1
|
Guo K, Lu Y, Wang X, Duan Y, Li H, Gao F, Wang J. Multi-level exploration of auricular acupuncture: from traditional Chinese medicine theory to modern medical application. Front Neurosci 2024; 18:1426618. [PMID: 39376538 PMCID: PMC11456840 DOI: 10.3389/fnins.2024.1426618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/22/2024] [Indexed: 10/09/2024] Open
Abstract
As medical research advances and technology rapidly develops, auricular acupuncture has emerged as a point of growing interest. This paper delves into the intricate anatomy of auricular points, their significance and therapeutic principles in traditional Chinese medicine (TCM), and the underlying mechanisms of auricular acupuncture in contemporary medicine. The aim is to delve deeply into this ancient and mysterious medical tradition, unveiling its multi-layered mysteries in the field of neurostimulation. The anatomical structure of auricular points is complex and delicate, and their unique neurovascular network grants them a special status in neurostimulation therapy. Through exploration of these anatomical features, we not only comprehend the position of auricular points in TCM theory but also provide a profound foundation for their modern medical applications. Through systematic review, we synthesize insights from traditional Chinese medical theory for modern medical research. Building upon anatomical and classical theoretical foundations, we focus on the mechanisms of auricular acupuncture as a unique neurostimulation therapy. This field encompasses neuroregulation, pain management, psychological wellbeing, metabolic disorders, and immune modulation. The latest clinical research not only confirms the efficacy of auricular stimulation in alleviating pain symptoms and modulating metabolic diseases at the endocrine level but also underscores its potential role in regulating patients' psychological wellbeing. This article aims to promote a comprehensive understanding of auricular acupuncture by demonstrating its diverse applications and providing substantial evidence to support its broader adoption in clinical practice.
Collapse
Affiliation(s)
- Kaixin Guo
- Department of Acupuncture, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Lu
- Department of Acupuncture, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiuping Wang
- Department of Acupuncture, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yunfeng Duan
- Department of Acupuncture, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hui Li
- Department of Acupuncture, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fengxiao Gao
- Department of Acupuncture, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jian Wang
- Department of Acupuncture, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Acupuncture and Moxibustion, The First Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
2
|
Hua T, Kong E, Zhang H, Lu J, Huang K, Ding R, Wang H, Li J, Han C, Yuan H. PRMT6 deficiency or inhibition alleviates neuropathic pain by decreasing glycolysis and inflammation in microglia. Brain Behav Immun 2024; 118:101-114. [PMID: 38402915 DOI: 10.1016/j.bbi.2024.02.027] [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: 09/04/2023] [Revised: 02/16/2024] [Accepted: 02/23/2024] [Indexed: 02/27/2024] Open
Abstract
Microglia induced chronic inflammation is the critical pathology of Neuropathic pain (NP). Metabolic reprogramming of macrophage has been intensively reported in various chronic inflammation diseases. However, the metabolic reprogramming of microglia in chronic pain remains to be elusive. Here, we reported that immuno-metabolic markers (HIF-1α, PKM2, GLUT1 and lactate) were related with increased expression of PRMT6 in the ipsilateral spinal cord dorsal horn of the chronic construction injury (CCI) mice. PRMT6 deficiency or prophylactic and therapeutic intrathecal administration of PRMT6 inhibitor (EPZ020411) ameliorated CCI-induced NP, inflammation and glycolysis in the ipsilateral spinal cord dorsal horn. PRMT6 knockout or knockdown inhibited LPS-induced inflammation, proliferation and glycolysis in microglia cells. While PRMT6 overexpression exacerbated LPS-induced inflammation, proliferation and glycolysis in BV2 cells. Recent research revealed that PRMT6 could interact with and methylate HIF-1α, which increased HIF-1α protein stability. In sum, increased expression of PRMT6 exacerbates NP progress by increasing glycolysis and neuroinflammation through interacting with and stabilizing HIF-1α in a methyltransferase manner, which outlines novel pathological mechanism and drug target for NP.
Collapse
Affiliation(s)
- Tong Hua
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Erliang Kong
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, Shanghai, China; Department of Anesthesiology, The No. 988 Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Zhengzhou, China
| | - Hailing Zhang
- Department of Neurology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jinfang Lu
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Kesheng Huang
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ruifeng Ding
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Haowei Wang
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jian Li
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chaofeng Han
- Department of Histology and Embryology, and Shanghai Key Laboratory of Cell Engineering, Naval Medical University, Shanghai, China.
| | - Hongbin Yuan
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
3
|
Malapert P, Robert G, Brunet E, Chemin J, Bourinet E, Moqrich A. A novel Na v1.8-FLPo driver mouse for intersectional genetics to uncover the functional significance of primary sensory neuron diversity. iScience 2024; 27:109396. [PMID: 38510134 PMCID: PMC10952036 DOI: 10.1016/j.isci.2024.109396] [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: 08/18/2023] [Revised: 11/08/2023] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
The recent development of single-cell and single-nucleus RNA sequencing has highlighted the extraordinary diversity of dorsal root ganglia neurons. However, the few available genetic tools limit our understanding of the functional significance of this heterogeneity. We generated a new mouse line expressing the flippase recombinase from the scn10a locus. By crossing Nav1.8Ires-FLPo mice with the AdvillinCre and RC::FL-hM3Dq mouse lines in an intersectional genetics approach, we were able to obtain somatodendritic expression of hM3Dq-mCherry selectively in the Nav1.8 lineage. The bath application of clozapine N-oxide triggered strong calcium responses selectively in mCherry+ neurons. The intraplantar injection of CNO caused robust flinching, shaking, and biting responses accompanied by strong cFos activation in the ipsilateral lumbar spinal cord. The Nav1.8Ires-FLPo mouse model will be a valuable tool for extending our understanding of the in vivo functional specialization of neuronal subsets of the Nav1.8 lineage for which inducible Cre lines are available.
Collapse
Affiliation(s)
- Pascale Malapert
- Aix-Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, Marseille, France
| | - Guillaume Robert
- Aix-Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, Marseille, France
| | - Elena Brunet
- Aix-Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, Marseille, France
| | - Jean Chemin
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Emmanuel Bourinet
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Aziz Moqrich
- Aix-Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, Marseille, France
| |
Collapse
|
4
|
Lavaud S, Bichara C, D'Andola M, Yeh SH, Takeoka A. Two inhibitory neuronal classes govern acquisition and recall of spinal sensorimotor adaptation. Science 2024; 384:194-201. [PMID: 38603479 DOI: 10.1126/science.adf6801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 03/06/2024] [Indexed: 04/13/2024]
Abstract
Spinal circuits are central to movement adaptation, yet the mechanisms within the spinal cord responsible for acquiring and retaining behavior upon experience remain unclear. Using a simple conditioning paradigm, we found that dorsal inhibitory neurons are indispensable for adapting protective limb-withdrawal behavior by regulating the transmission of a specific set of somatosensory information to enhance the saliency of conditioning cues associated with limb position. By contrast, maintaining previously acquired motor adaptation required the ventral inhibitory Renshaw cells. Manipulating Renshaw cells does not affect the adaptation itself but flexibly alters the expression of adaptive behavior. These findings identify a circuit basis involving two distinct populations of spinal inhibitory neurons, which enables lasting sensorimotor adaptation independently from the brain.
Collapse
Affiliation(s)
- Simon Lavaud
- VIB-Neuroelectronics Research Flanders (NERF), 3001 Leuven, Belgium
- KU Leuven, Department of Neuroscience and Leuven Brain Institute, 3000 Leuven, Belgium
| | - Charlotte Bichara
- VIB-Neuroelectronics Research Flanders (NERF), 3001 Leuven, Belgium
- KU Leuven, Department of Neuroscience and Leuven Brain Institute, 3000 Leuven, Belgium
| | - Mattia D'Andola
- VIB-Neuroelectronics Research Flanders (NERF), 3001 Leuven, Belgium
- KU Leuven, Department of Neuroscience and Leuven Brain Institute, 3000 Leuven, Belgium
| | - Shu-Hao Yeh
- VIB-Neuroelectronics Research Flanders (NERF), 3001 Leuven, Belgium
- KU Leuven, Department of Neuroscience and Leuven Brain Institute, 3000 Leuven, Belgium
| | - Aya Takeoka
- VIB-Neuroelectronics Research Flanders (NERF), 3001 Leuven, Belgium
- KU Leuven, Department of Neuroscience and Leuven Brain Institute, 3000 Leuven, Belgium
- IMEC, 3001 Leuven, Belgium
- RIKEN Center for Brain Science, Laboratory for Motor Circuit Plasticity, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| |
Collapse
|
5
|
Gautam M, Yamada A, Yamada AI, Wu Q, Kridsada K, Ling J, Yu H, Dong P, Ma M, Gu J, Luo W. Distinct local and global functions of mouse Aβ low-threshold mechanoreceptors in mechanical nociception. Nat Commun 2024; 15:2911. [PMID: 38575590 PMCID: PMC10995180 DOI: 10.1038/s41467-024-47245-0] [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: 05/15/2023] [Accepted: 03/13/2024] [Indexed: 04/06/2024] Open
Abstract
The roles of Aβ low-threshold mechanoreceptors (LTMRs) in transmitting mechanical hyperalgesia and in alleviating chronic pain have been of great interest but remain contentious. Here we utilized intersectional genetic tools, optogenetics, and high-speed imaging to specifically examine functions of SplitCre labeled mouse Aβ-LTMRs in this regard. Genetic ablation of SplitCre-Aβ-LTMRs increased mechanical nociception but not thermosensation in both acute and chronic inflammatory pain conditions, indicating a modality-specific role in gating mechanical nociception. Local optogenetic activation of SplitCre-Aβ-LTMRs triggered nociception after tissue inflammation, whereas their broad activation at the dorsal column still alleviated mechanical hypersensitivity of chronic inflammation. Taking all data into consideration, we propose a model, in which Aβ-LTMRs play distinctive local and global roles in transmitting or alleviating mechanical hyperalgesia of chronic pain, respectively. Our model suggests a strategy of global activation plus local inhibition of Aβ-LTMRs for treating mechanical hyperalgesia.
Collapse
Affiliation(s)
- Mayank Gautam
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Akihiro Yamada
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Ayaka I Yamada
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Qinxue Wu
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kim Kridsada
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jennifer Ling
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Huasheng Yu
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Peter Dong
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Minghong Ma
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jianguo Gu
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Wenqin Luo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
6
|
Rankin G, Chirila AM, Emanuel AJ, Zhang Z, Woolf CJ, Drugowitsch J, Ginty DD. Nerve injury disrupts temporal processing in the spinal cord dorsal horn through alterations in PV + interneurons. Cell Rep 2024; 43:113718. [PMID: 38294904 PMCID: PMC11101906 DOI: 10.1016/j.celrep.2024.113718] [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: 03/15/2023] [Revised: 11/13/2023] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
How mechanical allodynia following nerve injury is encoded in patterns of neural activity in the spinal cord dorsal horn (DH) remains incompletely understood. We address this in mice using the spared nerve injury model of neuropathic pain and in vivo electrophysiological recordings. Surprisingly, despite dramatic behavioral over-reactivity to mechanical stimuli following nerve injury, an overall increase in sensitivity or reactivity of DH neurons is not observed. We do, however, observe a marked decrease in correlated neural firing patterns, including the synchrony of mechanical stimulus-evoked firing, across the DH. Alterations in DH temporal firing patterns are recapitulated by silencing DH parvalbumin+ (PV+) interneurons, previously implicated in mechanical allodynia, as are allodynic pain-like behaviors. These findings reveal decorrelated DH network activity, driven by alterations in PV+ interneurons, as a prominent feature of neuropathic pain and suggest restoration of proper temporal activity as a potential therapeutic strategy to treat chronic neuropathic pain.
