1
|
Wu L, He J, Shen N, Chen S. Molecular and cellular mechanisms underlying peripheral nerve injury-induced cellular ecological shifts: Implications for neuroregeneration. IBRO Neurosci Rep 2025; 18:120-129. [PMID: 39877591 PMCID: PMC11773043 DOI: 10.1016/j.ibneur.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
The peripheral nervous system is a complex ecological network, and its injury triggers a series of fine-grained intercellular regulations that play a crucial role in the repair process. The peripheral nervous system is a sophisticated ecological network, and its injury initiates a cascade of intricate intercellular regulatory processes that are instrumental in the repair process. Despite the advent of sophisticated microsurgical techniques, the repair of peripheral nerve injuries frequently proves inadequate, resulting in adverse effects on patients' quality of life. Accordingly, the continued pursuit of more efficacious treatments is of paramount importance. In this paper, a review of the relevant literature from recent years was conducted to identify the key cell types involved after peripheral nerve injury. These included Schwann cells, macrophages, neutrophils, endothelial cells, and fibroblasts. The review was conducted in depth. This paper analyses the phenotypic changes of these cells after injury, the relevant factors affecting these changes, and how they coordinate with neurons and other cell types. In addition, it explores the potential mechanisms that mediate the behaviour of these cells. Understanding the interactions between these cells and their mutual regulation with neurons is of great significance for the discovery of new neuroregenerative treatments and the identification of potential therapeutic targets.
Collapse
Affiliation(s)
- Limao Wu
- School of Clinical Medicine, Hebei University of Engineering, No.81 Congtai Road, Congtai District, Handan City, Hebei Province 056004, China
| | - Jinglan He
- Affiliated Hospital of Hebei University of Engineering, No. 80, Jianshe Street, Fuxing District, Handan City, Hebei Province 056003, China
| | - Na Shen
- Department of Science and Education, Affiliated Hospital of Hebei University of Engineering, No.81 Congtai Road, Congtai District, Handan City, Hebei Province 056004, China
| | - Song Chen
- Orthopaedic Center, Affiliated Hospital of Hebei University of Engineering, No.81 Congtai Road, Congtai District, Handan City, Hebei Province 56004, China
| |
Collapse
|
2
|
Huang Y, Ye Q, Wang J, Zhu K, Yang H, Jiang X, Shen M. Recent progress in the identification and in vitro culture of skin organoids. Regen Ther 2025; 29:341-351. [PMID: 40242086 PMCID: PMC12000699 DOI: 10.1016/j.reth.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/13/2024] [Accepted: 01/04/2025] [Indexed: 04/18/2025] Open
Abstract
An organoid is a cell-based structure that shows organ-specific properties and shares a similar spatial organization as the corresponding organ. Organoids possess powerful capability to reproduce the key functions of the associated organ structures, and their similarity to the organs makes them physiologically relevant systems. The primary challenge associated with the development of skin organoids is the complexity of the human skin architecture, which encompasses the epidermis and the dermis as well as accessory structures, including hair follicles, sweat glands, and sebaceous glands, that perform various functions such as thermoregulation. The ultimate objectives of developing skin organoids are to regenerate the complete skin structure in vitro and reconstruct the skin in vivo. Consequently, safety, reliability, and the fidelity of the tissue interfaces are key considerations in this process. For this purpose, the present article reviews the most recent advances in this field, focusing on the cell sources, culture methods, culture conditions, and biomarkers for identifying the structure and function of skin organoids developed in vitro or in vivo. The subsequent sections summarize the recent applications of skin organoids in related disease diagnosis and treatments, and discuss the future prospects of these organoids in terms of clinical applications. This review of skin organoids can provide an important foundation for studies on human skin development, disease modeling, and reconstructive surgery, with broad utility for promising future opportunities in both biomedical research and clinical practice.
Collapse
Affiliation(s)
- Yanan Huang
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| | - Qing Ye
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| | | | - Kaimin Zhu
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| | - Haojie Yang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, China
| | - Xiaoping Jiang
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| | - Meihua Shen
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| |
Collapse
|
3
|
Crosson T, Bretaud N, Ugolini S. Role of specialized sensory neuron subtypes in modulating peripheral immune responses. Immunity 2025; 58:1161-1174. [PMID: 40324383 DOI: 10.1016/j.immuni.2025.04.008] [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: 02/14/2025] [Revised: 04/07/2025] [Accepted: 04/07/2025] [Indexed: 05/07/2025]
Abstract
The immune and sensory nervous systems detect diverse threats, from tissue damage to infection, and coordinate protective responses to restore homeostasis. Like immune cells, sensory neurons exhibit remarkable heterogeneity, with advanced genetic models revealing that distinct subsets differentially regulate immune responses. Here, we review how various immune signals engage distinct subtypes of sensory neurons to mediate inflammatory pain, itch, relief, protective behavioral adaptations, and autonomic reflexes. We also highlight how specialized sensory neuron populations modulate immune function through the release of neuropeptides, neurokines, or glutamate. This functional specialization enables precise immunomodulation adapted to the kinetics and nature of immune responses, positioning sensory neurons as key regulators of host defense and tissue homeostasis.
Collapse
Affiliation(s)
- Théo Crosson
- Aix-Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Ninon Bretaud
- Aix-Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Sophie Ugolini
- Aix-Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France.
| |
Collapse
|
4
|
Franck MCM, Weman HM, Ceder MM, Ahemaiti A, Henriksson K, Bengtsson E, Magnusson KA, Koning HK, Öhman-Mägi C, Lagerström MC. Spinal lumbar Urocortin 3-expressing neurons are associated with both scratching and Compound 48/80-induced sensations. Pain 2025; 166:1070-1087. [PMID: 39432740 PMCID: PMC12004988 DOI: 10.1097/j.pain.0000000000003435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 08/23/2024] [Accepted: 09/02/2024] [Indexed: 10/23/2024]
Abstract
ABSTRACT Urocortin 3 is a neuropeptide that belongs to the corticotropin-releasing hormone family and is involved in mechanosensation and stress regulation. In this study, we show that Urocortin 3 marks a population of excitatory neurons in the mouse spinal cord, divided into 2 nonoverlapping subpopulations expressing protein kinase C gamma or calretinin/calbindin 2, populations previously associated with mechanosensation. Electrophysiological experiments demonstrated that lumbar spinal Urocortin 3 neurons receive both glycinergic and GABAergic local tonic inhibition, and monosynaptic inputs from both Aβ and C fibers, which could be confirmed by retrograde trans-synaptic rabies tracing. Furthermore, fos analyses showed that subpopulations of lumbar Urocortin 3 neurons are activated by artificial scratching or Compound 48/80-induced sensations. Chemogenetic activation of lumbar Urocortin 3-Cre neurons evoked a targeted biting/licking behavior towards the corresponding dermatome and chemogenetic inhibition decreased Compound 48/80-induced behavior. Hence, spinal lumbar Urocortin 3 neurons represent a mechanically associated population with roles in both scratching and Compound 48/80-induced sensations.
Collapse
Affiliation(s)
- Marina C. M. Franck
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Hannah M. Weman
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Mikaela M. Ceder
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Aikeremu Ahemaiti
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Katharina Henriksson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Erica Bengtsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Kajsa A. Magnusson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Harmen K. Koning
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Caroline Öhman-Mägi
- Department of Materials Science and Engineering, Applied Materials Science, Uppsala University, Uppsala, Sweden
| | - Malin C. Lagerström
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
5
|
Hafez OA, Chang RB. Regulation of Cardiac Function by the Autonomic Nervous System. Physiology (Bethesda) 2025; 40:0. [PMID: 39585760 DOI: 10.1152/physiol.00018.2024] [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: 04/05/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024] Open
Abstract
The autonomic nervous system is critical for regulating cardiovascular physiology. The neurocardiac axis encompasses multiple levels of control, including the motor circuits of the sympathetic and parasympathetic nervous systems, sensory neurons that contribute to cardiac reflexes, and the intrinsic cardiac nervous system that provides localized sensing and regulation of the heart. Disruption of these systems can lead to significant clinical conditions. Recent advances have enhanced our understanding of the autonomic control of the heart, detailing the specific neuronal populations involved and their physiologic roles. In this review, we discuss this research at each level of the neurocardiac axis. We conclude by discussing the clinical field of neurocardiology and attempts to translate this new understanding of neurocardiac physiology to the clinic. We highlight the contributions of autonomic dysfunction in prevalent cardiovascular diseases and assess the current status of novel neuroscience-based treatment approaches.
Collapse
Affiliation(s)
- Omar A Hafez
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, United States
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, United States
- M.D.-Ph.D. Program, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Rui B Chang
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, United States
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, United States
| |
Collapse
|
6
|
Qualls KA, Kadakia FK, Serafin EK, Lückemeyer DDN, Davidson S, Strong JA, Zhang JM. mRNA Expression of Mineralocorticoid and Glucocorticoid Receptors in Human and Mouse Sensory Neurons of the Dorsal Root Ganglia. Anesth Analg 2025; 140:1216-1226. [PMID: 39808573 PMCID: PMC11919799 DOI: 10.1213/ane.0000000000007133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
BACKGROUND Corticosteroid receptors, including mineralocorticoid receptor (MR) and glucocorticoid receptor (GR), play important roles in inflammatory pain in the dorsal root ganglion (DRG). Although it is widely known that activating the GR reduces inflammatory pain, it has recently been shown that MR activation contributes to pain and neuronal excitability in rodent studies. Moreover, little is known about the translation of this work to humans, or the mechanisms through which corticosteroid receptors regulate inflammatory pain. METHODS Corticosteroid receptor expression in human and mouse DRGs was characterized. RNAscope was used to perform high-resolution in situ hybridization for GR and MR mRNAs and to examine their colocalization with markers for nociceptors ( SCN10A , Na V 1.8 mRNA) and Aβ mechanoreceptors ( KCNS1 , Kv9.1 mRNA) in human DRG and C57BL/6J mouse DRG samples. RESULTS GR and MR mRNAs are expressed in almost all DRG neurons across species. The 2 receptors colocalize in 99.2% of human DRG neurons and 95.9% of mouse DRG neurons ( P = .0004, Fisher exact test). In both human and mouse DRGs, the large-diameter KCNS1+ Aβ mechanoreceptors showed a significantly higher MR/GR ratio (MR-leaning) compared to KCNS1- neurons (human: 0.23 vs 0.04, P = .0002; mouse: 0.35 vs -0.24, P < .0001; log ratios, unpaired t test), whereas small-diameter SCN10A+ nociceptive neurons showed a significantly lower MR/GR ratio (GR-leaning) compared to SCN10A- neurons (human: -0.02 vs 0.18, P = .0001; mouse: -0.16 vs 0.08, P < .0001; log ratios, unpaired t test). CONCLUSIONS These findings indicate that mouse corticosteroid receptor mRNA expression reflects human expression in the DRG, and that mice could be a suitable model for studying corticosteroid receptor involvement in pain. Additionally, this study supports the translatability of rodent data to humans for the use of more selective corticosteroids at the DRG in pain treatments.
