the mechanisms of the STING-IFN-I pathway and discussed its function in nociception.
Collapse
Affiliation(s)
- Jinghan Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Ding
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Shuai
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Zhang
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Yan Zhang,
| | - Yan Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
50
|
Maximizing treatment efficacy through patient stratification in neuropathic pain trials. Nat Rev Neurol 2023; 19:53-64. [PMID: 36400867 DOI: 10.1038/s41582-022-00741-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2022] [Indexed: 11/19/2022]
Abstract
Treatment of neuropathic pain remains inadequate despite the elucidation of multiple pathophysiological mechanisms and the development of promising therapeutic compounds. The lack of success in translating knowledge into clinical practice has discouraged pharmaceutical companies from investing in pain medicine; however, new patient stratification approaches could help bridge the translation gap and develop individualized therapeutic approaches. As we highlight in this article, subgrouping of patients according to sensory profiles and other baseline characteristics could aid the prediction of treatment success. Furthermore, novel outcome measures have been developed for patients with neuropathic pain. The extent to which sensory profiles and outcome measures can be employed in routine clinical practice and clinical trials and across distinct neuropathic pain aetiologies is yet to be determined. Improvements in animal models, drawing on our knowledge of human pain, and robust public-private partnerships will be needed to pave the way to innovative and effective pain medicine in the future.
Collapse
|