Collapse
Affiliation(s)
- Genelle Rankin
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Anda M Chirila
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Alan J Emanuel
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Zihe Zhang
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Clifford J Woolf
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jan Drugowitsch
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - David D Ginty
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
7
|
Gilbert JE, Zhang T, Esteller R, Grill WM. Network model of nociceptive processing in the superficial spinal dorsal horn reveals mechanisms of hyperalgesia, allodynia, and spinal cord stimulation. J Neurophysiol 2023; 130:1103-1117. [PMID: 37727912 DOI: 10.1152/jn.00186.2023] [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: 05/09/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/21/2023] Open
Abstract
The spinal dorsal horn (DH) processes sensory information and plays a key role in transmitting nociception to supraspinal centers. Loss of DH inhibition during neuropathic pain unmasks a pathway from nonnociceptive Aβ-afferent inputs to superficial dorsal horn (SDH) nociceptive-specific (NS) projection neurons, and this change may contribute to hyperalgesia and allodynia. We developed and validated a computational model of SDH neuronal circuitry that links nonnociceptive Aβ-afferent inputs in lamina II/III to a NS projection neuron in lamina I via a network of excitatory interneurons. The excitatory pathway and the NS projection neuron were in turn gated by inhibitory interneurons with connections based on prior patch-clamp recordings. Changing synaptic weights in the computational model to replicate neuropathic pain states unmasked a low-threshold excitatory pathway to NS neurons similar to experimental recordings. Spinal cord stimulation (SCS) is an effective therapy for neuropathic pain, and accumulating experimental evidence indicates that NS neurons in the SDH also respond to SCS. Accounting for these responses may inform therapeutic improvements, and we quantified responses to SCS in the SDH network model and examined the role of different modes of inhibitory control in modulating NS neuron responses to SCS. We combined the SDH network model with a previously published model of the deep dorsal horn (DDH) and identified optimal stimulation frequencies across different neuropathic pain conditions. Finally, we found that SCS-generated inhibition did not completely suppress model NS activity during simulated pinch inputs, providing an explanation of why SCS does not eliminate acute pain.NEW & NOTEWORTHY Chronic pain is a severe public health problem that reduces the quality of life for those affected and exacts an enormous socio-economic burden worldwide. Spinal cord stimulation (SCS) is an effective treatment for chronic pain, but SCS efficacy has not significantly improved over time, in part because the mechanisms of action remain unclear. Most preclinical studies investigating pain and SCS mechanisms have focused on the responses of deep dorsal horn (DDH) neurons, but neural networks in the superficial dorsal horn (SDH) are also important for processing nociceptive information. This work synthesizes heterogeneous experimental recordings from the SDH into a computational model that replicates experimental responses and that can be used to quantify neuronal responses to SCS under neuropathic pain conditions.
Collapse
Affiliation(s)
- John E Gilbert
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States
| | - Tianhe Zhang
- Neuromodulation Research and Advanced Concepts, Boston Scientific Neuromodulation, Valencia, California, United States
| | - Rosana Esteller
- Neuromodulation Research and Advanced Concepts, Boston Scientific Neuromodulation, Valencia, California, United States
| | - Warren M Grill
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States
- Department of Electrical and Computer Engineering, Duke University, Durham, North Carolina, United States
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, United States
| |
Collapse
|
8
|
Du F, Yin G, Han L, Liu X, Dong D, Duan K, Huo J, Sun Y, Cheng L. Targeting Peripheral μ-opioid Receptors or μ-opioid Receptor-Expressing Neurons Does not Prevent Morphine-induced Mechanical Allodynia and Anti-allodynic Tolerance. Neurosci Bull 2023; 39:1210-1228. [PMID: 36622575 PMCID: PMC10387027 DOI: 10.1007/s12264-022-01009-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/19/2022] [Indexed: 01/10/2023] Open
Abstract
The chronic use of morphine and other opioids is associated with opioid-induced hypersensitivity (OIH) and analgesic tolerance. Among the different forms of OIH and tolerance, the opioid receptors and cell types mediating opioid-induced mechanical allodynia and anti-allodynic tolerance remain unresolved. Here we demonstrated that the loss of peripheral μ-opioid receptors (MORs) or MOR-expressing neurons attenuated thermal tolerance, but did not affect the expression and maintenance of morphine-induced mechanical allodynia and anti-allodynic tolerance. To confirm this result, we made dorsal root ganglia-dorsal roots-sagittal spinal cord slice preparations and recorded low-threshold Aβ-fiber stimulation-evoked inputs and outputs in superficial dorsal horn neurons. Consistent with the behavioral results, peripheral MOR loss did not prevent the opening of Aβ mechanical allodynia pathways in the spinal dorsal horn. Therefore, the peripheral MOR signaling pathway may not be an optimal target for preventing mechanical OIH and analgesic tolerance. Future studies should focus more on central mechanisms.
Collapse
Affiliation(s)
- Feng Du
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Guangjuan Yin
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Lei Han
- Department of Anesthesiology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, China
| | - Xi Liu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Dong Dong
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Kaifang Duan
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jiantao Huo
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yanyan Sun
- Department of Anesthesiology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, China.
| | - Longzhen Cheng
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China.
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
9
|
Bohic M, Upadhyay A, Eisdorfer JT, Keating J, Simon RC, Briones BA, Azadegan C, Nacht HD, Oputa O, Martinez AM, Bethell BN, Gradwell MA, Romanienko P, Ramer MS, Stuber GD, Abraira VE. A new Hoxb8FlpO mouse line for intersectional approaches to dissect developmentally defined adult sensorimotor circuits. Front Mol Neurosci 2023; 16:1176823. [PMID: 37603775 PMCID: PMC10437123 DOI: 10.3389/fnmol.2023.1176823] [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: 02/28/2023] [Accepted: 07/04/2023] [Indexed: 08/23/2023] Open
Abstract
Improvements in the speed and cost of expression profiling of neuronal tissues offer an unprecedented opportunity to define ever finer subgroups of neurons for functional studies. In the spinal cord, single cell RNA sequencing studies support decades of work on spinal cord lineage studies, offering a unique opportunity to probe adult function based on developmental lineage. While Cre/Flp recombinase intersectional strategies remain a powerful tool to manipulate spinal neurons, the field lacks genetic tools and strategies to restrict manipulations to the adult mouse spinal cord at the speed at which new tools develop. This study establishes a new workflow for intersectional mouse-viral strategies to dissect adult spinal function based on developmental lineages in a modular fashion. To restrict manipulations to the spinal cord, we generate a brain-sparing Hoxb8FlpO mouse line restricting Flp recombinase expression to caudal tissue. Recapitulating endogenous Hoxb8 gene expression, Flp-dependent reporter expression is present in the caudal embryo starting day 9.5. This expression restricts Flp activity in the adult to the caudal brainstem and below. Hoxb8FlpO heterozygous and homozygous mice do not develop any of the sensory or locomotor phenotypes evident in Hoxb8 heterozygous or mutant animals, suggesting normal developmental function of the Hoxb8 gene and protein in Hoxb8FlpO mice. Compared to the variability of brain recombination in available caudal Cre and Flp lines, Hoxb8FlpO activity is not present in the brain above the caudal brainstem, independent of mouse genetic background. Lastly, we combine the Hoxb8FlpO mouse line with dorsal horn developmental lineage Cre mouse lines to express GFP in developmentally determined dorsal horn populations. Using GFP-dependent Cre recombinase viruses and Cre recombinase-dependent inhibitory chemogenetics, we target developmentally defined lineages in the adult. We show how developmental knock-out versus transient adult silencing of the same ROR𝛃 lineage neurons affects adult sensorimotor behavior. In summary, this new mouse line and viral approach provides a blueprint to dissect adult somatosensory circuit function using Cre/Flp genetic tools to target spinal cord interneurons based on genetic lineage.
Collapse
Affiliation(s)
- Manon Bohic
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Aman Upadhyay
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- Neuroscience PhD Program at Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Jaclyn T. Eisdorfer
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Jessica Keating
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- School of Medicine, Oregon Health and Science University, Portland, OR, United States
- M.D./PhD Program in Neuroscience, School of Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Rhiana C. Simon
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Brandy A. Briones
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Chloe Azadegan
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Hannah D. Nacht
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Olisemeka Oputa
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Alana M. Martinez
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Bridget N. Bethell
- International Collaboration on Repair Discoveries and Department of Zoology, The University of British Columbia, Vancouver, BC, Canada
| | - Mark A. Gradwell
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Peter Romanienko
- Genome Editing Shared Resource, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Matt S. Ramer
- International Collaboration on Repair Discoveries and Department of Zoology, The University of British Columbia, Vancouver, BC, Canada
| | - Garret D. Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Victoria E. Abraira
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| |
Collapse
|
10
|
Ma Q, Su D, Huo J, Yin G, Dong D, Duan K, Cheng H, Xu H, Ma J, Liu D, Mou B, Peng J, Cheng L. Microglial Depletion does not Affect the Laterality of Mechanical Allodynia in Mice. Neurosci Bull 2023; 39:1229-1245. [PMID: 36637789 PMCID: PMC10387012 DOI: 10.1007/s12264-022-01017-2] [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: 07/31/2022] [Accepted: 10/12/2022] [Indexed: 01/14/2023] Open
Abstract
Mechanical allodynia (MA), including punctate and dynamic forms, is a common and debilitating symptom suffered by millions of chronic pain patients. Some peripheral injuries result in the development of bilateral MA, while most injuries usually led to unilateral MA. To date, the control of such laterality remains poorly understood. Here, to study the role of microglia in the control of MA laterality, we used genetic strategies to deplete microglia and tested both dynamic and punctate forms of MA in mice. Surprisingly, the depletion of central microglia did not prevent the induction of bilateral dynamic and punctate MA. Moreover, in dorsal root ganglion-dorsal root-sagittal spinal cord slice preparations we recorded the low-threshold Aβ-fiber stimulation-evoked inputs and outputs of superficial dorsal horn neurons. Consistent with behavioral results, microglial depletion did not prevent the opening of bilateral gates for Aβ pathways in the superficial dorsal horn. This study challenges the role of microglia in the control of MA laterality in mice. Future studies are needed to further understand whether the role of microglia in the control of MA laterality is etiology-or species-specific.
Collapse
Affiliation(s)
- Quan Ma
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Dongmei Su
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jiantao Huo
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Guangjuan Yin
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Dong Dong
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Kaifang Duan
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Hong Cheng
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Huiling Xu
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jiao Ma
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Dong Liu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Bin Mou
- Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Jiyun Peng
- Institute of Life Science, Nanchang University, Nanchang, 330031, China.
| | - Longzhen Cheng
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China.
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China.
| |
Collapse
|
11
|
Gautam M, Yamada A, Yamada A, Wu Q, Kridsada K, Ling J, Yu H, Dong P, Ma M, Gu J, Luo W. Distinct Local and Global Functions of Aβ Low-Threshold Mechanoreceptors in Mechanical Pain Transmission. RESEARCH SQUARE 2023:rs.3.rs-2939309. [PMID: 37398333 PMCID: PMC10312941 DOI: 10.21203/rs.3.rs-2939309/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: 07/04/2023]
Abstract
The roles of Aβ low-threshold mechanoreceptors (LTMRs) in transmitting mechanical hyperalgesia and in alleviating chronic pain have been of great interest but remain contentious. Here we utilized intersectional genetic tools, optogenetics, and high-speed imaging to specifically examine functions of SplitCre labeled Aβ-LTMRs in this regard. Genetic ablation of SplitCre-Aβ-LTMRs increased mechanical pain but not thermosensation in both acute and chronic inflammatory pain conditions, indicating their modality-specific role in gating mechanical pain transmission. Local optogenetic activation of SplitCre-Aβ-LTMRs triggered nociception after tissue inflammation, whereas their broad activation at the dorsal column still alleviated mechanical hypersensitivity of chronic inflammation. Taking all data into consideration, we propose a new model, in which Aβ-LTMRs play distinctive local and global roles in transmitting and alleviating mechanical hyperalgesia of chronic pain, respectively. Our model suggests a new strategy of global activation plus local inhibition of Aβ-LTMRs for treating mechanical hyperalgesia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Minghong Ma
- University of Pennsylvania School of Medicine
| | | | | |
Collapse
|
12
|
Gautam M, Yamada A, Yamada AI, Wu Q, Kridsada K, Ling J, Yu H, Dong P, Ma M, Gu J, Luo W. Distinct Local and Global Functions of Aβ Low-Threshold Mechanoreceptors in Mechanical Pain Transmission. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.16.540962. [PMID: 37293085 PMCID: PMC10245756 DOI: 10.1101/2023.05.16.540962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The roles of Aβ low-threshold mechanoreceptors (LTMRs) in transmitting mechanical hyperalgesia and in alleviating chronic pain have been of great interest but remain contentious. Here we utilized intersectional genetic tools, optogenetics, and high-speed imaging to specifically examine functions of Split Cre labeled Aβ-LTMRs in this regard. Genetic ablation of Split Cre -Aβ-LTMRs increased mechanical pain but not thermosensation in both acute and chronic inflammatory pain conditions, indicating their modality-specific role in gating mechanical pain transmission. Local optogenetic activation of Split Cre -Aβ-LTMRs triggered nociception after tissue inflammation, whereas their broad activation at the dorsal column still alleviated mechanical hypersensitivity of chronic inflammation. Taking all data into consideration, we propose a new model, in which Aβ-LTMRs play distinctive local and global roles in transmitting and alleviating mechanical hyperalgesia of chronic pain, respectively. Our model suggests a new strategy of global activation plus local inhibition of Aβ-LTMRs for treating mechanical hyperalgesia.