Collapse
MESH Headings
- Ganglia, Spinal/metabolism
- Animals
- Receptors, Mineralocorticoid/genetics
- Receptors, Mineralocorticoid/biosynthesis
- Receptors, Mineralocorticoid/metabolism
- Humans
- Receptors, Glucocorticoid/genetics
- Receptors, Glucocorticoid/biosynthesis
- Receptors, Glucocorticoid/metabolism
- Mice, Inbred C57BL
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Messenger/biosynthesis
- Sensory Receptor Cells/metabolism
- Male
- Mice
- Female
- Middle Aged
- Species Specificity
- Nociceptors/metabolism
- Adult
- NAV1.8 Voltage-Gated Sodium Channel/genetics
- NAV1.8 Voltage-Gated Sodium Channel/metabolism
Collapse
Affiliation(s)
- Katherine A Qualls
- From the Department of Anesthesiology, Pain Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | | | | | | | | | | |
Collapse
|
7
|
Szemes T, Sabaté San José A, Azouz A, Sitte M, Salinas G, Achouri Y, Kricha S, Ris L, Red-Horse K, Bellefroid EJ, Desiderio S. Temporal refinement of Dach1 expression contributes to the development of somatosensory neurons. EMBO J 2025; 44:2882-2905. [PMID: 40205161 DOI: 10.1038/s44318-025-00427-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 04/11/2025] Open
Abstract
During somatosensory neurogenesis, neurons are born in an unspecialized transcriptional state. Several transcription factors in these cells follow a broad-to-restricted expression trajectory as development proceeds, giving rise to neuron subtypes with different identities. The relevance of this temporal refinement of transcription factor expression remains unclear as the functions of transcription factors with broad-to-restricted expression patterns have been mostly studied in those neuron subtypes in which they remain active. Here we show that Dach1 encodes a bona fide transcription factor with a broad-to-restricted expression pattern retained and required in tactile somatosensory neurons. In developing nociceptors, Prdm12 contributes to Dach1 silencing. Using genetic approaches to prevent its temporal restriction during mouse somatosensory development, we reveal that Dach1 expression refinement is a prerequisite for the acquisition of an appropriate transcriptional profile in those somatosensory neuron subtypes in which it becomes ultimately silenced. These findings highlight the essential role played by Dach1 during somatosensory neuron development and demonstrate that the temporal pattern of broad-to-restricted expression followed by several transcription factors is physiologically important for the development of somatosensory neurons.
Collapse
Affiliation(s)
- Tünde Szemes
- Department of Molecular Biology, ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Laboratory of Neuroscience, UMONS Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Alba Sabaté San José
- Department of Molecular Biology, ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Abdulkader Azouz
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), ULB, Brussels, Belgium
| | - Maren Sitte
- NGS Integrative Genomics, Institute of Pathology at the University Medical Center Göttingen (UMG), 37075, Göttingen, Germany
| | - Gabriela Salinas
- NGS Integrative Genomics, Institute of Pathology at the University Medical Center Göttingen (UMG), 37075, Göttingen, Germany
| | - Younes Achouri
- Transgenesis Platform, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Sadia Kricha
- Department of Molecular Biology, ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Laurence Ris
- Laboratory of Neuroscience, UMONS Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford, CA, USA
| | - Eric J Bellefroid
- Department of Molecular Biology, ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium.
| | - Simon Desiderio
- Department of Molecular Biology, ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium.
| |
Collapse
|
8
|
Wang R, Lu Y, Zhao J, Duan X, Chen Y, Zhang Z, Huang R. A combined protocol for isolation, culture, and patch-clamp recording of dorsal root ganglion neurons. BIOPHYSICS REPORTS 2025; 11:87-95. [PMID: 40308938 PMCID: PMC12035743 DOI: 10.52601/bpr.2024.240036] [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: 09/07/2024] [Accepted: 09/25/2024] [Indexed: 05/02/2025] Open
Abstract
The dorsal root ganglion (DRG) neurons are crucial in transmitting sensory information from the peripheral nervous system to the central nervous system, including touch, pain, temperature, and proprioception. Understanding the functions and mechanisms of DRG neurons is essential for studying sensory processing and developing efficient treatments for sensory disorders. In addition, electrophysiological patch-clamp recording is a powerful and classical tool to study the functions and mechanisms of the nervous system. Building upon the strategies outlined in published works and our group's abundant research experience in DRG neurons' functions by patch-clamp, we have summarized and put forward a comprehensive step-by-step protocol combining juvenile rat DRG neuron isolation and culture, and patch-clamp recording. This protocol would be a powerful guidance document for neuroscience researchers to study sensory DRG neurons' physiological and pathological functions using electrophysiological tools.
Collapse
Affiliation(s)
- Ruolin Wang
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710000, China
| | - Yu Lu
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710000, China
| | - Jianbo Zhao
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710000, China
| | - Xueting Duan
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710000, China
| | - Yang Chen
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710000, China
| | - Zhuoyu Zhang
- Neurological Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai 200333, China
| | - Rong Huang
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710000, China
| |
Collapse
|
9
|
Kim JI, Imaizumi K, Jurjuț O, Kelley KW, Wang D, Thete MV, Hudacova Z, Amin ND, Levy RJ, Scherrer G, Pașca SP. Human assembloid model of the ascending neural sensory pathway. Nature 2025:10.1038/s41586-025-08808-3. [PMID: 40205039 DOI: 10.1038/s41586-025-08808-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/19/2025] [Indexed: 04/11/2025]
Abstract
Somatosensory pathways convey crucial information about pain, touch, itch and body part movement from peripheral organs to the central nervous system1,2. Despite substantial needs to understand how these pathways assemble and to develop pain therapeutics, clinical translation remains challenging. This is probably related to species-specific features and the lack of in vitro models of the polysynaptic pathway. Here we established a human ascending somatosensory assembloid (hASA), a four-part assembloid generated from human pluripotent stem cells that integrates somatosensory, spinal, thalamic and cortical organoids to model the spinothalamic pathway. Transcriptomic profiling confirmed the presence of key cell types of this circuit. Rabies tracing and calcium imaging showed that sensory neurons connect to dorsal spinal cord neurons, which further connect to thalamic neurons. Following noxious chemical stimulation, calcium imaging of hASA demonstrated a coordinated response. In addition, extracellular recordings and imaging revealed synchronized activity across the assembloid. Notably, loss of the sodium channel NaV1.7, which causes pain insensitivity, disrupted synchrony across hASA. By contrast, a gain-of-function SCN9A variant associated with extreme pain disorder induced hypersynchrony. These experiments demonstrated the ability to functionally assemble the essential components of the human sensory pathway, which could accelerate our understanding of sensory circuits and facilitate therapeutic development.
Collapse
Affiliation(s)
- Ji-Il Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Kent Imaizumi
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Ovidiu Jurjuț
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Kevin W Kelley
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Dong Wang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Mayuri Vijay Thete
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Zuzana Hudacova
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Neal D Amin
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Rebecca J Levy
- Department of Neurology & Neurological Sciences, Division of Child Neurology, Stanford University, Stanford, CA, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA.
| |
Collapse
|
10
|
Jiang M, Ding Z, Huang Y, Jiang T, Xia Y, Gu D, Gu X, Bai H, Yao D. TGF-β1 Improves Nerve Regeneration and Functional Recovery After Sciatic Nerve Injury by Alleviating Inflammation. Biomedicines 2025; 13:872. [PMID: 40299436 PMCID: PMC12024759 DOI: 10.3390/biomedicines13040872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Peripheral nerves have a certain regenerative ability, but their repair and regeneration after injury is a complex process, usually involving a large number of genes and proteins. In a previous study, we analyzed the gene expression profile in rats after sciatic nerve injury and found significant changes in transforming growth factor-beta 1 (TGF-β1) expression, suggesting that TGF-β1 may be involved in the process of nerve regeneration after injury. Methods: In this study, we first detected the time-course expression and localization of TGF-β1 in dorsal root ganglion (DRG) tissues in a rat sciatic nerve transection model via RT-qPCR. Secondly, we investigated the bioactive roles of TGF-β1 in primary cultured DRG neuron cells through a CCK8 assay, TUNEL assay, and immunofluorescence staining. Thirdly, we explored the neuroprotective roles of TGF-β1 in an in vivo model of sciatic nerve regeneration through morphological observation, behavioral, and electrophysiological tests, and a molecular biological measure. Results: We found that TGF-β1 expression was increased after injury and mainly located in the cytoplasm and nuclei of neuron cells in the DRG. TGF-β1 may regulate the viability, apoptosis, and neurite outgrowth of primary DRG neuron cells. In our in vivo model of sciatic nerve regeneration, TGF-β1 improved nerve regeneration and neuronal function recovery after sciatic nerve injury, alleviated the inflammatory response, and relieved neuropathic pain via the TGF-β1/smad2 pathway. Conclusions: This study provides an experimental and theoretical basis for using TGF-β1 as a neuroprotective agent after peripheral nerve injury in clinical practice in the future.
Collapse
Affiliation(s)
- Maorong Jiang
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, No. 9 Seyuan Road, Nantong 226019, China; (M.J.)
| | - Zihan Ding
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, No. 9 Seyuan Road, Nantong 226019, China; (M.J.)
| | - Yuxiao Huang
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, No. 9 Seyuan Road, Nantong 226019, China; (M.J.)
| | - Taoran Jiang
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, No. 9 Seyuan Road, Nantong 226019, China; (M.J.)
| | - Yiming Xia
- Medical School of Nantong University, Nantong 226001, China
| | - Dandan Gu
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, No. 9 Seyuan Road, Nantong 226019, China; (M.J.)
| | - Xi Gu
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, No. 9 Seyuan Road, Nantong 226019, China; (M.J.)
| | - Huiyuan Bai
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, No. 9 Seyuan Road, Nantong 226019, China; (M.J.)
| | - Dengbing Yao
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, No. 9 Seyuan Road, Nantong 226019, China; (M.J.)
| |
Collapse
|
11
|
Zhu N, Smallwood PM, Williams J, Wang Y, Nathans JH. Utility of binding protein fusions to immunoglobulin heavy chain constant regions from mammalian and avian species. J Biol Chem 2025; 301:108324. [PMID: 39971160 PMCID: PMC11964738 DOI: 10.1016/j.jbc.2025.108324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/21/2025] Open
Abstract
Antibodies are of central importance as reagents for the localization of proteins and other biomolecules in cells and tissues. To expand the repertoire of antibody-based reagents, we have constructed a series of plasmid vectors that permit expression of amino-terminal fusions to the hinge and Fc regions from goat, guinea pig, human, mouse, and rabbit immunoglobulin Gs, and chicken immunogloblin Y. The resulting fusion proteins can be produced in transfected mammalian cells and detected with commercially available and species-specific secondary antibody reagents. We demonstrate the utility of this platform by constructing and testing Fc fusions with DARPin, single-chain Fv, nanobody, toxin, and chemokine partners. The resulting fusion proteins were used to detect their targets in tissue sections or on the surface of transfected cells by immunofluorescent staining or on the surface of immune cells by flow cytometry. By expanding the range of Fc sequences available for fusion protein production, this platform will expand the repertoire of primary antibody reagents for multiplexed immunostaining and fluorescence-activated cell sorting analyses.
Collapse
Affiliation(s)
- Ningyu Zhu
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Philip M Smallwood
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John Williams
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yanshu Wang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeremy H Nathans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
12
|
Erdogan O, Hu XQ, Chiu IM. Sensory neurons on guard: roles in pathogen defense and host immunity. Curr Opin Immunol 2025; 93:102541. [PMID: 40015178 PMCID: PMC11884989 DOI: 10.1016/j.coi.2025.102541] [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: 12/22/2024] [Revised: 02/11/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
The nervous system, like the immune system, constantly interfaces with the environment, encountering threats, including pathogens. Recent discoveries reveal an emerging role for sensory neurons in host defense and immunity. Sensory neurons detect infections either by directly sensing microbial signals or through immune mediators. Beyond pathogen detection, they modulate immune responses and local inflammation by interacting with immune cells, influencing inflammation and pathogen clearance. Additionally, sensory neurons trigger protective reflexes - such as pain, coughing, sneezing, and itching - that can help expel pathogens but may also facilitate their spread. Sensory neurons may also encode and shape long-term immunity. Understanding the roles of neurons in pathogen defense could offer new insights into infectious diseases and highlight therapeutic opportunities for immune modulation.
Collapse
Affiliation(s)
- Ozge Erdogan
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Department of Restorative Dentistry and Biomaterial Sciences, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Xiao-Qian Hu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
13
|
Kheyrollah M, Brandt N, Bräuer AU, Schrader S, Mertsch S. The role of lysophosphatidic acid and its receptors in corneal nerve regeneration. Ocul Surf 2025; 36:10-18. [PMID: 39709127 DOI: 10.1016/j.jtos.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 12/23/2024]
Abstract
The integrity of corneal nerves is critical for ocular surface health, and damages can lead to Neurotrophic Keratopathy (NK). Despite the regenerative abilities of the peripheral nerve system (PNS), corneal nerve regeneration is often incomplete, and the underlying mechanisms are poorly understood. This study aims to identify potential factors that can enhance corneal nerve regeneration for NK treatment, with a focus on Lysophosphatidic acid (LPA). Thus, the effect of LPA and its underlying pathways in nerve regeneration is investigated in detail using in vitro mouse sensory neurons. To elucidate the impact of LPA as well as to reveal the responsible receptor, several functional assays as well as siRNA-based knock-down experiments were conducted. Additionally, possible changes in underlying pathways were investigated on mRNA levels. LPA-treated neurons significantly reduced fiber growth. However, LPAR2 knockdown neurons (Lpar2-KD) following LPA treatment showed a significant increase in fiber length. Additionally, LPA-treated neurons demonstrated enhanced levels of Lpar2 mRNA. On the other hand, nerve regeneration indicators such as Ngf, Gap-43, and Cdc42, along with LPA downstream signaling components like Pi3k and Ras, were elevated in Lpar2-KD neurons. In conclusion, this study elucidates the inhibitory effects of LPA on fiber outgrowth of sensory neurons. Furthermore, LPAR2 was identified as the responsible receptor for the LPA effect. Thus, Lpar2 knockdown might be a promising therapeutic approach to enhance neuronal regeneration in patients with NK.
Collapse
Affiliation(s)
- Maryam Kheyrollah
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany; Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany
| | - Nicola Brandt
- Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany
| | - Anja U Bräuer
- Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany; Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Stefan Schrader
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany; Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Sonja Mertsch
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany.
| |
Collapse
|
14
|
Koh ZM, Arceo RA, Hammer J, Chau K, Light SE, Dolojan A, Januszewski M, Svara F, Smith CJ. An ultrastructural map of a spinal sensorimotor circuit reveals the potential of astroglial modulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.05.641432. [PMID: 40093104 PMCID: PMC11908220 DOI: 10.1101/2025.03.05.641432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Information flow through circuits is dictated by the precise connectivity of neurons and glia. While a single astrocyte can contact many synapses, how glial-synaptic interactions are arranged within a single circuit to impact information flow remains understudied. Here, we use the local spinal sensorimotor circuit in zebrafish as a model to understand how neurons and astroglia are connected in a vertebrate circuit. With semi-automated cellular reconstructions and automated approaches to map all the synaptic connections, we identified the precise synaptic connections of the local sensorimotor circuit, from dorsal root ganglia neurons to spinal interneurons and finally to motor neurons. This revealed a complex network of interneurons that interact in the local sensorimotor circuit. We then mapped the glial processes within tripartite synapses in the circuit. We demonstrate that tripartite synapses are equally distributed across the circuit, supporting the idea that glia can modulate information flow through the circuit at different levels. We show that multiple astroglia, including bona fide astrocytes, contact synapses within a single sensory neuron's circuit and that each of these astroglia can contact multiple parts of the circuit. This detailed map reveals an extensive network of connected neurons and astroglia that process sensory stimuli in a vertebrate. We then utilized this ultrastructural map to model how synaptic thresholding and glial modulation could alter information flow in circuits. We validated this circuit map with GCaMP6s imaging of dorsal root ganglia, spinal neurons and astroglia. This work provides a foundational resource detailing the ultrastructural organization of neurons and glia in a vertebrate circuit, offering insights in how glia could influence information flow in complex neural networks.
Collapse
Affiliation(s)
- Zachary M. Koh
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN
- The Center for Stem Cells and Regenerative Medicine University of Notre Dame, Notre Dame, IN
| | - Ricky Avalos Arceo
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN
- The Center for Stem Cells and Regenerative Medicine University of Notre Dame, Notre Dame, IN
| | - Jacob Hammer
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN
- The Center for Stem Cells and Regenerative Medicine University of Notre Dame, Notre Dame, IN
| | - Khang Chau
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN
- The Center for Stem Cells and Regenerative Medicine University of Notre Dame, Notre Dame, IN
| | - Sarah E.W. Light
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN
- The Center for Stem Cells and Regenerative Medicine University of Notre Dame, Notre Dame, IN
| | - Antonio Dolojan
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN
- The Center for Stem Cells and Regenerative Medicine University of Notre Dame, Notre Dame, IN
| | | | - Fabian Svara
- Zürich, Switzerland. ariadne.ai ag, Buchrain, Switzerland
| | - Cody J. Smith
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN
- The Center for Stem Cells and Regenerative Medicine University of Notre Dame, Notre Dame, IN
| |
Collapse
|
15
|
Silagi ES, Nduka E, Pazyra-Murphy MF, Paiz JZ, Bhuiyan SA, Segal RA. Profiling local translatomes and RNA binding proteins of somatosensory neurons reveals specializations of individual axons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640799. [PMID: 40364912 PMCID: PMC12073832 DOI: 10.1101/2025.02.28.640799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Individual neurons have one or more axons that often extend long distances and traverse multiple microenvironments. However, it is not known how the composition of individual axons is established or locally modulated to enable neuronal function and plasticity. Here, we use spatial translatomics to identify local axonal translatomes in anatomically and functionally specialized neurons in the dorsal root ganglia (DRG). DRG neurons extend long central and peripheral axons in opposite directions and distinct microenvironments to enable somatosensation. Using Translating Ribosome Affinity Purification and RNA sequencing, we generated a comprehensive resource of mRNAs preferentially translated within each axon. Locally translated proteins include pain receptors, ion channels, and translational machinery, which establish distinct electrophysiologic properties and regenerative capacities for each axon. We identify RNA-binding proteins associated with sorting and transporting functionally related mRNAs. These findings provide resources for addressing how axonal translation shapes the spatial organization of neurons and enables subcellular neuroplasticity. HIGHLIGHTS Distinct mRNAs are localized to and translated in individual axons.Axonal translatomes govern regenerative capacity, translational machinery, and electrophysiology.The RBP, SFPQ, coordinates mRNA sorting towards peripheral somatosensory axons.Axonal translatome data can be explored at painseq.shinyapps.io/CompartmentTRAP/.