Collapse
|
13
|
Wilson AC, Sweeney LB. Spinal cords: Symphonies of interneurons across species. Front Neural Circuits 2023; 17:1146449. [PMID: 37180760 PMCID: PMC10169611 DOI: 10.3389/fncir.2023.1146449] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/23/2023] [Indexed: 05/16/2023] Open
Abstract
Vertebrate movement is orchestrated by spinal inter- and motor neurons that, together with sensory and cognitive input, produce dynamic motor behaviors. These behaviors vary from the simple undulatory swimming of fish and larval aquatic species to the highly coordinated running, reaching and grasping of mice, humans and other mammals. This variation raises the fundamental question of how spinal circuits have changed in register with motor behavior. In simple, undulatory fish, exemplified by the lamprey, two broad classes of interneurons shape motor neuron output: ipsilateral-projecting excitatory neurons, and commissural-projecting inhibitory neurons. An additional class of ipsilateral inhibitory neurons is required to generate escape swim behavior in larval zebrafish and tadpoles. In limbed vertebrates, a more complex spinal neuron composition is observed. In this review, we provide evidence that movement elaboration correlates with an increase and specialization of these three basic interneuron types into molecularly, anatomically, and functionally distinct subpopulations. We summarize recent work linking neuron types to movement-pattern generation across fish, amphibians, reptiles, birds and mammals.
Collapse
Affiliation(s)
| | - Lora B. Sweeney
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Lower Austria, Austria
| |
Collapse
|
14
|
Huo J, Du F, Duan K, Yin G, Liu X, Ma Q, Dong D, Sun M, Hao M, Su D, Huang T, Ke J, Lai S, Zhang Z, Guo C, Sun Y, Cheng L. Identification of brain-to-spinal circuits controlling the laterality and duration of mechanical allodynia in mice. Cell Rep 2023; 42:112300. [PMID: 36952340 DOI: 10.1016/j.celrep.2023.112300] [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: 06/09/2021] [Revised: 12/22/2022] [Accepted: 03/07/2023] [Indexed: 03/24/2023] Open
Abstract
Mechanical allodynia (MA) represents one prevalent symptom of chronic pain. Previously we and others have identified spinal and brain circuits that transmit or modulate the initial establishment of MA. However, brain-derived descending pathways that control the laterality and duration of MA are still poorly understood. Here we report that the contralateral brain-to-spinal circuits, from Oprm1 neurons in the lateral parabrachial nucleus (lPBNOprm1), via Pdyn neurons in the dorsal medial regions of hypothalamus (dmHPdyn), to the spinal dorsal horn (SDH), act to prevent nerve injury from inducing contralateral MA and reduce the duration of bilateral MA induced by capsaicin. Ablating/silencing dmH-projecting lPBNOprm1 neurons or SDH-projecting dmHPdyn neurons, deleting Dyn peptide from dmH, or blocking spinal κ-opioid receptors all led to long-lasting bilateral MA. Conversely, activation of dmHPdyn neurons or their axonal terminals in SDH can suppress sustained bilateral MA induced by lPBN lesion.
Collapse
Affiliation(s)
- Jiantao Huo
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Feng Du
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Kaifang Duan
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Guangjuan Yin
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xi Liu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Quan Ma
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Dong Dong
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mengge Sun
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mei Hao
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Dongmei Su
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Tianwen Huang
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Jin Ke
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Shishi Lai
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Zhi Zhang
- Division of Life Sciences and Medicine, CAS Key Laboratory of Brain Function and Diseases, University of Science and Technology of China, Hefei 230027, China
| | - Chao Guo
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yuanjie Sun
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Longzhen Cheng
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen 518055, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China.
| |
Collapse
|
15
|
Rankin G, Chirila AM, Emanuel AJ, Zhang Z, Woolf CJ, Drugowitsch J, Ginty DD. Nerve injury disrupts temporal processing in the spinal cord dorsal horn through alterations in PV + interneurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.20.533541. [PMID: 36993199 PMCID: PMC10055222 DOI: 10.1101/2023.03.20.533541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
How mechanical allodynia following nerve injury is encoded in patterns of neural activity in the spinal cord dorsal horn (DH) is not known. We addressed this using the spared nerve injury model of neuropathic pain and in vivo electrophysiological recordings. Surprisingly, despite dramatic behavioral over-reactivity to mechanical stimuli following nerve injury, an overall increase in sensitivity or reactivity of DH neurons was not observed. We did, however, observe a marked decrease in correlated neural firing patterns, including the synchrony of mechanical stimulus-evoked firing, across the DH. Alterations in DH temporal firing patterns were recapitulated by silencing DH parvalbumin + (PV + ) inhibitory interneurons, previously implicated in mechanical allodynia, as were allodynic pain-like behaviors in mice. These findings reveal decorrelated DH network activity, driven by alterations in PV + interneurons, as a prominent feature of neuropathic pain, and suggest that restoration of proper temporal activity is a potential treatment for chronic neuropathic pain.
Collapse
|
16
|
Ca 2+-Permeable AMPA Receptors Contribute to Changed Dorsal Horn Neuronal Firing and Inflammatory Pain. Int J Mol Sci 2023; 24:ijms24032341. [PMID: 36768663 PMCID: PMC9916706 DOI: 10.3390/ijms24032341] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/26/2023] Open
Abstract
The dorsal horn (DH) neurons of the spinal cord play a critical role in nociceptive input integration and processing in the central nervous system. Engaged neuronal classes and cell-specific excitability shape nociceptive computation within the DH. The DH hyperexcitability (central sensitisation) has been considered a fundamental mechanism in mediating nociceptive hypersensitivity, with the proven role of Ca2+-permeable AMPA receptors (AMPARs). However, whether and how the DH hyperexcitability relates to changes in action potential (AP) parameters in DH neurons and if Ca2+-permeable AMPARs contribute to these changes remain unknown. We examined the cell-class heterogeneity of APs generated by DH neurons in inflammatory pain conditions to address these. Inflammatory-induced peripheral hypersensitivity increased DH neuronal excitability. We found changes in the AP threshold and amplitude but not kinetics (spike waveform) in DH neurons generating sustained or initial bursts of firing patterns. In contrast, there were no changes in AP parameters in the DH neurons displaying a single spike firing pattern. Genetic knockdown of the molecular mechanism responsible for the upregulation of Ca2+-permeable AMPARs allowed the recovery of cell-specific AP changes in peripheral inflammation. Selective inhibition of Ca2+-permeable AMPARs in the spinal cord alleviated nociceptive hypersensitivity, both thermal and mechanical modalities, in animals with peripheral inflammation. Thus, Ca2+-permeable AMPARs contribute to shaping APs in DH neurons and nociceptive hypersensitivity. This may represent a neuropathological mechanism in the DH circuits, leading to aberrant signal transfer to other nociceptive pathways.
Collapse
|
17
|
Ishibashi T, Yoshikawa Y, Sueto D, Tashima R, Tozaki-Saitoh H, Koga K, Yamaura K, Tsuda M. Selective Involvement of a Subset of Spinal Dorsal Horn Neurons Operated by a Prodynorphin Promoter in Aβ Fiber-Mediated Neuropathic Allodynia-Like Behavioral Responses in Rats. Front Mol Neurosci 2022; 15:911122. [PMID: 35813063 PMCID: PMC9260077 DOI: 10.3389/fnmol.2022.911122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022] Open
Abstract
Mechanical allodynia (pain produced by innocuous stimuli such as touch) is the main symptom of neuropathic pain. Its underlying mechanism remains to be elucidated, but peripheral nerve injury (PNI)-induced malfunction of neuronal circuits in the central nervous system, including the spinal dorsal horn (SDH), is thought to be involved in touch-pain conversion. Here, we found that intra-SDH injection of adeno-associated viral vectors including a prodynorphin promoter (AAV-PdynP) captured a subset of neurons that were mainly located in the superficial laminae, including lamina I, and exhibited mostly inhibitory characteristics. Using transgenic rats that enable optogenetic stimulation of touch-sensing Aβ fibers, we found that the light-evoked paw withdrawal behavior and aversive responses after PNI were attenuated by selective ablation of AAV-PdynP-captured SDH neurons. Notably, the ablation had no effect on withdrawal behavior from von Frey filaments. Furthermore, Aβ fiber stimulation did not excite AAV-PdynP+ SDH neurons under normal conditions, but after PNI, this induced excitation, possibly due to enhanced Aβ fiber-evoked excitatory synaptic inputs and elevated resting membrane potentials of these neurons. Moreover, the chemogenetic silencing of AAV-PdynP+ neurons of PNI rats attenuated the Aβ fiber-evoked paw withdrawal behavior and c-FOS expression in superficial SDH neurons. Our findings suggest that PNI renders AAV-PdynP-captured neurons excitable to Aβ fiber stimulation, which selectively contributes to the conversion of Aβ fiber-mediated touch signal to nociceptive. Thus, reducing the excitability of AAV-PdynP-captured neurons may be a new option for the treatment of neuropathic allodynia.
Collapse
Affiliation(s)
- Tadayuki Ishibashi
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yu Yoshikawa
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Daichi Sueto
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryoichi Tashima
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Hidetoshi Tozaki-Saitoh
- Department of Pharmaceutical Sciences, International University of Health and Welfare, Fukuoka, Japan
| | - Keisuke Koga
- Department of Neurophysiology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Ken Yamaura
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
- Kyushu University Institute for Advanced Study, Fukuoka, Japan
- *Correspondence: Makoto Tsuda
| |
Collapse
|
18
|
Zhi YR, Cao F, Su XJ, Gao SW, Zheng HN, Jiang JY, Su L, Liu J, Wang Y, Zhang Y, Zhang Y. The T-Type Calcium Channel Cav3.2 in Somatostatin Interneurons in Spinal Dorsal Horn Participates in Mechanosensation and Mechanical Allodynia in Mice. Front Cell Neurosci 2022; 16:875726. [PMID: 35465611 PMCID: PMC9024096 DOI: 10.3389/fncel.2022.875726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Somatostatin-positive (SOM+) neurons have been proposed as one of the key populations of excitatory interneurons in the spinal dorsal horn involved in mechanical pain. However, the molecular mechanism for their role in pain modulation remains unknown. Here, we showed that the T-type calcium channel Cav3.2 was highly expressed in spinal SOM+ interneurons. Colocalization of Cacna1h (which codes for Cav3.2) and SOMtdTomato was observed in the in situ hybridization studies. Fluorescence-activated cell sorting of SOMtdTomato cells in spinal dorsal horn also proved a high expression of Cacna1h in SOM+ neurons. Behaviorally, virus-mediated knockdown of Cacna1h in spinal SOM+ neurons reduced the sensitivity to light touch and responsiveness to noxious mechanical stimuli in naïve mice. Furthermore, knockdown of Cacna1h in spinal SOM+ neurons attenuated thermal hyperalgesia and dynamic allodynia in the complete Freund’s adjuvant-induced inflammatory pain model, and reduced both dynamic and static allodynia in a neuropathic pain model of spared nerve injury. Mechanistically, a decrease in the percentage of neurons with Aβ-eEPSCs and Aβ-eAPs in superficial dorsal horn was observed after Cacna1h knockdown in spinal SOM+ neurons. Altogether, our results proved a crucial role of Cav3.2 in spinal SOM+ neurons in mechanosensation under basal conditions and in mechanical allodynia under pathological pain conditions. This work reveals a molecular basis for SOM+ neurons in transmitting mechanical pain and shows a functional role of Cav3.2 in tactile and pain processing at the level of spinal cord in addition to its well-established peripheral role.