Collapse
|
16
|
Fernandez A, Sarn N, Eng C, Wright KM. Altered primary somatosensory neuron development in a Pten heterozygous model for autism spectrum disorder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.08.04.552039. [PMID: 37781577 PMCID: PMC10541114 DOI: 10.1101/2023.08.04.552039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder characterized by deficits in social interactions, repetitive behaviors, and hyper- or hyposensitivity to sensory stimuli. The mechanisms underlying the emergence of sensory features in ASD are not fully understood, but recent studies in rodent models highlight that these may result from differences in primary sensory neurons themselves. We examined sensory behaviors in a Pten haploinsufficient mouse model ( Pten Het ) for syndromic ASD and identified elevated responses to mechanical stimuli and a higher threshold to thermal responses. Transcriptomic and in vivo anatomical analysis identified alterations in subtype-specific markers of primary somatosensory neurons in Pten Het dorsal root ganglia (DRG). These defects emerge early during DRG development and involve dysregulation of multiple signaling pathways downstream of Pten . Finally, we show that mice harboring an ASD-associated mutation ( Pten Y69H ) also show altered expression of somatosensory neuron subtype-specific markers. Together, these results show that precise levels of Pten are required for proper somatosensory development and provide insight into the molecular and cellular basis of sensory abnormalities in a model for syndromic ASD.
Collapse
|
17
|
Song K, Kim BS. The peripheral neuroimmune system. J Leukoc Biol 2024; 116:1291-1300. [PMID: 39422243 PMCID: PMC11599120 DOI: 10.1093/jleuko/qiae230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/18/2024] [Accepted: 10/16/2024] [Indexed: 10/19/2024] Open
Abstract
Historically, the nervous and immune systems were studied as separate entities. The nervous system relays signals between the body and the brain by processing sensory inputs and executing motor outputs, whereas the immune system provides protection against injury and infection through inflammation. However, recent developments have demonstrated that these systems mount tightly integrated responses. In particular, the peripheral nervous system acts in concert with the immune system to control reflexes that maintain and restore homeostasis. Notwithstanding their homeostatic mechanisms, dysregulation of these neuroimmune interactions may underlie various pathological conditions. Understanding how these two distinct systems communicate is an emerging field of peripheral neuroimmunology that promises to reveal new insights into tissue physiology and identify novel targets to treat disease.
Collapse
Affiliation(s)
- Keaton Song
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY 10029, USA
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, 787 11th Ave, New York, NY 10019, USA
- Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai, 787 11th Ave, New York, NY 10019, USA
| | - Brian S Kim
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY 10029, USA
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, 787 11th Ave, New York, NY 10019, USA
- Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai, 787 11th Ave, New York, NY 10019, USA
| |
Collapse
|
18
|
Medlock L, Al-Basha D, Halawa A, Dedek C, Ratté S, Prescott SA. Encoding of Vibrotactile Stimuli by Mechanoreceptors in Rodent Glabrous Skin. J Neurosci 2024; 44:e1252242024. [PMID: 39379153 PMCID: PMC11561868 DOI: 10.1523/jneurosci.1252-24.2024] [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: 07/02/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024] Open
Abstract
Somatosensory coding in rodents has been mostly studied in the whisker system and hairy skin, whereas the function of low-threshold mechanoreceptors (LTMRs) in the rodent glabrous skin has received scant attention, unlike in primates where the glabrous skin has been the focus. The relative activation of different LTMR subtypes carries information about vibrotactile stimuli, as does the rate and temporal patterning of LTMR spikes. Rate coding depends on the probability of a spike occurring on each stimulus cycle (reliability), whereas temporal coding depends on the timing of spikes relative to the stimulus cycle (precision). Using in vivo extracellular recordings in male rats and mice of either sex, we measured the reliability and precision of LTMR responses to tactile stimuli including sustained pressure and vibration. Similar to other species, rodent LTMRs were separated into rapid-adapting (RA) or slow-adapting based on their response to sustained pressure. However, unlike the dichotomous frequency preference characteristic of RA1 and RA2/Pacinian afferents in other species, rodent RAs fell along a continuum. Fitting generalized linear models to experimental data reproduced the reliability and precision of rodent RAs. The resulting model parameters highlight key mechanistic differences across the RA spectrum; specifically, the integration window of different RAs transitions from wide to narrow as tuning preferences across the population move from low to high frequencies. Our results show that rodent RAs can support both rate and temporal coding, but their heterogeneity suggests that coactivation patterns play a greater role in population coding than for dichotomously tuned primate RAs.
Collapse
Affiliation(s)
- Laura Medlock
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Dhekra Al-Basha
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Adel Halawa
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Christopher Dedek
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Stéphanie Ratté
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Steven A Prescott
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
19
|
Ben-Shaanan TL, Knöpper K, Duan L, Liu R, Taglinao H, Xu Y, An J, Plikus MV, Cyster JG. Dermal TRPV1 innervations engage a macrophage- and fibroblast-containing pathway to activate hair growth in mice. Dev Cell 2024; 59:2818-2833.e7. [PMID: 38851191 PMCID: PMC11537826 DOI: 10.1016/j.devcel.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/25/2024] [Accepted: 05/15/2024] [Indexed: 06/10/2024]
Abstract
Pain, detected by nociceptors, is an integral part of injury, yet whether and how it can impact tissue physiology and recovery remain understudied. Here, we applied chemogenetics in mice to locally activate dermal TRPV1 innervations in naive skin and found that it triggered new regenerative cycling by dormant hair follicles (HFs). This was preceded by rapid apoptosis of dermal macrophages, mediated by the neuropeptide calcitonin gene-related peptide (CGRP). TRPV1 activation also triggered a macrophage-dependent induction of osteopontin (Spp1)-expressing dermal fibroblasts. The neuropeptide CGRP and the extracellular matrix protein Spp1 were required for the nociceptor-triggered hair growth. Finally, we showed that epidermal abrasion injury induced Spp1-expressing dermal fibroblasts and hair growth via a TRPV1 neuron and CGRP-dependent mechanism. Collectively, these data demonstrated a role for TRPV1 nociceptors in orchestrating a macrophage and fibroblast-supported mechanism to promote hair growth and enabling the efficient restoration of this mechano- and thermo-protective barrier after wounding.
Collapse
Affiliation(s)
- Tamar L Ben-Shaanan
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Konrad Knöpper
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lihui Duan
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ruiqi Liu
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Hanna Taglinao
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ying Xu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jinping An
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
20
|
Navajas Acedo J. Complete persistence of the primary somatosensory system in zebrafish. Dev Biol 2024; 515:178-185. [PMID: 39021074 DOI: 10.1016/j.ydbio.2024.05.004] [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: 02/12/2024] [Revised: 03/20/2024] [Accepted: 05/07/2024] [Indexed: 07/20/2024]
Abstract
The somatosensory system detects peripheral stimuli that are translated into behaviors necessary for survival. Fishes and amphibians possess two somatosensory systems in the trunk: the primary somatosensory system, formed by the Rohon-Beard neurons, and the secondary somatosensory system, formed by the neural crest cell-derived neurons of the Dorsal Root Ganglia. Rohon-Beard neurons have been characterized as a transient population that mostly disappears during the first days of life and is functionally replaced by the Dorsal Root Ganglia. Here, I follow Rohon-Beard neurons in vivo and show that the entire repertoire remains present in zebrafish from 1-day post-fertilization until the juvenile stage, 15-days post-fertilization. These data indicate that zebrafish retain two complete somatosensory systems until at least a developmental stage when the animals display complex behavioral repertoires.
Collapse
Affiliation(s)
- Joaquín Navajas Acedo
- Biozentrum at University of Basel, Spitalstrasse 41, Basel, Switzerland; Allen Discovery Center for Cell Lineage Tracing, University of Washington, Seattle, WA, USA.
| |
Collapse
|
21
|
Fitzgerald M. On the relation of injury to pain-an infant perspective. Pain 2024; 165:S33-S38. [PMID: 39560413 DOI: 10.1097/j.pain.0000000000003366] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/03/2024] [Indexed: 11/20/2024]
Abstract
ABSTRACT Forty-five years ago, Patrick Wall published his John J Bonica lecture "On the relation of injury to pain."90 In this lecture, he argued that pain is better classified as an awareness of a need-state than as a sensation. This need state, he argued, serves more to promote healing than to avoid injury. Here I reframe Wall's prescient proposal to pain in early life and propose a set of different need states that are triggered when injury occurs in infancy. This paper, and my own accompanying Bonica lecture, is dedicated to his memory and to his unique contribution to the neuroscience of pain. The IASP definition of pain includes a key statement, "through their life experiences, individuals learn the concept of pain."69 But the relation between injury and pain is not fixed from birth. In early life, the links between nociception (the sense) and pain (the need state) are very different from those of adults, although no less important. I propose that injury evokes three pain need states in infancy, all of which depend on the state of maturity of the central nervous system: (1) the need to attract maternal help; (2) the need to learn the concept of pain; and (3) the need to maintain healthy activity dependent brain development.
Collapse
Affiliation(s)
- Maria Fitzgerald
- Department of Neuroscience, Physiology & Pharamcology, University College London, London, United Kingdom
| |
Collapse
|
22
|
Hayward R, Moore S, Artun D, Madhavan A, Harte E, Torres-Pérez JV, Nagy I. Transcriptional reprogramming post-peripheral nerve injury: A systematic review. Neurobiol Dis 2024; 200:106624. [PMID: 39097036 DOI: 10.1016/j.nbd.2024.106624] [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: 06/28/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024] Open
Abstract
Neuropathic pain is characterised by periodic or continuous hyperalgesia, numbness, or allodynia, and results from insults to the somatosensory nervous system. Peripheral nerve injury induces transcriptional reprogramming in peripheral sensory neurons, contributing to increased spinal nociceptive input and the development of neuropathic pain. Effective treatment for neuropathic pain remains an unmet medical need as current therapeutics offer limited effectiveness and have undesirable effects. Understanding transcriptional changes in peripheral nerve injury-induced neuropathy might offer a path for novel analgesics. Our literature search identified 65 papers exploring transcriptomic changes post-peripheral nerve injury, many of which were conducted in animal models. We scrutinize their transcriptional changes data and conduct gene ontology enrichment analysis to reveal their common functional profile. Focusing on genes involved in 'sensory perception of pain' (GO:0019233), we identified transcriptional changes for different ion channels, receptors, and neurotransmitters, shedding light on its role in nociception. Examining peripheral sensory neurons subtype-specific transcriptional reprograming and regeneration-associated genes, we delved into downstream regulation of hypersensitivity. Identifying the temporal program of transcription regulatory mechanisms might help develop better therapeutics to target them effectively and selectively, thus preventing the development of neuropathic pain without affecting other physiological functions.
Collapse
Affiliation(s)
- R Hayward
- Nociception Group, Department of Surgery and Cancer, Division of Anaesthetics, Pain Medicine and Intensive Care, Chelsea and Westminster Hospital Campus, Imperial College London, 369 Fulham Road, London SW10 9FJ, UK
| | - S Moore
- Nociception Group, Department of Surgery and Cancer, Division of Anaesthetics, Pain Medicine and Intensive Care, Chelsea and Westminster Hospital Campus, Imperial College London, 369 Fulham Road, London SW10 9FJ, UK
| | - D Artun
- Nociception Group, Department of Surgery and Cancer, Division of Anaesthetics, Pain Medicine and Intensive Care, Chelsea and Westminster Hospital Campus, Imperial College London, 369 Fulham Road, London SW10 9FJ, UK
| | - A Madhavan
- Nociception Group, Department of Surgery and Cancer, Division of Anaesthetics, Pain Medicine and Intensive Care, Chelsea and Westminster Hospital Campus, Imperial College London, 369 Fulham Road, London SW10 9FJ, UK
| | - E Harte
- Nociception Group, Department of Surgery and Cancer, Division of Anaesthetics, Pain Medicine and Intensive Care, Chelsea and Westminster Hospital Campus, Imperial College London, 369 Fulham Road, London SW10 9FJ, UK
| | - J V Torres-Pérez
- Departament de Biologia Cel·lular, Biologia Funcional i Antropologia Física, Facultat de Ciències Biològiques, Universitat de València, C/Dr. Moliner 50, 46100 Burjassot, Spain.
| | - I Nagy
- Nociception Group, Department of Surgery and Cancer, Division of Anaesthetics, Pain Medicine and Intensive Care, Chelsea and Westminster Hospital Campus, Imperial College London, 369 Fulham Road, London SW10 9FJ, UK.
| |
Collapse
|
23
|
Chiu IM, Sokol CL. Neuroimmune recognition of allergens. Curr Opin Immunol 2024; 90:102458. [PMID: 39213825 PMCID: PMC11423315 DOI: 10.1016/j.coi.2024.102458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/07/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Interactions between the nervous system and the immune system play crucial roles in initiating and directing the type 2 immune response. Sensory neurons can initiate innate and adaptive type 2 immunity through their ability to detect allergens and promote dendritic cell and mast cell responses. Neurons also indirectly promote type 2 inflammation through suppression of type 1 immune responses. Type 2 cytokines promote neuronal function by directly activating or sensitizing neurons. This positive neuroimmune feedback loop may not only enhance allergic inflammation but also promote the system-wide responses of aversion, anaphylaxis, and allergen polysensitization that are characteristic of allergic immunity.
Collapse
Affiliation(s)
- Isaac M Chiu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| | - Caroline L Sokol
- Center for Immunology & Inflammatory Diseases, Division of Rheumatology, Allergy & Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
24
|
Singh K, Jayaram M, Hanumantharaju A, Tõnissoo T, Jagomäe T, Mikheim K, Muthuraman S, Gilbert SF, Plaas M, Schäfer MKE, Innos J, Lilleväli K, Philips MA, Vasar E. The IgLON family of cell adhesion molecules expressed in developing neural circuits ensure the proper functioning of the sensory system in mice. Sci Rep 2024; 14:22593. [PMID: 39349721 PMCID: PMC11442611 DOI: 10.1038/s41598-024-73358-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024] Open
Abstract
Deletions and malfunctions of the IgLON family of cell adhesion molecules are associated with anatomical, behavioral, and metabolic manifestations of neuropsychiatric disorders. We have previously shown that IgLON genes are expressed in sensory nuclei/pathways and that IgLON proteins modulate sensory processing. Here, we examined the expression of IgLON alternative promoter-specific isoforms during embryonic development and studied the sensory consequences of the anatomical changes when one of the IgLON genes, Negr1, is knocked out. At the embryonal age of E12.5 and E13.5, various IgLONs were distributed differentially and dynamically in the developing sensory areas within the central and peripheral nervous system, as well as in limbs and mammary glands. Sensory tests showed that Negr1 deficiency causes differences in vestibular function and temperature sensitivity in the knockout mice. Sex-specific differences were noted across olfaction, vestibular functioning, temperature regulation, and mechanical sensitivity. Our findings highlight the involvement of IgLON molecules during sensory circuit formation and suggest Negr1's critical role in somatosensory processing.
Collapse
Affiliation(s)
- Katyayani Singh
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia.
| | - Mohan Jayaram
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Arpana Hanumantharaju
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Tambet Tõnissoo
- Institute of Molecular and Cell Biology, University of Tartu, Vanemuise 46-221, Ria 23-204, 51010, Tartu, Estonia
| | - Toomas Jagomäe
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411, Tartu, Estonia
| | - Kaie Mikheim
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Srirathi Muthuraman
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Scott F Gilbert
- Department of Biology, Swarthmore College, Swarthmore, PA, USA
| | - Mario Plaas
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411, Tartu, Estonia
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, 55131, Mainz, Germany
- Focus Program Translational Neurosciences, Johannes Gutenberg-University Mainz, 55131, Mainz, Germany
- Research Center for Immunotherapy, Johannes Gutenberg-University Mainz, 55131, Mainz, Germany
| | - Jürgen Innos
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Kersti Lilleväli
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
- The Centre of Estonian Rural Research and Knowledge, 48309, Jõgeva Alevik, Estonia
| | - Mari-Anne Philips
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| |
Collapse
|
25
|
Pham QTT, Weng HJ. Uncovering the missing mutations in pain. Pain 2024; 165:1901-1903. [PMID: 39159470 DOI: 10.1097/j.pain.0000000000003253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 03/12/2024] [Indexed: 08/21/2024]
Affiliation(s)
- Quoc Thao Trang Pham
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Dermatology, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Hao-Jui Weng
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Dermatology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
- Department of Dermatology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan
| |
Collapse
|
26
|
Cowan ZL, Green L, Clark TD, Blewett TA, De Bonville J, Gagnon T, Hoots E, Kuchenmüller L, Leeuwis RHJ, Navajas Acedo J, Rowsey LE, Scheuffele H, Skeeles MR, Silva-Garay L, Jutfelt F, Binning SA. Global change and premature hatching of aquatic embryos. GLOBAL CHANGE BIOLOGY 2024; 30:e17488. [PMID: 39238185 DOI: 10.1111/gcb.17488] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 09/07/2024]
Abstract
Anthropogenically induced changes to the natural world are increasingly exposing organisms to stimuli and stress beyond that to which they are adapted. In aquatic systems, it is thought that certain life stages are more vulnerable than others, with embryos being flagged as highly susceptible to environmental stressors. Interestingly, evidence from across a wide range of taxa suggests that aquatic embryos can hatch prematurely, potentially as an adaptive response to external stressors, despite the potential for individual costs linked with underdeveloped behavioural and/or physiological functions. However, surprisingly little research has investigated the prevalence, causes and consequences of premature hatching, and no compilation of the literature exists. Here, we review what is known about premature hatching in aquatic embryos and discuss how this phenomenon is likely to become exacerbated with anthropogenically induced global change. Specifically, we (1) review the mechanisms of hatching, including triggers for premature hatching in experimental and natural systems; (2) discuss the potential implications of premature hatching at different levels of biological organisation from individuals to ecosystems; and (3) outline knowledge gaps and future research directions for understanding the drivers and consequences of premature hatching. We found evidence that aquatic embryos can hatch prematurely in response to a broad range of abiotic (i.e. temperature, oxygen, toxicants, light, pH, salinity) and biotic (i.e. predators, pathogens) stressors. We also provide empirical evidence that premature hatching appears to be a common response to rapid thermal ramping across fish species. We argue that premature hatching represents a fascinating yet untapped area of study, and the phenomenon may provide some additional resilience to aquatic communities in the face of ongoing global change.