Collapse
Affiliation(s)
- Yu-Ru Zhi
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, Peking University, Beijing, China
| | - Feng Cao
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, Peking University, Beijing, China
| | - Xiao-Jing Su
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shu-Wen Gao
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, Peking University, Beijing, China
| | - Hao-Nan Zheng
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Jin-Yan Jiang
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, Peking University, Beijing, China
| | - Li Su
- Center of Medical and Health Analysis, Peking University Health Science Center, Beijing, China
| | - Jiao Liu
- Center of Medical and Health Analysis, Peking University Health Science Center, Beijing, China
| | - Yun Wang
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Yan Zhang
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Ying Zhang,
| | - Ying Zhang
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, Peking University, Beijing, China
- Yan Zhang,
| |
Collapse
|
19
|
Glutamate in primary afferents is required for itch transmission. Neuron 2022; 110:809-823.e5. [PMID: 34986325 PMCID: PMC8898340 DOI: 10.1016/j.neuron.2021.12.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/21/2021] [Accepted: 12/06/2021] [Indexed: 12/13/2022]
Abstract
Whether glutamate or itch-selective neurotransmitters are used to confer itch specificity is still under debate. We focused on an itch-selective population of primary afferents expressing MRGPRA3, which highly expresses Vglut2 and the neuropeptide neuromedin B (Nmb), to investigate this question. Optogenetic stimulation of MRGPRA3+ afferents triggers scratching and other itch-related avoidance behaviors. Using a combination of optogenetics, spinal cord slice recordings, Vglut2 conditional knockout mice, and behavior assays, we showed that glutamate is essential for MRGPRA3+ afferents to transmit itch. We further demonstrated that MRGPRA3+ afferents form monosynaptic connections with both NMBR+ and NMBR- neurons and that NMB and glutamate together can enhance the activity of NMBR+ spinal DH neurons. Moreover, Nmb in MRGPRA3+ afferents and NMBR+ DH neurons are required for chloroquine-induced scratching. Together, our results establish a new model in which glutamate is an essential neurotransmitter in primary afferents for itch transmission, whereas NMB signaling enhances its activities.
Collapse
|
20
|
Toussaint AB, Foster W, Jones JM, Kaufmann S, Wachira M, Hughes R, Bongiovanni AR, Famularo ST, Dunham BP, Schwark R, Karbalaei R, Dressler C, Bavley CC, Fried NT, Wimmer ME, Abdus-Saboor I. Chronic paternal morphine exposure increases sensitivity to morphine-derived pain relief in male progeny. SCIENCE ADVANCES 2022; 8:eabk2425. [PMID: 35171664 PMCID: PMC8849295 DOI: 10.1126/sciadv.abk2425] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Parental history of opioid exposure is seldom considered when prescribing opioids for pain relief. To explore whether parental opioid exposure may affect sensitivity to morphine in offspring, we developed a "rat pain scale" with high-speed imaging, machine learning, and mathematical modeling in a multigenerational model of paternal morphine self-administration. We find that the most commonly used tool to measure mechanical sensitivity in rodents, the von Frey hair, is not painful in rats during baseline conditions. We also find that male progeny of morphine-treated sires had no baseline changes in mechanical pain sensitivity but were more sensitive to the pain-relieving effects of morphine. Using RNA sequencing across pain-relevant brain regions, we identify gene expression changes within the regulator of G protein signaling family of proteins that may underlie this multigenerational phenotype. Together, this rat pain scale revealed that paternal opioid exposure increases sensitivity to morphine's pain-relieving effects in male offspring.
Collapse
Affiliation(s)
- Andre B. Toussaint
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - William Foster
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Jessica M. Jones
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Samuel Kaufmann
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Meghan Wachira
- Department of Biology, Rutgers Camden University, Camden, NJ, USA
| | - Robert Hughes
- Department of Biology, Rutgers Camden University, Camden, NJ, USA
| | - Angela R. Bongiovanni
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Sydney T. Famularo
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Benjamin P. Dunham
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Ryan Schwark
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Reza Karbalaei
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Carmen Dressler
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Charlotte C. Bavley
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Nathan T. Fried
- Department of Biology, Rutgers Camden University, Camden, NJ, USA
| | - Mathieu E. Wimmer
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Ishmail Abdus-Saboor
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
- Corresponding author.
| |
Collapse
|
21
|
Mujinya R, Kalange M, Ochieng JJ, Ninsiima HI, Eze ED, Afodun AM, Nabirumbi R, Sulaiman SO, Kairania E, Echoru I, Okpanachi AO, Matama K, Asiimwe OH, Nambuya G, Usman IM, Obado OL, Zirintunda G, Ssempijja F, Nansunga M, Matovu H, Ayikobua ET, Nganda PE, Onanyang D, Ekou J, Musinguzi SP, Ssimbwa G, Kasozi KI. Cerebral Cortical Activity During Academic Stress Amongst Undergraduate Medical Students at Kampala International University (Uganda). Front Psychiatry 2022; 13:551508. [PMID: 35757206 PMCID: PMC9231459 DOI: 10.3389/fpsyt.2022.551508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 03/29/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Stress among medical students is related to their academic lifespan; however, information on brain health among medical students from developing countries continues to be scarce. The objective of this study was to establish perceived academic stress levels, assess the ability to cope with stress, and investigate its effects on the visual reaction time (VRT), audio reaction time (ART), and tactile reaction time (TRT) in the somatosensory cortex among medical students of Uganda. METHODS This was a cross-sectional study conducted among preclinical (n = 88) and clinical (n = 96) undergraduate medical students at Kampala International University Western Campus. A standard Perceived Stress Scale (PSS) was used to categorize stress into low, moderate, and severe while the ability to cope with stress was categorized into below average, average, above average, and superior stresscoper (SS). Data on reaction time were acquired through VRT, ART, and TRT using the catch-a-ruler experiment, and this was analyzed using SPSS version 20. RESULTS This study shows that preclinical students are more stressed than clinical students (PSS prevalence for low stress = preclinical; clinical: 40, 60%). Moderate stress was 48.4 and 51.6% while high perceived stress was 75 and 25% among preclinical and clinical students. Among male and female students in preclinical years, higher TRT and VRT were found in clinical students showing that stress affects the tactile and visual cortical areas in the brain, although the VRT scores were only significantly (P = 0.0123) poor in male students than female students in biomedical sciences. Also, highly stressed individuals had higher TRT and ART and low VRT. SS had high VRT and ART and low TRT in preclinical students, demonstrating the importance of the visual cortex in stress plasticity. Multiple regression showed a close relationship between PSS, ability to cope with stress, age, and educational level (P < 0.05), demonstrating the importance of social and psychological support, especially in the biomedical sciences. CONCLUSION Preclinical students suffer more from stress and are poorer SS than clinical students. This strongly impairs their cortical regions in the brain, thus affecting their academic productivity.
Collapse
Affiliation(s)
- Regan Mujinya
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | - Muhamudu Kalange
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | - Juma John Ochieng
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | | | | | - Adam Moyosore Afodun
- Department of Anatomy and Cell Biology, Faculty of Health Sciences, Busitema University, Tororo, Uganda
| | | | - Sheu Oluwadare Sulaiman
- Graduate Program in Cell Biology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Emmanuel Kairania
- Department of Anatomy and Cell Biology, Faculty of Health Sciences, Busitema University, Tororo, Uganda
| | - Isaac Echoru
- School of Medicine, Kabale University, Kabale, Uganda
| | | | - Kevin Matama
- Department of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy, Kampala International University Western Campus, Bushenyi, Uganda
| | - Oscar Hilary Asiimwe
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | - Grace Nambuya
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | - Ibe Michael Usman
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | | | - Gerald Zirintunda
- Department of Animal Production and Management, Faculty of Agriculture and Animal Sciences, Busitema University, Tororo, Uganda
| | - Fred Ssempijja
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | - Miriam Nansunga
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda.,Department of Physiology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Henry Matovu
- Department of Animal Production and Management, Faculty of Agriculture and Animal Sciences, Busitema University, Tororo, Uganda
| | | | - Ponsiano Ernest Nganda
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | - David Onanyang
- Department of Biology, Faculty of Science, Gulu University, Gulu, Uganda
| | - Justine Ekou
- Department of Animal Production and Management, Faculty of Agriculture and Animal Sciences, Busitema University, Tororo, Uganda
| | - Simon Peter Musinguzi
- Department of Agriculture Production, Faculty of Agriculture, Kyambogo University, Kampala, Uganda
| | - Godfrey Ssimbwa
- Department of Physiology, Faculty of Health Sciences, Muni University, Arua, Uganda
| | | |
Collapse
|
22
|
Ran C, Kamalani GNA, Chen X. Modality-Specific Modulation of Temperature Representations in the Spinal Cord after Injury. J Neurosci 2021; 41:8210-8219. [PMID: 34408066 PMCID: PMC8482863 DOI: 10.1523/jneurosci.1104-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/06/2021] [Accepted: 08/11/2021] [Indexed: 12/25/2022] Open
Abstract
Different types of tissue injury, such as inflammatory and neuropathic conditions, cause modality-specific alternations on temperature perception. There are profound changes in peripheral sensory neurons after injury, but how patterned neuronal activities in the CNS encode injury-induced sensitization to temperature stimuli is largely unknown. Using in vivo calcium imaging and mouse genetics, we show that formalin- and prostaglandin E2-induced inflammation dramatically increase spinal responses to heating and decrease responses to cooling in male and female mice. The reduction of cold response is largely eliminated on ablation of TRPV1-expressing primary sensory neurons, indicating a crossover inhibition of cold response from the hyperactive heat inputs in the spinal cord. Interestingly, chemotherapy medication oxaliplatin can rapidly increase spinal responses to cooling and suppress responses to heating. Together, our results suggest a push-pull mechanism in processing cold and heat inputs and reveal a synergic mechanism to shift thermosensation after injury.SIGNIFICANCE STATEMENT In this paper, we combine our novel in vivo spinal cord two-photon calcium imaging, mouse genetics, and persistent pain models to study how tissue injury alters the sensation of temperature. We discover modality-specific changes of spinal temperature responses in different models of injury. Chemotherapy medication oxaliplatin leads to cold hypersensitivity and heat hyposensitivity. By contrast, inflammation increases heat sensitivity and decreases cold sensitivity. This decrease in cold sensitivity results from the stronger crossover inhibition from the hyperactive heat inputs. Our work reveals the bidirectional change of thermosensitivity by injury and suggests that the crossover inhibitory circuit underlies the shifted thermosensation, providing a mechanism to the biased perception toward a unique thermal modality that was observed clinically in chronic pain patients.
Collapse
Affiliation(s)
- Chen Ran
- Department of Biology, Stanford University, Stanford, California 94305
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | | | - Xiaoke Chen
- Department of Biology, Stanford University, Stanford, California 94305
| |
Collapse
|
23
|
Russ DE, Cross RBP, Li L, Koch SC, Matson KJE, Yadav A, Alkaslasi MR, Lee DI, Le Pichon CE, Menon V, Levine AJ. A harmonized atlas of mouse spinal cord cell types and their spatial organization. Nat Commun 2021; 12:5722. [PMID: 34588430 PMCID: PMC8481483 DOI: 10.1038/s41467-021-25125-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
Single-cell RNA sequencing data can unveil the molecular diversity of cell types. Cell type atlases of the mouse spinal cord have been published in recent years but have not been integrated together. Here, we generate an atlas of spinal cell types based on single-cell transcriptomic data, unifying the available datasets into a common reference framework. We report a hierarchical structure of postnatal cell type relationships, with location providing the highest level of organization, then neurotransmitter status, family, and finally, dozens of refined populations. We validate a combinatorial marker code for each neuronal cell type and map their spatial distributions in the adult spinal cord. We also show complex lineage relationships among postnatal cell types. Additionally, we develop an open-source cell type classifier, SeqSeek, to facilitate the standardization of cell type identification. This work provides an integrated view of spinal cell types, their gene expression signatures, and their molecular organization.