Collapse
Affiliation(s)
- Zara-Louise Cowan
- Natural Resources Institute Finland (Luke), Oulu, Finland
- Department of Biology, Faculty of Natural Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Leon Green
- Department of Biology and Environmental Sciences, Faculty of Natural Sciences, Kristineberg Center, University of Gothenburg, Fiskebäckskil, Sweden
| | - Timothy D Clark
- School of Life and Environmental Sciences, Deakin University, Geelong, Victoria, Australia
| | - Tamzin A Blewett
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Jeremy De Bonville
- Département de Sciences Biologiques, Université de Montréal, Montreal, Quebec, Canada
| | - Thomas Gagnon
- Département de Sciences Biologiques, Université de Montréal, Montreal, Quebec, Canada
| | - Elizabeth Hoots
- School of Life and Environmental Sciences, Deakin University, Geelong, Victoria, Australia
| | - Luis Kuchenmüller
- School of Life and Environmental Sciences, Deakin University, Geelong, Victoria, Australia
| | - Robine H J Leeuwis
- Department of Biology, Faculty of Natural Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Lauren E Rowsey
- Department of Biological Sciences, University of New Brunswick, Saint John, New Brunswick, Canada
| | - Hanna Scheuffele
- School of Life and Environmental Sciences, Deakin University, Geelong, Victoria, Australia
| | | | - Lorena Silva-Garay
- Department of Biology, Faculty of Natural Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Fredrik Jutfelt
- Department of Biology, Faculty of Natural Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Biological and Environmental Sciences, Faculty of Science, University of Gothenburg, Gothenburg, Sweden
| | - Sandra A Binning
- Département de Sciences Biologiques, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
27
|
Desbois M, Grill B. Molecular regulation of axon termination in mechanosensory neurons. Development 2024; 151:dev202945. [PMID: 39268828 PMCID: PMC11698068 DOI: 10.1242/dev.202945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Spatially and temporally accurate termination of axon outgrowth, a process called axon termination, is required for efficient, precise nervous system construction and wiring. The mechanosensory neurons that sense low-threshold mechanical stimulation or gentle touch have proven exceptionally valuable for studying axon termination over the past 40 years. In this Review, we discuss progress made in deciphering the molecular and genetic mechanisms that govern axon termination in touch receptor neurons. Findings across model organisms, including Caenorhabditis elegans, Drosophila, zebrafish and mice, have revealed that complex signaling is required for termination with conserved principles and players beginning to surface. A key emerging theme is that axon termination is mediated by complex signaling networks that include ubiquitin ligase signaling hubs, kinase cascades, transcription factors, guidance/adhesion receptors and growth factors. Here, we begin a discussion about how these signaling networks could represent termination codes that trigger cessation of axon outgrowth in different species and types of mechanosensory neurons.
Collapse
Affiliation(s)
- Muriel Desbois
- School of Life Sciences, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - Brock Grill
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98101, USA
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98101, USA
| |
Collapse
|
28
|
Zhang ZX, Tian Y, Li S, Jing HB, Cai J, Li M, Xing GG. Involvement of HDAC2-mediated kcnq2/kcnq3 genes transcription repression activated by EREG/EGFR-ERK-Runx1 signaling in bone cancer pain. Cell Commun Signal 2024; 22:416. [PMID: 39192337 PMCID: PMC11350972 DOI: 10.1186/s12964-024-01797-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/18/2024] [Indexed: 08/29/2024] Open
Abstract
Bone cancer pain (BCP) represents a prevalent symptom among cancer patients with bone metastases, yet its underlying mechanisms remain elusive. This study investigated the transcriptional regulation mechanism of Kv7(KCNQ)/M potassium channels in DRG neurons and its involvement in the development of BCP in rats. We show that HDAC2-mediated transcriptional repression of kcnq2/kcnq3 genes, which encode Kv7(KCNQ)/M potassium channels in dorsal root ganglion (DRG), contributes to the sensitization of DRG neurons and the pathogenesis of BCP in rats. Also, HDAC2 requires the formation of a corepressor complex with MeCP2 and Sin3A to execute transcriptional regulation of kcnq2/kcnq3 genes. Moreover, EREG is identified as an upstream signal molecule for HDAC2-mediated kcnq2/kcnq3 genes transcription repression. Activation of EREG/EGFR-ERK-Runx1 signaling, followed by the induction of HDAC2-mediated transcriptional repression of kcnq2/kcnq3 genes in DRG neurons, leads to neuronal hyperexcitability and pain hypersensitivity in tumor-bearing rats. Consequently, the activation of EREG/EGFR-ERK-Runx1 signaling, along with the subsequent transcriptional repression of kcnq2/kcnq3 genes by HDAC2 in DRG neurons, underlies the sensitization of DRG neurons and the pathogenesis of BCP in rats. These findings uncover a potentially targetable mechanism contributing to bone metastasis-associated pain in cancer patients.
Collapse
Affiliation(s)
- Zi-Xian Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center and Neuroscience Research Institute, Peking University, Beijing, China
| | - Yue Tian
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center and Neuroscience Research Institute, Peking University, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education of China & National Health Commission of China, Beijing, 100191, China
| | - Song Li
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center and Neuroscience Research Institute, Peking University, Beijing, China
| | - Hong-Bo Jing
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center and Neuroscience Research Institute, Peking University, Beijing, China
| | - Jie Cai
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center and Neuroscience Research Institute, Peking University, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education of China & National Health Commission of China, Beijing, 100191, China
| | - Min Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, 100191, China.
| | - Guo-Gang Xing
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center and Neuroscience Research Institute, Peking University, Beijing, China.
- Key Laboratory for Neuroscience, Ministry of Education of China & National Health Commission of China, Beijing, 100191, China.
| |
Collapse
|
29
|
LeBlang CJ, Pazyra-Murphy MF, Silagi ES, Dasgupta S, Tsolias M, Miller T, Petrova V, Zhen S, Jovanovic V, Castellano D, Gerrish K, Ormanoglu P, Tristan C, Singeç I, Woolf CJ, Tasdemir-Yilmaz O, Segal RA. Satellite glial contact enhances differentiation and maturation of human iPSC-derived sensory neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604966. [PMID: 39211268 PMCID: PMC11361066 DOI: 10.1101/2024.07.24.604966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Sensory neurons generated from induced pluripotent stem cells (iSNs) are used to model human peripheral neuropathies, however current differentiation protocols produce sensory neurons with an embryonic phenotype. Peripheral glial cells contact sensory neurons early in development and contribute to formation of the canonical pseudounipolar morphology, but these signals are not encompassed in current iSN differentiation protocols. Here, we show that terminal differentiation of iSNs in co-culture with rodent Dorsal Root Ganglion satellite glia (rSG) advances their differentiation and maturation. Co-cultured iSNs develop a pseudounipolar morphology through contact with rSGs. This transition depends on semaphorin-plexin guidance cues and on glial gap junction signaling. In addition to morphological changes, iSNs terminally differentiated in co-culture exhibit enhanced spontaneous action potential firing, more mature gene expression, and increased susceptibility to paclitaxel induced axonal degeneration. Thus, iSNs differentiated in coculture with rSGs provide a better model for investigating human peripheral neuropathies.
Collapse
|
30
|
Kantarci H, Elvira PD, Thottumkara AP, O'Connell EM, Iyer M, Donovan LJ, Dugan MQ, Ambiel N, Granados A, Zeng H, Saw NL, Brosius Lutz A, Sloan SA, Gray EE, Tran KV, Vichare A, Yeh AK, Münch AE, Huber M, Agrawal A, Morri M, Zhong H, Shamloo M, Anderson TA, Tawfik VL, Du Bois J, Zuchero JB. Schwann cell-secreted PGE 2 promotes sensory neuron excitability during development. Cell 2024; 187:4690-4712.e30. [PMID: 39142281 PMCID: PMC11967275 DOI: 10.1016/j.cell.2024.07.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/18/2024] [Accepted: 06/21/2024] [Indexed: 08/16/2024]
Abstract
Electrical excitability-the ability to fire and propagate action potentials-is a signature feature of neurons. How neurons become excitable during development and whether excitability is an intrinsic property of neurons remain unclear. Here, we demonstrate that Schwann cells, the most abundant glia in the peripheral nervous system, promote somatosensory neuron excitability during development. We find that Schwann cells secrete prostaglandin E2, which is necessary and sufficient to induce developing somatosensory neurons to express normal levels of genes required for neuronal function, including voltage-gated sodium channels, and to fire action potential trains. Inactivating this signaling pathway in Schwann cells impairs somatosensory neuron maturation, causing multimodal sensory defects that persist into adulthood. Collectively, our studies uncover a neurodevelopmental role for prostaglandin E2 distinct from its established role in inflammation, revealing a cell non-autonomous mechanism by which glia regulate neuronal excitability to enable the development of normal sensory functions.
Collapse
Affiliation(s)
- Husniye Kantarci
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Pablo D Elvira
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | | | - Emma M O'Connell
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Manasi Iyer
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lauren J Donovan
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Micaela Quinn Dugan
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nicholas Ambiel
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Hong Zeng
- Transgenic, Knockout and Tumor model Center (TKTC), Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nay L Saw
- Behavioral and Functional Neuroscience Laboratory, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amanda Brosius Lutz
- Department of Obstetrics and Gynecology, University Hospital, Bern, Switzerland
| | - Steven A Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Erin E Gray
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Khanh V Tran
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aditi Vichare
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ashley K Yeh
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alexandra E Münch
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Max Huber
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Aditi Agrawal
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | | | - Haining Zhong
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Mehrdad Shamloo
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Behavioral and Functional Neuroscience Laboratory, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Thomas Anthony Anderson
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Vivianne L Tawfik
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - J Du Bois
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA.
| | - J Bradley Zuchero
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
31
|
Koyanagi M, Ogido R, Moriya A, Saigo M, Ihida S, Teranishi T, Kawada J, Katsuno T, Matsubara K, Terada T, Yamashita A, Imai S. Development of a 3-dimensional organotypic model with characteristics of peripheral sensory nerves. CELL REPORTS METHODS 2024; 4:100835. [PMID: 39116883 PMCID: PMC11384078 DOI: 10.1016/j.crmeth.2024.100835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/02/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024]
Abstract
We developed a rat dorsal root ganglion (DRG)-derived sensory nerve organotypic model by culturing DRG explants on an organoid culture device. With this method, a large number of organotypic cultures can be produced simultaneously with high reproducibility simply by seeding DRG explants derived from rat embryos. Unlike previous DRG explant models, this organotypic model consists of a ganglion and an axon bundle with myelinated A fibers, unmyelinated C fibers, and stereo-myelin-forming nodes of Ranvier. The model also exhibits Ca2+ signaling in cell bodies in response to application of chemical stimuli to nerve terminals. Further, axonal transection increases the activating transcription factor 3 mRNA level in ganglia. Axons and myelin are shown to regenerate 14 days following transection. Our sensory organotypic model enables analysis of neuronal excitability in response to pain stimuli and tracking of morphological changes in the axon bundle over weeks.