Collapse
Affiliation(s)
- Daniel E Russ
- Division of Cancer Epidemiology and Genetics, Data Science Research Group, National Cancer Institute, NIH, Rockville, MD, USA
| | - Ryan B Patterson Cross
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Li Li
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Stephanie C Koch
- Department of Neuroscience, Physiology and Pharmacology, Division of Biosciences, University College London, London, UK
| | - Kaya J E Matson
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Archana Yadav
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA
| | - Mor R Alkaslasi
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA.,Department of Neuroscience, Brown University, Providence, RI, USA
| | - Dylan I Lee
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA
| | - Claire E Le Pichon
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Vilas Menon
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA
| | - Ariel J Levine
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA.
| |
Collapse
|
24
|
Miranda CO, Hegedüs K, Wildner H, Zeilhofer HU, Antal M. Morphological and neurochemical characterization of glycinergic neurons in laminae I-IV of the mouse spinal dorsal horn. J Comp Neurol 2021; 530:607-626. [PMID: 34382691 DOI: 10.1002/cne.25232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 08/04/2021] [Accepted: 08/09/2021] [Indexed: 12/18/2022]
Abstract
A growing body of experimental evidence shows that glycinergic inhibition plays vital roles in spinal pain processing. In spite of this, however, our knowledge about the morphology, neurochemical characteristics, and synaptic relations of glycinergic neurons in the spinal dorsal horn is very limited. The lack of this knowledge makes our understanding about the specific contribution of glycinergic neurons to spinal pain processing quite vague. Here we investigated the morphology and neurochemical characteristics of glycinergic neurons in laminae I-IV of the spinal dorsal horn using a GlyT2::CreERT2-tdTomato transgenic mouse line. Confirming previous reports, we show that glycinergic neurons are sparsely distributed in laminae I-II, but their densities are much higher in lamina III and especially in lamina IV. First in the literature, we provide experimental evidence indicating that in addition to neurons in which glycine colocalizes with GABA, there are glycinergic neurons in laminae I-II that do not express GABA and can thus be referred to as glycine-only neurons. According to the shape and size of cell bodies and dendritic morphology, we divided the tdTomato-labeled glycinergic neurons into three and six morphological groups in laminae I-II and laminae III-IV, respectively. We also demonstrate that most of the glycinergic neurons co-express neuronal nitric oxide synthase, parvalbumin, the receptor tyrosine kinase RET, and the retinoic acid-related orphan nuclear receptor β (RORβ), but there might be others that need further neurochemical characterization. The present findings may foster our understanding about the contribution of glycinergic inhibition to spinal pain processing.
Collapse
Affiliation(s)
- Camila Oliveira Miranda
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Krisztina Hegedüs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Hendrik Wildner
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Miklós Antal
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
25
|
Xia LP, Luo H, Ma Q, Xie YK, Li W, Hu H, Xu ZZ. GPR151 in nociceptors modulates neuropathic pain via regulating P2X3 function and microglial activation. Brain 2021; 144:3405-3420. [PMID: 34244727 DOI: 10.1093/brain/awab245] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 05/17/2021] [Accepted: 06/01/2021] [Indexed: 11/14/2022] Open
Abstract
Neuropathic pain is a major health problem that affects up to 7-10% of the population worldwide. Currently, neuropathic pain is difficult to treat due to its elusive mechanisms. Here we report that orphan G protein-coupled receptor 151 (GPR151) in nociceptive sensory neurons controls neuropathic pain induced by nerve injury. GPR151 was mainly expressed in nonpeptidergic C-fiber dorsal root ganglion (DRG) neurons and highly upregulated after nerve injury. Importantly, conditional knockout of Gpr151 in adult nociceptive sensory neurons significantly alleviated chronic constriction injury (CCI)-induced neuropathic pain-like behavior but did not affect basal nociception. Moreover, GPR151 in DRG neurons was required for CCI-induced neuronal hyperexcitability and upregulation of colony-stimulating factor 1 (CSF1), which is necessary for microglial activation in the spinal cord after nerve injury. Mechanistically, GPR151 coupled with P2X3 ion channels and promoted their functional activities in neuropathic pain-like hypersensitivity. Knockout of Gpr151 suppressed P2X3-mediated calcium elevation and spontaneous pain behavior in CCI mice. Conversely, overexpression of Gpr151 significantly enhanced P2X3-mediated calcium elevation and DRG neuronal excitability. Furthermore, knockdown of P2X3 in DRGs reversed CCI-induced CSF1 upregulation, spinal microglial activation, and neuropathic pain-like behavior. Finally, the co-expression of GPR151 and P2X3 was confirmed in small-diameter human DRG neurons, indicating the clinical relevance of our findings. Together, our results suggest that GPR151 in nociceptive DRG neurons plays a key role in the pathogenesis of neuropathic pain and could be a potential target for treating neuropathic pain.
Collapse
Affiliation(s)
- Li-Ping Xia
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Hao Luo
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Qiang Ma
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ya-Kai Xie
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Wei Li
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Hailan Hu
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zhen-Zhong Xu
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
26
|
A subset of spinal dorsal horn interneurons crucial for gating touch-evoked pain-like behavior. Proc Natl Acad Sci U S A 2021; 118:2021220118. [PMID: 33431693 DOI: 10.1073/pnas.2021220118] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
A cardinal, intractable symptom of neuropathic pain is mechanical allodynia, pain caused by innocuous stimuli via low-threshold mechanoreceptors such as Aβ fibers. However, the mechanism by which Aβ fiber-derived signals are converted to pain remains incompletely understood. Here we identify a subset of inhibitory interneurons in the spinal dorsal horn (SDH) operated by adeno-associated viral vectors incorporating a neuropeptide Y promoter (AAV-NpyP+) and show that specific ablation or silencing of AAV-NpyP+ SDH interneurons converted touch-sensing Aβ fiber-derived signals to morphine-resistant pain-like behavioral responses. AAV-NpyP+ neurons received excitatory inputs from Aβ fibers and transmitted inhibitory GABA signals to lamina I neurons projecting to the brain. In a model of neuropathic pain developed by peripheral nerve injury, AAV-NpyP+ neurons exhibited deeper resting membrane potentials, and their excitation by Aβ fibers was impaired. Conversely, chemogenetic activation of AAV-NpyP+ neurons in nerve-injured rats reversed Aβ fiber-derived neuropathic pain-like behavior that was shown to be morphine-resistant and reduced pathological neuronal activation of superficial SDH including lamina I. These findings suggest that identified inhibitory SDH interneurons that act as a critical brake on conversion of touch-sensing Aβ fiber signals into pain-like behavioral responses. Thus, enhancing activity of these neurons may offer a novel strategy for treating neuropathic allodynia.
Collapse
|
27
|
Spinal Inhibitory Interneurons: Gatekeepers of Sensorimotor Pathways. Int J Mol Sci 2021; 22:ijms22052667. [PMID: 33800863 PMCID: PMC7961554 DOI: 10.3390/ijms22052667] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/26/2021] [Accepted: 03/04/2021] [Indexed: 12/20/2022] Open
Abstract
The ability to sense and move within an environment are complex functions necessary for the survival of nearly all species. The spinal cord is both the initial entry site for peripheral information and the final output site for motor response, placing spinal circuits as paramount in mediating sensory responses and coordinating movement. This is partly accomplished through the activation of complex spinal microcircuits that gate afferent signals to filter extraneous stimuli from various sensory modalities and determine which signals are transmitted to higher order structures in the CNS and to spinal motor pathways. A mechanistic understanding of how inhibitory interneurons are organized and employed within the spinal cord will provide potential access points for therapeutics targeting inhibitory deficits underlying various pathologies including sensory and movement disorders. Recent studies using transgenic manipulations, neurochemical profiling, and single-cell transcriptomics have identified distinct populations of inhibitory interneurons which express an array of genetic and/or neurochemical markers that constitute functional microcircuits. In this review, we provide an overview of identified neural components that make up inhibitory microcircuits within the dorsal and ventral spinal cord and highlight the importance of inhibitory control of sensorimotor pathways at the spinal level.
Collapse
|
28
|
Shadrach JL, Gomez-Frittelli J, Kaltschmidt JA. Proprioception revisited: where do we stand? CURRENT OPINION IN PHYSIOLOGY 2021; 21:23-28. [PMID: 34222735 DOI: 10.1016/j.cophys.2021.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Originally referred to as 'muscle sense', the notion that skeletal muscle held a peripheral sensory function was first described early in the 19th century. Foundational experiments by Sherrington in the early 20th century definitively demonstrated that proprioceptors contained within skeletal muscle, tendons, and joints are innervated by sensory neurons and play an important role in the control of movement. In this review, we will highlight several recent advances in the ongoing effort to further define the molecular diversity underlying the proprioceptive sensorimotor system. Together, the work summarized here represents our current understanding of sensorimotor circuit formation during development and the mechanisms that regulate the integration of proprioceptive feedback into the spinal circuits that control locomotion in both normal and diseased states.
Collapse
Affiliation(s)
- Jennifer L Shadrach
- Department of Neurosurgery, Stanford University, Stanford, CA, 94305, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, 94305, USA
| | - Julieta Gomez-Frittelli
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, 94305, USA.,Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Julia A Kaltschmidt
- Department of Neurosurgery, Stanford University, Stanford, CA, 94305, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
29
|
Presynaptic Inhibition of Pain and Touch in the Spinal Cord: From Receptors to Circuits. Int J Mol Sci 2021; 22:ijms22010414. [PMID: 33401784 PMCID: PMC7795800 DOI: 10.3390/ijms22010414] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 02/07/2023] Open
Abstract
Sensory primary afferent fibers, conveying touch, pain, itch, and proprioception, synapse onto spinal cord dorsal horn neurons. Primary afferent central terminals express a wide variety of receptors that modulate glutamate and peptide release. Regulation of the amount and timing of neurotransmitter release critically affects the integration of postsynaptic responses and the coding of sensory information. The role of GABA (γ-aminobutyric acid) receptors expressed on afferent central terminals is particularly important in sensory processing, both in physiological conditions and in sensitized states induced by chronic pain. During the last decade, techniques of opto- and chemogenetic stimulation and neuronal selective labeling have provided interesting insights on this topic. This review focused on the recent advances about the modulatory effects of presynaptic GABAergic receptors in spinal cord dorsal horn and the neural circuits involved in these mechanisms.
Collapse
|
30
|
Liu P, Zhang X, He X, Jiang Z, Wang Q, Lu Y. Spinal GABAergic neurons are under feed-forward inhibitory control driven by A δ and C fibers in Gad2 td-Tomato mice. Mol Pain 2021; 17:1744806921992620. [PMID: 33586515 PMCID: PMC7890716 DOI: 10.1177/1744806921992620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Spinal GABAergic neurons act as a critical modulator in sensory transmission like pain or itch. The monosynaptic or polysynaptic primary afferent inputs onto GABAergic neurons, along with other interneurons or projection neurons make up the direct and feed-forward inhibitory neural circuits. Previous research indicates that spinal GABAergic neurons mainly receive excitatory inputs from Aδ and C fibers. However, whether they are controlled by other inhibitory sending signals is not well understood. METHODS We applied a transgenic mouse line in which neurons co-expressed the GABA-synthesizing enzyme Gad65 and the enhanced red fluorescence (td-Tomato) to characterize the features of morphology and electrophysiology of GABAergic neurons. Patch-clamp whole cell recordings were used to record the evoked postsynaptic potentials of fluorescent neurons in spinal slices in response to dorsal root stimulation. RESULTS We demonstrated that GABAergic neurons not only received excitatory drive from peripheral Aβ, Aδ and C fibers, but also received inhibitory inputs driven by Aδ and C fibers. The evoked inhibitory postsynaptic potentials (eIPSPs) mediated by C fibers were mainly Glycinergic (66.7%) as well as GABAergic mixed with Glycinergic (33.3%), whereas the inhibition mediated by Aδ fibers was predominately both GABA and Glycine-dominant (57.1%), and the rest of which was purely Glycine-dominant (42.9%). CONCLUSION These results indicated that spinal GABAergic inhibitory neurons are under feedforward inhibitory control driven by primary C and Aδ fibers, suggesting that this feed-forward inhibitory pathway may play an important role in balancing the excitability of GABAergic neurons in spinal dorsal horn.
Collapse
Affiliation(s)
- Peng Liu
- Department of Pain Medicine, Department of Anesthesiology & Perioprative Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiao Zhang
- Department of Pain Medicine, Department of Anesthesiology & Perioprative Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiaolan He
- Department of Pain Medicine, Department of Anesthesiology & Perioprative Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhenhua Jiang
- Department of Pain Medicine, Department of Anesthesiology & Perioprative Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Qun Wang
- Department of Pain Medicine, Department of Anesthesiology & Perioprative Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yan Lu
- Department of Pain Medicine, Department of Anesthesiology & Perioprative Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
31
|
Abdus-Saboor I, Fried NT, Lay M, Burdge J, Swanson K, Fischer R, Jones J, Dong P, Cai W, Guo X, Tao YX, Bethea J, Ma M, Dong X, Ding L, Luo W. Development of a Mouse Pain Scale Using Sub-second Behavioral Mapping and Statistical Modeling. Cell Rep 2020; 28:1623-1634.e4. [PMID: 31390574 PMCID: PMC6724534 DOI: 10.1016/j.celrep.2019.07.017] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 02/18/2019] [Accepted: 07/08/2019] [Indexed: 01/05/2023] Open
Abstract
Rodents are the main model systems for pain research, but determining their pain state is challenging. To develop an objective method to assess pain sensation in mice, we adopt high-speed videography to capture sub-second behavioral features following hind paw stimulation with both noxious and innocuous stimuli and identify several differentiating parameters indicating the affective and reflexive aspects of nociception. Using statistical modeling and machine learning, we integrate these parameters into a single index and create a "mouse pain scale," which allows us to assess pain sensation in a graded manner for each withdrawal. We demonstrate the utility of this method by determining sensations triggered by three different von Frey hairs and optogenetic activation of two different nociceptor populations. Our behavior-based "pain scale" approach will help improve the rigor and reproducibility of using withdrawal reflex assays to assess pain sensation in mice.
Collapse
Affiliation(s)
- Ishmail Abdus-Saboor
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nathan T Fried
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biology, Rutgers University, Camden, NJ 08102, USA
| | - Mark Lay
- Howard Hughes Medical Institute and Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Justin Burdge
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathryn Swanson
- Department of Biology, Drexel University, College of Arts and Sciences, Philadelphia, PA 19104, USA
| | - Roman Fischer
- Department of Biology, Drexel University, College of Arts and Sciences, Philadelphia, PA 19104, USA
| | - Jessica Jones
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter Dong
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Weihua Cai
- Department of Anesthesiology, Rutgers University Medical School, Newark, NJ 07101, USA
| | - Xinying Guo
- Department of Anesthesiology, Rutgers University Medical School, Newark, NJ 07101, USA
| | - Yuan-Xiang Tao
- Department of Anesthesiology, Rutgers University Medical School, Newark, NJ 07101, USA
| | - John Bethea
- Department of Biology, Drexel University, College of Arts and Sciences, Philadelphia, PA 19104, USA
| | - Minghong Ma
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xinzhong Dong
- Howard Hughes Medical Institute and Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Long Ding
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wenqin Luo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
32
|
Jones JM, Foster W, Twomey CR, Burdge J, Ahmed OM, Pereira TD, Wojick JA, Corder G, Plotkin JB, Abdus-Saboor I. A machine-vision approach for automated pain measurement at millisecond timescales. eLife 2020; 9:e57258. [PMID: 32758355 PMCID: PMC7434442 DOI: 10.7554/elife.57258] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 08/05/2020] [Indexed: 12/28/2022] Open
Abstract
Objective and automatic measurement of pain in mice remains a barrier for discovery in neuroscience. Here, we capture paw kinematics during pain behavior in mice with high-speed videography and automated paw tracking with machine and deep learning approaches. Our statistical software platform, PAWS (Pain Assessment at Withdrawal Speeds), uses a univariate projection of paw position over time to automatically quantify seven behavioral features that are combined into a single, univariate pain score. Automated paw tracking combined with PAWS reveals a behaviorally divergent mouse strain that displays hypersensitivity to mechanical stimuli. To demonstrate the efficacy of PAWS for detecting spinally versus centrally mediated behavioral responses, we chemogenetically activated nociceptive neurons in the amygdala, which further separated the pain-related behavioral features and the resulting pain score. Taken together, this automated pain quantification approach will increase objectivity in collecting rigorous behavioral data, and it is compatible with other neural circuit dissection tools for determining the mouse pain state.
Collapse
Affiliation(s)
- Jessica M Jones
- Department of Biology, University of PennsylvaniaPhiladelphiaUnited States
| | - William Foster
- Department of Biology, University of PennsylvaniaPhiladelphiaUnited States
| | - Colin R Twomey
- Department of Biology, University of PennsylvaniaPhiladelphiaUnited States
| | - Justin Burdge
- Department of Biology, University of PennsylvaniaPhiladelphiaUnited States
| | - Osama M Ahmed
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Talmo D Pereira
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Jessica A Wojick
- Departments of Psychiatry and Neuroscience, University of PennsylvaniaPhiladelphiaUnited States
| | - Gregory Corder
- Departments of Psychiatry and Neuroscience, University of PennsylvaniaPhiladelphiaUnited States
| | - Joshua B Plotkin
- Department of Biology, University of PennsylvaniaPhiladelphiaUnited States
| | | |
Collapse
|
33
|
Synaptic Dynamics of the Feed-forward Inhibitory Circuitry Gating Mechanical Allodynia in Mice. Anesthesiology 2020; 132:1212-1228. [PMID: 32101975 DOI: 10.1097/aln.0000000000003194] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND The authors' previous studies have found that spinal protein kinase C γ expressing neurons are involved in the feed-forward inhibitory circuit gating mechanical allodynia in the superficial dorsal horn. The authors hypothesize that nerve injury enhances the excitability of spinal protein kinase C γ expressing interneurons due to disinhibition of the feed-forward inhibitory circuit, and enables Aβ primary inputs to activate spinal protein kinase C γ expressing interneurons. METHODS Prkcg-P2A-tdTomato mice were constructed using the clustered regularly interspaced short palindromic repeats and clustered regularly interspaced short palindromic repeats-associated nuclease 9 technology, and were used to analyze the electrophysiologic properties of spinal protein kinase C γ expressing neurons in both normal conditions and pathologic conditions induced by chronic constriction injury of the sciatic nerve. Patch-clamp whole cell recordings were used to identify the nature of the dynamic synaptic drive to protein kinase C γ expressing neurons. RESULTS Aβ fiber stimulation evoked a biphasic synaptic response in 42% (31 of 73) of protein kinase C γ expressing neurons. The inhibitory components of the biphasic synaptic response were blocked by both strychnine and bicuculline in 57% (16 of 28) of neurons. Toll-like receptor 5 immunoreactive fibers made close contact with protein kinase C γ expressing neurons. After nerve injury, the percentage of neurons double-labeled for c-fos and Prkcg-P2A-tdTomato in animals walking on a rotarod was significantly higher than that in the nerve injury animals (4.1% vs. 9.9%, 22 of 539 vs. 54 of 548,P < 0.001). Aβ fiber stimulation evoked burst action potentials in 25.8% (8 of 31) of protein kinase C γ expressing neurons in control animals, while the proportion increased to 51.1% (23 of 45) in nerve injury animals (P = 0.027). CONCLUSIONS The Prkcg-P2A-tdTomato mice the authors constructed provide a useful tool for further analysis on how the spinal allodynia gate works. The current study indicated that nerve injury enhanced the excitability of spinal protein kinase C γ expressing interneurons due to disinhibition of the feed-forward inhibitory circuit, and enabled Aβ primary inputs to activate spinal protein kinase C γ expressing interneurons.
Collapse
|
34
|
Toll-like receptor 7 contributes to neuropathic pain by activating NF-κB in primary sensory neurons. Brain Behav Immun 2020; 87:840-851. [PMID: 32205121 PMCID: PMC7316623 DOI: 10.1016/j.bbi.2020.03.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/17/2020] [Accepted: 03/19/2020] [Indexed: 01/01/2023] Open
Abstract
Toll like receptor 7 (TLR7) is expressed in neurons of the dorsal root ganglion (DRG), but whether it contributes to neuropathic pain is elusive. We found that peripheral nerve injury caused by ligation of the fourth lumbar (L4) spinal nerve (SNL) or chronic constriction injury of sciatic nerve led to a significant increase in the expression of TLR7 at mRNA and protein levels in mouse injured DRG. Blocking this increase through microinjection of the adeno-associated virus (AAV) 5 expressing TLR7 shRNA into the ipsilateral L4 DRG alleviated the SNL-induced mechanical, thermal and cold pain hypersensitivities in both male and female mice. This microinjection also attenuated the SNL-induced increases in the levels of phosphorylated extracellular signal-regulated kinase ½ (p-ERK1/2) and glial fibrillary acidic protein (GFAP) in L4 dorsal horn on the ipsilateral side during both development and maintenance periods. Conversely, mimicking this increase through microinjection of AAV5 expressing full-length TLR7 into unilateral L3/4 DRGs led to elevations in the amounts of p-ERK1/2 and GFAP in the dorsal horn, augmented responses to mechanical, thermal and cold stimuli, and induced the spontaneous pain on the ipsilateral side in the absence of SNL. Mechanistically, the increased TLR7 activated the NF-κB signaling pathway through promoting the translocation of p65 into the nucleus and phosphorylation of p65 in the nucleus from the injured DRG neurons. Our findings suggest that DRG TLR7 contributes to neuropathic pain by activating NF-κB in primary sensory neurons. TLR7 may be a potential target for therapeutic treatment of this disorder.
Collapse
|
35
|
Graham RD, Bruns TM, Duan B, Lempka SF. The Effect of Clinically Controllable Factors on Neural Activation During Dorsal Root Ganglion Stimulation. Neuromodulation 2020; 24:655-671. [PMID: 32583523 DOI: 10.1111/ner.13211] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/08/2020] [Accepted: 05/10/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Dorsal root ganglion stimulation (DRGS) is an effective therapy for chronic pain, though its mechanisms of action are unknown. Currently, we do not understand how clinically controllable parameters (e.g., electrode position, stimulus pulse width) affect the direct neural response to DRGS. Therefore, the goal of this study was to utilize a computational modeling approach to characterize how varying clinically controllable parameters changed neural activation profiles during DRGS. MATERIALS AND METHODS We coupled a finite element model of a human L5 DRG to multicompartment models of primary sensory neurons (i.e., Aα-, Aβ-, Aδ-, and C-neurons). We calculated the stimulation amplitudes necessary to elicit one or more action potentials in each neuron, and examined how neural activation profiles were affected by varying clinically controllable parameters. RESULTS In general, DRGS predominantly activated large myelinated Aα- and Aβ-neurons. Shifting the electrode more than 2 mm away from the ganglion abolished most DRGS-induced neural activation. Increasing the stimulus pulse width to 500 μs or greater increased the number of activated Aδ-neurons, while shorter pulse widths typically only activated Aα- and Aβ-neurons. Placing a cathode near a nerve root, or an anode near the ganglion body, maximized Aβ-mechanoreceptor activation. Guarded active contact configurations did not activate more Aβ-mechanoreceptors than conventional bipolar configurations. CONCLUSIONS Our results suggest that DRGS applied with stimulation parameters within typical clinical ranges predominantly activates Aβ-mechanoreceptors. In general, varying clinically controllable parameters affects the number of Aβ-mechanoreceptors activated, although longer pulse widths can increase Aδ-neuron activation. Our data support several Neuromodulation Appropriateness Consensus Committee guidelines on the clinical implementation of DRGS.
Collapse
Affiliation(s)
- Robert D Graham
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Tim M Bruns
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Bo Duan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Scott F Lempka
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.,Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
36
|
Liang L, Wu S, Lin C, Chang YJ, Tao YX. Alternative Splicing of Nrcam Gene in Dorsal Root Ganglion Contributes to Neuropathic Pain. THE JOURNAL OF PAIN 2020; 21:892-904. [PMID: 31917219 DOI: 10.1016/j.jpain.2019.12.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 11/04/2019] [Accepted: 12/11/2019] [Indexed: 01/23/2023]
Abstract
NrCAM, a neuronal cell adhesion molecule in the L1 family of the immunoglobulin superfamily, is subjected to extensively alternative splicing and involved in neural development and some disorders. The aim of this study was to explore the role of Nrcam mRNA alternative splicing in neuropathic pain. A next generation RNA sequencing analysis of dorsal root ganglions (DRGs) showed the differential expression of two splicing variants of Nrcam, Nrcam+10 and Nrcam-10, in the injured DRG after the fourth lumbar spinal nerve ligation (SNL) in mice. SNL increased the exon 10 insertion, resulting in an increase in the amount of Nrcam+10 and a corresponding decrease in the level of Nrcam-10 in the injured DRG. An antisense oligonucleotide (ASO) that specifically targeted exon 10 of Nrcam gene (Nrcam ASO) repressed RNA expression of Nrcam+10 and increased RNA expression of Nrcam-10 in in vitro DRG cell culture. Either DRG microinjection or intrathecal injection of Nrcam ASO attenuated SNL-induced the development of mechanical allodynia, thermal hyperalgesia, or cold allodynia. Nrcam ASO also relieved SNL- or chronic compression of DRG (CCD)-induced the maintenance of pain hypersensitivities in male and female mice. PERSPECTIVE: We conclude that the relative levels of alternatively spliced Nrcam variants are critical for neuropathic pain genesis. Targeting Nrcam alternative splicing via the antisense oligonucleotides may be a new potential avenue in neuropathic pain management.