Collapse
Affiliation(s)
- Madoka Koyanagi
- Department of Medical Neuropharmacology, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 640-8156, Japan
| | - Ryosuke Ogido
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Akari Moriya
- Department of Clinical Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Mamiko Saigo
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto 606-8507, Japan
| | - Satoshi Ihida
- New Business Promotion Division, Business Development Unit, Panel Semicon Laboratories, Sharp Corporation, Tenri, Nara 632-8567, Japan
| | - Tomoko Teranishi
- New Business Promotion Division, Business Development Unit, Panel Semicon Laboratories, Sharp Corporation, Tenri, Nara 632-8567, Japan
| | - Jiro Kawada
- Jiksak Bioengineering, Inc., Kawasaki, Kanagawa 210-0821, Japan
| | - Tatsuya Katsuno
- Division of Electron Microscopic Study, Center for Anatomical Studies, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Kazuo Matsubara
- School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 640-8156, Japan
| | - Tomohiro Terada
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto 606-8507, Japan
| | - Akira Yamashita
- Department of Medical Neuropharmacology, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 640-8156, Japan
| | - Satoshi Imai
- Department of Medical Neuropharmacology, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 640-8156, Japan; Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto 606-8507, Japan.
| |
Collapse
|
32
|
Baltar J, Miranda RM, Cabral M, Rebelo S, Grahammer F, Huber TB, Reguenga C, Monteiro FA. Neph1 is required for neurite branching and is negatively regulated by the PRRXL1 homeodomain factor in the developing spinal cord dorsal horn. Neural Dev 2024; 19:13. [PMID: 39049046 PMCID: PMC11271021 DOI: 10.1186/s13064-024-00190-6] [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: 05/16/2024] [Accepted: 07/05/2024] [Indexed: 07/27/2024] Open
Abstract
The cell-adhesion molecule NEPH1 is required for maintaining the structural integrity and function of the glomerulus in the kidneys. In the nervous system of Drosophila and C. elegans, it is involved in synaptogenesis and axon branching, which are essential for establishing functional circuits. In the mammalian nervous system, the expression regulation and function of Neph1 has barely been explored. In this study, we provide a spatiotemporal characterization of Neph1 expression in mouse dorsal root ganglia (DRGs) and spinal cord. After the neurogenic phase, Neph1 is broadly expressed in the DRGs and in their putative targets at the dorsal horn of the spinal cord, comprising both GABAergic and glutamatergic neurons. Interestingly, we found that PRRXL1, a homeodomain transcription factor that is required for proper establishment of the DRG-spinal cord circuit, prevents a premature expression of Neph1 in the superficial laminae of the dorsal spinal cord at E14.5, but has no regulatory effect on the DRGs or on either structure at E16.5. By chromatin immunoprecipitation analysis of the dorsal spinal cord, we identified four PRRXL1-bound regions within the Neph1 introns, suggesting that PRRXL1 directly regulates Neph1 transcription. We also showed that Neph1 is required for branching, especially at distal neurites. Together, our work showed that Prrxl1 prevents the early expression of Neph1 in the superficial dorsal horn, suggesting that Neph1 might function as a downstream effector gene for proper assembly of the DRG-spinal nociceptive circuit.
Collapse
Affiliation(s)
- João Baltar
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Rafael Mendes Miranda
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Maria Cabral
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Sandra Rebelo
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Departamento de Patologia Clínica, Centro Hospitalar Universitário São João, Porto, Portugal
| | - Florian Grahammer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carlos Reguenga
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Filipe Almeida Monteiro
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal.
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal.
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
33
|
Xu M, Thottappillil N, Cherief M, Li Z, Zhu M, Xing X, Gomez-Salazar M, Mwirigi JM, Sankaranarayanan I, Tavares-Ferreira D, Zhang C, Wang XW, Archer M, Guan Y, Tower RJ, Cahan P, Price TJ, Clemens TL, James AW. Mapping Somatosensory Afferent Circuitry to Bone Identifies Neurotrophic Signals Required for Fracture Healing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597786. [PMID: 38895367 PMCID: PMC11185682 DOI: 10.1101/2024.06.06.597786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The profound pain accompanying bone fracture is mediated by somatosensory neurons, which also appear to be required to initiate bone regeneration following fracture. Surprisingly, the precise neuroanatomical circuitry mediating skeletal nociception and regeneration remains incompletely understood. Here, we characterized somatosensory dorsal root ganglia (DRG) afferent neurons innervating murine long bones before and after experimental long bone fracture in mice. Retrograde labeling of DRG neurons by an adeno-associated virus with peripheral nerve tropism showed AAV-tdT signal. Single cell transcriptomic profiling of 6,648 DRG neurons showed highest labeling across CGRP+ neuron clusters (6.9-17.2%) belonging to unmyelinated C fibers, thinly myelinated Aδ fibers and Aβ-Field LTMR (9.2%). Gene expression profiles of retrograde labeled DRG neurons over multiple timepoints following experimental stress fracture revealed dynamic changes in gene expression corresponding to the acute inflammatory ( S100a8 , S100a9 ) and mechanical force ( Piezo2 ). Reparative phase after fracture included morphogens such as Tgfb1, Fgf9 and Fgf18 . Two methods to surgically or genetically denervate fractured bones were used in combination with scRNA-seq to implicate defective mesenchymal cell proliferation and osteodifferentiation as underlying the poor bone repair capacity in the presence of attenuated innervation. Finally, multi-tissue scRNA-seq and interactome analyses implicated neuron-derived FGF9 as a potent regulator of fracture repair, a finding compatible with in vitro assessments of neuron-to-skeletal mesenchyme interactions.
Collapse
|
34
|
Heiman MG, Bülow HE. Dendrite morphogenesis in Caenorhabditis elegans. Genetics 2024; 227:iyae056. [PMID: 38785371 PMCID: PMC11151937 DOI: 10.1093/genetics/iyae056] [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: 12/18/2023] [Accepted: 04/02/2024] [Indexed: 05/25/2024] Open
Abstract
Since the days of Ramón y Cajal, the vast diversity of neuronal and particularly dendrite morphology has been used to catalog neurons into different classes. Dendrite morphology varies greatly and reflects the different functions performed by different types of neurons. Significant progress has been made in our understanding of how dendrites form and the molecular factors and forces that shape these often elaborately sculpted structures. Here, we review work in the nematode Caenorhabditis elegans that has shed light on the developmental mechanisms that mediate dendrite morphogenesis with a focus on studies investigating ciliated sensory neurons and the highly elaborated dendritic trees of somatosensory neurons. These studies, which combine time-lapse imaging, genetics, and biochemistry, reveal an intricate network of factors that function both intrinsically in dendrites and extrinsically from surrounding tissues. Therefore, dendrite morphogenesis is the result of multiple tissue interactions, which ultimately determine the shape of dendritic arbors.
Collapse
Affiliation(s)
- Maxwell G Heiman
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Hannes E Bülow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
35
|
Weman HM, Ceder MM, Ahemaiti A, Magnusson KA, Henriksson K, Andréasson L, Lagerström MC. Spinal Glycine Receptor Alpha 3 Cells Communicate Sensations of Chemical Itch in Hairy Skin. J Neurosci 2024; 44:e1585232024. [PMID: 38553047 PMCID: PMC11079978 DOI: 10.1523/jneurosci.1585-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 01/31/2024] [Accepted: 02/15/2024] [Indexed: 05/12/2024] Open
Abstract
Glycinergic neurons regulate nociceptive and pruriceptive signaling in the spinal cord, but the identity and role of the glycine-regulated neurons are not fully known. Herein, we have characterized spinal glycine receptor alpha 3 (Glra3) subunit-expressing neurons in Glra3-Cre female and male mice. Glra3-Cre(+) neurons express Glra3, are located mainly in laminae III-VI, and respond to glycine. Chemogenetic activation of spinal Glra3-Cre(+) neurons induced biting/licking, stomping, and guarding behaviors, indicative of both a nociceptive and pruriceptive role for this population. Chemogenetic inhibition did not affect mechanical or thermal responses but reduced behaviors evoked by compound 48/80 and chloroquine, revealing a pruriceptive role for these neurons. Spinal cells activated by compound 48/80 or chloroquine express Glra3, further supporting the phenotype. Retrograde tracing revealed that spinal Glra3-Cre(+) neurons receive input from afferents associated with pain and itch, and dorsal root stimulation validated the monosynaptic input. In conclusion, these results show that spinal Glra3(+) neurons contribute to acute communication of compound 48/80- and chloroquine-induced itch in hairy skin.