Collapse
Affiliation(s)
- Lingli Liang
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Shaogen Wu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Corinna Lin
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Yun-Juan Chang
- Office of advanced research computing, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey; Department of Cell Biology & Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey; Department of Physiology, Pharmacology & Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey.
| |
Collapse
|
37
|
Synaptic control of spinal GRPR + neurons by local and long-range inhibitory inputs. Proc Natl Acad Sci U S A 2019; 116:27011-27017. [PMID: 31806757 DOI: 10.1073/pnas.1905658116] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Spinal gastrin-releasing peptide receptor-expressing (GRPR+) neurons play an essential role in itch signal processing. However, the circuit mechanisms underlying the modulation of spinal GRPR+ neurons by direct local and long-range inhibitory inputs remain elusive. Using viral tracing and electrophysiological approaches, we dissected the neural circuits underlying the inhibitory control of spinal GRPR+ neurons. We found that spinal galanin+ GABAergic neurons form inhibitory synapses with GRPR+ neurons in the spinal cord and play an important role in gating the GRPR+ neuron-dependent itch signaling pathway. Spinal GRPR+ neurons also receive inhibitory inputs from local neurons expressing neuronal nitric oxide synthase (nNOS). Moreover, spinal GRPR+ neurons are gated by strong inhibitory inputs from the rostral ventromedial medulla. Thus, both local and long-range inhibitory inputs could play important roles in gating itch processing in the spinal cord by directly modulating the activity of spinal GRPR+ neurons.
Collapse
|
38
|
Propriospinal Neurons of L3-L4 Segments Involved in Control of the Rat External Urethral Sphincter. Neuroscience 2019; 425:12-28. [PMID: 31785359 DOI: 10.1016/j.neuroscience.2019.11.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/31/2019] [Accepted: 11/11/2019] [Indexed: 12/17/2022]
Abstract
Coordination of activity of external urethral sphincter (EUS) striated muscle and bladder (BL) smooth muscle is essential for efficient voiding. In this study we examined the morphological and electrophysiological properties of neurons in the L3/L4 spinal cord (SC) that are likely to have an important role in EUS-BL coordination in rats. EUS-related SC neurons were identified by retrograde transsynaptic tracing following injection of pseudorabies virus (PRV) co-expressing fluorescent markers into the EUS of P18-P20 male rats. Tracing revealed not only EUS motoneurons in L6/S1 but also interneurons in lamina X of the L6/S1 and L3/L4 SC. Physiological properties of fluorescently labeled neurons were assessed during whole-cell recordings in SC slices followed by reconstruction of biocytin-filled neurons. Reconstructions of neuronal processes from transverse or longitudinal slices showed that some L3/L4 neurons have axons projecting toward and into the ventro-medial funiculus (VMf) where axons extended caudally. Other neurons had axons projecting within laminae X and VII. Dendrites of L3/L4 neurons were distributed within laminae X and VII. The majority of L3/L4 neurons exhibited tonic firing in response to depolarizing currents. In transverse slices focal electrical stimulation (FES) in the VMf or in laminae X and VII elicited antidromic axonal spikes and/or excitatory synaptic responses in L3/L4 neurons; while in longitudinal slices FES elicited excitatory synaptic inputs from sites up to 400 μm along the central canal. Inhibitory inputs were rarely observed. These data suggest that L3/L4 EUS-related circuitry consists of at least two neuronal populations: segmental interneurons and propriospinal neurons projecting to L6/S1.
Collapse
|
39
|
Smith KM, Browne TJ, Davis OC, Coyle A, Boyle KA, Watanabe M, Dickinson SA, Iredale JA, Gradwell MA, Jobling P, Callister RJ, Dayas CV, Hughes DI, Graham BA. Calretinin positive neurons form an excitatory amplifier network in the spinal cord dorsal horn. eLife 2019; 8:49190. [PMID: 31713514 PMCID: PMC6908433 DOI: 10.7554/elife.49190] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/09/2019] [Indexed: 12/20/2022] Open
Abstract
Nociceptive information is relayed through the spinal cord dorsal horn, a critical area in sensory processing. The neuronal circuits in this region that underpin sensory perception must be clarified to better understand how dysfunction can lead to pathological pain. This study used an optogenetic approach to selectively activate spinal interneurons that express the calcium-binding protein calretinin (CR). We show that these interneurons form an interconnected network that can initiate and sustain enhanced excitatory signaling, and directly relay signals to lamina I projection neurons. Photoactivation of CR interneurons in vivo resulted in a significant nocifensive behavior that was morphine sensitive, caused a conditioned place aversion, and was enhanced by spared nerve injury. Furthermore, halorhodopsin-mediated inhibition of these interneurons elevated sensory thresholds. Our results suggest that dorsal horn circuits that involve excitatory CR neurons are important for the generation and amplification of pain and identify these interneurons as a future analgesic target.
Collapse
Affiliation(s)
- Kelly M Smith
- School of Biomedical Sciences & Pharmacy, Faculty of Health, University of Newcastle, Callaghan, Australia.,Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia.,Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, United States.,Department of Neurobiology, University of Pittsburgh, Pittsburgh, United States
| | - Tyler J Browne
- School of Biomedical Sciences & Pharmacy, Faculty of Health, University of Newcastle, Callaghan, Australia.,Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Olivia C Davis
- Institute of Neuroscience Psychology, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - A Coyle
- Institute of Neuroscience Psychology, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Kieran A Boyle
- Institute of Neuroscience Psychology, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo, Japan
| | - Sally A Dickinson
- School of Biomedical Sciences & Pharmacy, Faculty of Health, University of Newcastle, Callaghan, Australia.,Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Jacqueline A Iredale
- School of Biomedical Sciences & Pharmacy, Faculty of Health, University of Newcastle, Callaghan, Australia.,Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Mark A Gradwell
- School of Biomedical Sciences & Pharmacy, Faculty of Health, University of Newcastle, Callaghan, Australia.,Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Phillip Jobling
- School of Biomedical Sciences & Pharmacy, Faculty of Health, University of Newcastle, Callaghan, Australia.,Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Robert J Callister
- School of Biomedical Sciences & Pharmacy, Faculty of Health, University of Newcastle, Callaghan, Australia.,Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Christopher V Dayas
- School of Biomedical Sciences & Pharmacy, Faculty of Health, University of Newcastle, Callaghan, Australia.,Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - David I Hughes
- Institute of Neuroscience Psychology, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Brett A Graham
- School of Biomedical Sciences & Pharmacy, Faculty of Health, University of Newcastle, Callaghan, Australia.,Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| |
Collapse
|
40
|
Functional Reorganization of Local Circuit Connectivity in Superficial Spinal Dorsal Horn with Neuropathic Pain States. eNeuro 2019; 6:ENEURO.0272-19.2019. [PMID: 31533959 PMCID: PMC6787342 DOI: 10.1523/eneuro.0272-19.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/27/2019] [Accepted: 09/05/2019] [Indexed: 01/12/2023] Open
Abstract
The spinal dorsal horn is the first relay structure coding for pain transmission and modulation. Previous anatomical and electrophysiological studies have examined spinal dorsal horn circuit connections and network activity. Further work is required to understand spinal cord sensory information processing that underlies pathological neuropathic pain states. Our previous studies suggest that peripheral nerve injury enhances presynaptic excitatory input onto spinal superficial dorsal horn neurons, which in turn contributes to pathologic nociception. The potential changes in local postsynaptic circuits in the dorsal horn that lead to pathologically heightened behavioral responses to pain remain largely unexplored. We combined whole-cell electrophysiological recordings with laser-scanning photostimulation to test whether peripheral nerve injury in the spinal nerve ligation (SNL) mouse model of neuropathic pain leads to alterations in the functional connectivity of spinal cord circuits including lamina II excitatory interneurons. Here we show that SNL enhances excitation and decreases inhibition to lamina II excitatory interneurons along with their increased glutamate-evoked excitability. The enhanced excitatory postsynaptic input and connectivity evoked by SNL eventually return to normal levels concurrently with the resolution of the neuropathic pain states. The physiological pattern highly correlates with mouse pain behaviors following SNL, supporting a neurophysiological mechanism of central sensitization and neuropathic pain that is functionally localized to the spinal dorsal horn. Together, these data support that SNL induces functional changes in synaptic input and connectivity to lamina II excitatory interneurons that code for pain perception, and thus provide new insights into the mechanism and locus of pain hypersensitivity.
Collapse
|
41
|
Identification of Spinal Neurons Contributing to the Dorsal Column Projection Mediating Fine Touch and Corrective Motor Movements. Neuron 2019; 104:749-764.e6. [PMID: 31586516 DOI: 10.1016/j.neuron.2019.08.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 07/22/2019] [Accepted: 08/16/2019] [Indexed: 12/22/2022]
Abstract
Tactile stimuli are integrated and processed by neuronal circuits in the deep dorsal horn of the spinal cord. Several spinal interneuron populations have been implicated in tactile information processing. However, dorsal horn projection neurons that contribute to the postsynaptic dorsal column (PSDC) pathway transmitting tactile information to the brain are poorly characterized. Here, we show that spinal neurons marked by the expression of Zic2creER mediate light touch sensitivity and textural discrimination. A subset of Zic2creER neurons are PSDC neurons that project to brainstem dorsal column nuclei, and chemogenetic activation of Zic2 PSDC neurons increases sensitivity to light touch stimuli. Zic2 neurons receive direct input from the cortex and brainstem motor nuclei and are required for corrective motor movements. These results suggest that Zic2 neurons integrate sensory input from cutaneous afferents with descending signals from the brain to promote corrective movements and transmit processed touch information back to the brain. VIDEO ABSTRACT.
Collapse
|
42
|
Torsney C. Inflammatory pain neural plasticity. CURRENT OPINION IN PHYSIOLOGY 2019. [DOI: 10.1016/j.cophys.2019.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
43
|
Sadler KE, Langer SN, Menzel AD, Moehring F, Erb AN, Brandow AM, Stucky CL. Gabapentin alleviates chronic spontaneous pain and acute hypoxia-related pain in a mouse model of sickle cell disease. Br J Haematol 2019; 187:246-260. [PMID: 31247672 PMCID: PMC6786911 DOI: 10.1111/bjh.16067] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/07/2019] [Indexed: 12/23/2022]
Abstract
Pain is the main complication of sickle cell disease (SCD). Individuals with SCD experience acute pain episodes and chronic daily pain, both of which are managed with opioids. Opioids have deleterious side effects and use-associated stigma that make them less than ideal for SCD pain management. After recognizing the neuropathic qualities of SCD pain, clinically-approved therapies for neuropathic pain, including gabapentin, now present unique non-opioid based therapies for SCD pain management. These experiments explored the efficacy of gabapentin in relieving evoked and spontaneous chronic pain, and hypoxia/reoxygenation (H/R)-induced acute pain in mouse models of SCD. When administered following H/R, a single dose of gabapentin alleviated mechanical hypersensitivity in SCD mice by decreasing peripheral fibre activity. Gabapentin treatment also alleviated spontaneous ongoing pain in SCD mice. Longitudinal daily administration of gabapentin failed to alleviate H/R-induced pain or chronic evoked mechanical, cold or deep tissue hypersensitivity in SCD mice. Consistent with this observation, voltage-gated calcium channel (VGCC) α2 δ1 subunit expression was similar in sciatic nerve, dorsal root ganglia and lumbar spinal cord tissue from SCD and control mice. Based on these data, gabapentin may be an effective opioid alternative for the treatment of chronic spontaneous and acute H/R pain in SCD.
Collapse
Affiliation(s)
- Katelyn E Sadler
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sarah N Langer
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Anthony D Menzel
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Francie Moehring
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ashley N Erb
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Amanda M Brandow
- Department of Pediatrics, Section of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Cheryl L Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
44
|
Price TJ, Gold MS. From Mechanism to Cure: Renewing the Goal to Eliminate the Disease of Pain. PAIN MEDICINE 2019; 19:1525-1549. [PMID: 29077871 DOI: 10.1093/pm/pnx108] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objective Persistent pain causes untold misery worldwide and is a leading cause of disability. Despite its astonishing prevalence, pain is undertreated, at least in part because existing therapeutics are ineffective or cause intolerable side effects. In this review, we cover new findings about the neurobiology of pain and argue that all but the most transient forms of pain needed to avoid tissue damage should be approached as a disease where a cure can be the goal of all treatment plans, even if attaining this goal is not yet always possible. Design We reviewed the literature to highlight recent advances in the area of the neurobiology of pain. Results We discuss barriers that are currently hindering the achievement of this goal, as well as the development of new therapeutic strategies. We also discuss innovations in the field that are creating new opportunities to treat and even reverse persistent pain, some of which are in late-phase clinical trials. Conclusion We conclude that the confluence of new basic science discoveries and development of new technologies are creating a path toward pain therapeutics that should offer significant hope of a cure for patients and practitioners alike. Classification of Evidence. Our review points to new areas of inquiry for the pain field to advance the goal of developing new therapeutics to treat chronic pain.
Collapse
Affiliation(s)
- Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, Texas
| | - Michael S Gold
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
45
|
Petitjean H, Bourojeni FB, Tsao D, Davidova A, Sotocinal SG, Mogil JS, Kania A, Sharif-Naeini R. Recruitment of Spinoparabrachial Neurons by Dorsal Horn Calretinin Neurons. Cell Rep 2019; 28:1429-1438.e4. [DOI: 10.1016/j.celrep.2019.07.048] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 06/13/2019] [Accepted: 07/15/2019] [Indexed: 01/11/2023] Open
|
46
|
Ran C, Chen X. Probing the coding logic of thermosensation using spinal cord calcium imaging. Exp Neurol 2019; 318:42-49. [PMID: 31014574 PMCID: PMC6993943 DOI: 10.1016/j.expneurol.2019.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/25/2019] [Accepted: 04/19/2019] [Indexed: 12/20/2022]
Abstract
The spinal cord dorsal horn is the first relay station of the neural network for processing somatosensory information. High-throughput optical recording methods facilitate the study of sensory coding in the cortex but have not been successfully applied to study spinal cord circuitry until recently. Here, we review the development of an in vivo two-photon spinal calcium imaging preparation and biological findings from the first systematic characterization of the spinal response to cutaneous thermal stimuli, focusing on the difference between the coding of heat and cold, and the contribution of different peripheral inputs to thermosensory response in the spinal cord. Here we also report that knockout of TRPV1 channel impairs sensation of warmth, and somatostatin- and calbindin2-expressing neurons in the spinal dorsal horn preferentially respond to heat. Future work combining this technology with genetic tools and animal models of chronic pain will further elucidate the role of each neuronal type in the spinal thermosensory coding and their plasticity under pathological condition.
Collapse
Affiliation(s)
- Chen Ran
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| | - Xiaoke Chen
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
47
|
Khuong TM, Wang QP, Manion J, Oyston LJ, Lau MT, Towler H, Lin YQ, Neely GG. Nerve injury drives a heightened state of vigilance and neuropathic sensitization in Drosophila. SCIENCE ADVANCES 2019; 5:eaaw4099. [PMID: 31309148 PMCID: PMC6620091 DOI: 10.1126/sciadv.aaw4099] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 06/10/2019] [Indexed: 06/10/2023]
Abstract
Injury can lead to devastating and often untreatable chronic pain. While acute pain perception (nociception) evolved more than 500 million years ago, virtually nothing is known about the molecular origin of chronic pain. Here we provide the first evidence that nerve injury leads to chronic neuropathic sensitization in insects. Mechanistically, peripheral nerve injury triggers a loss of central inhibition that drives escape circuit plasticity and neuropathic allodynia. At the molecular level, excitotoxic signaling within GABAergic (γ-aminobutyric acid) neurons required the acetylcholine receptor nAChRα1 and led to caspase-dependent death of GABAergic neurons. Conversely, disruption of GABA signaling was sufficient to trigger allodynia without injury. Last, we identified the conserved transcription factor twist as a critical downstream regulator driving GABAergic cell death and neuropathic allodynia. Together, we define how injury leads to allodynia in insects, and describe a primordial precursor to neuropathic pain may have been advantageous, protecting animals after serious injury.
Collapse
Affiliation(s)
- Thang M. Khuong
- The Dr. John and Anne Chong Laboratory for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia
| | - Qiao-Ping Wang
- The Dr. John and Anne Chong Laboratory for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - John Manion
- The Dr. John and Anne Chong Laboratory for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia
| | - Lisa J. Oyston
- The Dr. John and Anne Chong Laboratory for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia
| | - Man-Tat Lau
- The Dr. John and Anne Chong Laboratory for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia
| | - Harry Towler
- The Dr. John and Anne Chong Laboratory for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia
| | - Yong Qi Lin
- The Dr. John and Anne Chong Laboratory for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia
| | - G. Gregory Neely
- The Dr. John and Anne Chong Laboratory for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia
- Genome Editing Initiative, The University of Sydney, NSW 2006, Australia
| |
Collapse
|
48
|
Greenspon CM, Battell EE, Devonshire IM, Donaldson LF, Chapman V, Hathway GJ. Lamina-specific population encoding of cutaneous signals in the spinal dorsal horn using multi-electrode arrays. J Physiol 2018; 597:377-397. [PMID: 30390415 PMCID: PMC6332738 DOI: 10.1113/jp277036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/31/2018] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Traditional, widely used in vivo electrophysiological techniques for the investigation of spinal processing of somatosensory information fail to account for the diverse functions of each lamina. To overcome this oversimplification, we have used multi-electrode arrays, in vivo, to simultaneously record neuronal activity across all laminae of the spinal dorsal horn. Multi-electrode arrays are sensitive enough to detect lamina- and region-specific encoding of different subtypes of afferent fibres and to detect short-lived changes in synaptic plasticity as measured by the application of cutaneous electrical stimulation of varying intensity and frequency. Differential encoding of innocuous and noxious thermal and mechanical stimuli were also detected across the laminae with the technique, as were the effects of the application of capsaicin. This new approach to the study of the dorsal spinal cord produces significantly more information per experiment, permitting accelerated research whilst also permitting the effects of pharmacological tools to modulate network responses. ABSTRACT The dorsal horn (DH) of the spinal cord is a complex laminar structure integrating peripheral signals into the central nervous system. Spinal somatosensory processing is commonly measured electrophysiologically in vivo by recording the activity of individual wide-dynamic-range neurons in the deep DH and extrapolating their behaviour to all cells in every lamina. This fails to account for the specialized processes that occur in each lamina and the considerable heterogeneity in cellular phenotype within and between laminae. Here we overcome this oversimplification by employing linear multi-electrode arrays (MEAs) in the DH of anaesthetized rats to simultaneously measure activity across all laminae. The MEAs, comprising 16 channels, were inserted into the lumbar dorsal horn and peripheral neurons activated electrically via transcutaneous electrodes and ethologically with von Frey hairs (vFHs) or an aluminium heating block. Ascending electrical stimuli showed fibre thresholds with distinct dorsoventral innervation profiles. Wind up was observed across the DH during the C-fibre and post-discharge latencies following 0.5 Hz stimulation. Intrathecal application of morphine (5 ng/50 μl) significantly reduced Aδ- and C-fibre-evoked activity in deep and superficial DH. Light vFHs (≤10 g) predominantly activated intermediate and deep laminae whereas noxious vFHs (26 g) also activated the superficial laminae. Noxious heat (55°C) induced significantly greater activity in the superficial and deep laminae than the innocuous control (30°C). The application of these arrays produced the first description of the processing of innocuous and noxious stimuli throughout the intact DH.
Collapse
Affiliation(s)
- Charles M Greenspon
- School of Life Sciences, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Emma E Battell
- School of Life Sciences, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Ian M Devonshire
- Bio-Support Unit, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Lucy F Donaldson
- School of Life Sciences, The University of Nottingham, Nottingham, NG7 2UH, UK.,Arthritis Research UK Pain Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Victoria Chapman
- School of Life Sciences, The University of Nottingham, Nottingham, NG7 2UH, UK.,Arthritis Research UK Pain Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Gareth J Hathway
- School of Life Sciences, The University of Nottingham, Nottingham, NG7 2UH, UK
| |
Collapse
|
49
|
MBD1 Contributes to the Genesis of Acute Pain and Neuropathic Pain by Epigenetic Silencing of Oprm1 and Kcna2 Genes in Primary Sensory Neurons. J Neurosci 2018; 38:9883-9899. [PMID: 30266739 DOI: 10.1523/jneurosci.0880-18.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 09/12/2018] [Accepted: 09/24/2018] [Indexed: 02/06/2023] Open
Abstract
The transmission of normal sensory and/or acute noxious information requires intact expression of pain-associated genes within the pain pathways of nervous system. Expressional changes of these genes after peripheral nerve injury are also critical for neuropathic pain induction and maintenance. Methyl-CpG-binding domain protein 1 (MBD1), an epigenetic repressor, regulates gene transcriptional activity. We report here that MBD1 in the primary sensory neurons of DRG is critical for the genesis of acute pain and neuropathic pain as DRG MBD1-deficient mice exhibit the reduced responses to acute mechanical, heat, cold, and capsaicin stimuli and the blunted nerve injury-induced pain hypersensitivities. Furthermore, DRG overexpression of MBD1 leads to spontaneous pain and evoked pain hypersensitivities in the WT mice and restores acute pain sensitivities in the MBD1-deficient mice. Mechanistically, MDB1 represses Oprm1 and Kcna2 gene expression by recruiting DNA methyltransferase DNMT3a into these two gene promoters in the DRG neurons. DRG MBD1 is likely a key player under the conditions of acute pain and neuropathic pain.SIGNIFICANCE STATEMENT In the present study, we revealed that the mice with deficiency of methyl-CpG-binding domain protein 1 (MBD1), an epigenetic repressor, in the DRG displayed the reduced responses to acute noxious stimuli and the blunted neuropathic pain. We also showed that DRG overexpression of MBD1 produced the hypersensitivities to noxious stimuli in the WT mice and rescued acute pain sensitivities in the MBD1-deficient mice. We have also provided the evidence that MDB1 represses Oprm1 and Kcna2 gene expression by recruiting DNA methyltransferase DNMT3a into these two gene promoters in the DRG neurons. DRG MBD1 may participate in the genesis of acute pain and neuropathic pain likely through regulating DNMT3a-controlled Oprm1 and Kcna2 gene expression in the DRG neurons.
Collapse
|
50
|
Timing Mechanisms Underlying Gate Control by Feedforward Inhibition. Neuron 2018; 99:941-955.e4. [PMID: 30122375 DOI: 10.1016/j.neuron.2018.07.026] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 05/20/2018] [Accepted: 07/17/2018] [Indexed: 01/20/2023]
Abstract
The gate control theory proposes that Aβ mechanoreceptor inputs to spinal pain transmission T neurons are gated via feedforward inhibition, but it remains unclear how monosynaptic excitation is gated by disynaptic inhibitory inputs that arrive later. Here we report that Aβ-evoked, non-NMDAR-dependent EPSPs in T neurons are subthreshold, allowing time for inhibitory inputs to prevent action potential firing that requires slow-onset NMDAR activation. Potassium channel activities-including IA, whose sizes are established constitutively by PreprodynorphinCre-derived inhibitory neurons-either completely filter away Aβ inputs or make them subthreshold, thereby creating a permissive condition to achieve gate control. Capsaicin-activated nociceptor inputs reduce IA and sensitize the T neurons, allowing Aβ inputs to cause firing before inhibitory inputs arrive. Thus, distinct kinetics of glutamate receptors and electric filtering by potassium channels solve the timing problem underlying the gating by feedforward inhibition, and their modulation offers a way to bypass the gate control.
Collapse
|