Collapse
Affiliation(s)
- Hannah M Weman
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75108, Sweden
| | - Mikaela M Ceder
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75108, Sweden
| | - Aikeremu Ahemaiti
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75108, Sweden
| | - Kajsa A Magnusson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75108, Sweden
| | - Katharina Henriksson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75108, Sweden
| | - Linn Andréasson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75108, Sweden
| | - Malin C Lagerström
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75108, Sweden
| |
Collapse
|
36
|
George DS, Jayaraj ND, Pacifico P, Ren D, Sriram N, Miller RE, Malfait AM, Miller RJ, Menichella DM. The Mas-related G protein-coupled receptor d (Mrgprd) mediates pain hypersensitivity in painful diabetic neuropathy. Pain 2024; 165:1154-1168. [PMID: 38147415 PMCID: PMC11017747 DOI: 10.1097/j.pain.0000000000003120] [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: 04/18/2023] [Revised: 08/22/2023] [Accepted: 08/22/2023] [Indexed: 12/28/2023]
Abstract
ABSTRACT Painful diabetic neuropathy (PDN) is one of the most common and intractable complications of diabetes. Painful diabetic neuropathy is characterized by neuropathic pain accompanied by dorsal root ganglion (DRG) nociceptor hyperexcitability, axonal degeneration, and changes in cutaneous innervation. However, the complete molecular profile underlying the hyperexcitable cellular phenotype of DRG nociceptors in PDN has not been elucidated. This gap in our knowledge is a critical barrier to developing effective, mechanism-based, and disease-modifying therapeutic approaches that are urgently needed to relieve the symptoms of PDN. Using single-cell RNA sequencing of DRGs, we demonstrated an increased expression of the Mas-related G protein-coupled receptor d (Mrgprd) in a subpopulation of DRG neurons in the well-established high-fat diet (HFD) mouse model of PDN. Importantly, limiting Mrgprd signaling reversed mechanical allodynia in the HFD mouse model of PDN. Furthermore, in vivo calcium imaging allowed us to demonstrate that activation of Mrgprd-positive cutaneous afferents that persist in diabetic mice skin resulted in an increased intracellular calcium influx into DRG nociceptors that we assess in vivo as a readout of nociceptors hyperexcitability. Taken together, our data highlight a key role of Mrgprd-mediated DRG neuron excitability in the generation and maintenance of neuropathic pain in a mouse model of PDN. Hence, we propose Mrgprd as a promising and accessible target for developing effective therapeutics currently unavailable for treating neuropathic pain in PDN.
Collapse
Affiliation(s)
| | | | | | - Dongjun Ren
- Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | | | - Rachel E. Miller
- Department of Internal Medicine, Rush Medical College, Chicago, IL, United States
| | - Anne-Marie Malfait
- Department of Internal Medicine, Rush Medical College, Chicago, IL, United States
| | - Richard J. Miller
- Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Daniela Maria Menichella
- Departments of Neurology and
- Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
37
|
Kim JH, Cetinkaya-Fisgin A, Zahn N, Sari MC, Hoke A, Barman I. Label-Free Visualization and Morphological Profiling of Neuronal Differentiation and Axonal Degeneration through Quantitative Phase Imaging. Adv Biol (Weinh) 2024; 8:e2400020. [PMID: 38548657 PMCID: PMC11090721 DOI: 10.1002/adbi.202400020] [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: 03/13/2024] [Indexed: 05/15/2024]
Abstract
Understanding the intricate processes of neuronal growth, degeneration, and neurotoxicity is paramount for unraveling nervous system function and holds significant promise in improving patient outcomes, especially in the context of chemotherapy-induced peripheral neuropathy (CIPN). These processes are influenced by a broad range of entwined events facilitated by chemical, electrical, and mechanical signals. The progress of each process is inherently linked to phenotypic changes in cells. Currently, the primary means of demonstrating morphological changes rely on measurements of neurite outgrowth and axon length. However, conventional techniques for monitoring these processes often require extensive preparation to enable manual or semi-automated measurements. Here, a label-free and non-invasive approach is employed for monitoring neuronal differentiation and degeneration using quantitative phase imaging (QPI). Operating on unlabeled specimens and offering little to no phototoxicity and photobleaching, QPI delivers quantitative maps of optical path length delays that provide an objective measure of cellular morphology and dynamics. This approach enables the visualization and quantification of axon length and other physical properties of dorsal root ganglion (DRG) neuronal cells, allowing greater understanding of neuronal responses to stimuli simulating CIPN conditions. This research paves new avenues for the development of more effective strategies in the clinical management of neurotoxicity.
Collapse
Affiliation(s)
- Jeong Hee Kim
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Aysel Cetinkaya-Fisgin
- Department of Neurology, Neuromuscular Division, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Noah Zahn
- Department Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Mehmet Can Sari
- Department of Neurology, Neuromuscular Division, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ahmet Hoke
- Department of Neurology, Neuromuscular Division, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Oncology, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
38
|
Dominguez-Bajo A, Clotman F. Potential Roles of Specific Subclasses of Premotor Interneurons in Spinal Cord Function Recovery after Traumatic Spinal Cord Injury in Adults. Cells 2024; 13:652. [PMID: 38667267 PMCID: PMC11048910 DOI: 10.3390/cells13080652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
The differential expression of transcription factors during embryonic development has been selected as the main feature to define the specific subclasses of spinal interneurons. However, recent studies based on single-cell RNA sequencing and transcriptomic experiments suggest that this approach might not be appropriate in the adult spinal cord, where interneurons show overlapping expression profiles, especially in the ventral region. This constitutes a major challenge for the identification and direct targeting of specific populations that could be involved in locomotor recovery after a traumatic spinal cord injury in adults. Current experimental therapies, including electrical stimulation, training, pharmacological treatments, or cell implantation, that have resulted in improvements in locomotor behavior rely on the modulation of the activity and connectivity of interneurons located in the surroundings of the lesion core for the formation of detour circuits. However, very few publications clarify the specific identity of these cells. In this work, we review the studies where premotor interneurons were able to create new intraspinal circuits after different kinds of traumatic spinal cord injury, highlighting the difficulties encountered by researchers, to classify these populations.
Collapse
Affiliation(s)
- Ana Dominguez-Bajo
- Université catholique de Louvain, Louvain Institute of Biomolecular Science and Technology (LIBST), Animal Molecular and Cellular Biology Group (AMCB), Place Croix du Sud 4–5, 1348 Louvain la Neuve, Belgium
| | - Frédéric Clotman
- Université catholique de Louvain, Louvain Institute of Biomolecular Science and Technology (LIBST), Animal Molecular and Cellular Biology Group (AMCB), Place Croix du Sud 4–5, 1348 Louvain la Neuve, Belgium
| |
Collapse
|
39
|
Deng J, Sun C, Zheng Y, Gao J, Cui X, Wang Y, Zhang L, Tang P. In vivo imaging of the neuronal response to spinal cord injury: a narrative review. Neural Regen Res 2024; 19:811-817. [PMID: 37843216 PMCID: PMC10664102 DOI: 10.4103/1673-5374.382225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 05/15/2023] [Accepted: 07/07/2023] [Indexed: 10/17/2023] Open
Abstract
Deciphering the neuronal response to injury in the spinal cord is essential for exploring treatment strategies for spinal cord injury (SCI). However, this subject has been neglected in part because appropriate tools are lacking. Emerging in vivo imaging and labeling methods offer great potential for observing dynamic neural processes in the central nervous system in conditions of health and disease. This review first discusses in vivo imaging of the mouse spinal cord with a focus on the latest imaging techniques, and then analyzes the dynamic biological response of spinal cord sensory and motor neurons to SCI. We then summarize and compare the techniques behind these studies and clarify the advantages of in vivo imaging compared with traditional neuroscience examinations. Finally, we identify the challenges and possible solutions for spinal cord neuron imaging.
Collapse
Affiliation(s)
- Junhao Deng
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Chang Sun
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
- Department of Orthopedics, Air Force Medical Center, PLA, Beijing, China
| | - Ying Zheng
- Medical School of Chinese PLA, Beijing, China
| | - Jianpeng Gao
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Xiang Cui
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Yu Wang
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
| | - Licheng Zhang
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Peifu Tang
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| |
Collapse
|
40
|
Qualls KA, Xie W, Zhang J, Lückemeyer DD, Lackey SV, Strong JA, Zhang JM. Mineralocorticoid Receptor Antagonism Reduces Inflammatory Pain Measures in Mice Independent of the Receptors on Sensory Neurons. Neuroscience 2024; 541:64-76. [PMID: 38307407 PMCID: PMC11959365 DOI: 10.1016/j.neuroscience.2024.01.024] [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: 12/04/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 02/04/2024]
Abstract
Corticosteroids are commonly used in the treatment of inflammatory low back pain, and their nominal target is the glucocorticoid receptor (GR) to relieve inflammation. They can also have similar potency at the mineralocorticoid receptor (MR). The MR has been shown to be widespread in rodent and human dorsal root ganglia (DRG) neurons and non-neuronal cells, and when MR antagonists are administered during a variety of inflammatory pain models in rats, pain measures are reduced. In this study we selectively knockout (KO) the MR in sensory neurons to determine the role of MR in sensory neurons of the mouse DRG in pain measures as MR antagonism during the local inflammation of the DRG (LID) pain model. We found that MR antagonism using eplerenone reduced evoked mechanical hypersensitivity during LID, but MR KO in paw-innervating sensory neurons only did not. This could be a result of differences between prolonged (MR KO) versus acute (drug) MR block or an indicator that non-neuronal cells in the DRG are driving the effect of MR antagonists. MR KO unmyelinated C neurons are more excitable under normal and inflamed conditions, while MR KO does not affect excitability of myelinated A cells. MR KO in sensory neurons causes a reduction in overall GR mRNA but is protective against reduction of the anti-inflammatory GRα isoform during LID. These effects of MR KO in sensory neurons expanded our understanding of MR's functional role in different neuronal subtypes (A and C neurons), and its interactions with the GR.
Collapse
Affiliation(s)
- Katherine A Qualls
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Wenrui Xie
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jietong Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Debora Denardin Lückemeyer
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sierra V Lackey
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Judith A Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
41
|
Cao Y, Li R, Bai L. Vagal sensory pathway for the gut-brain communication. Semin Cell Dev Biol 2024; 156:228-243. [PMID: 37558522 DOI: 10.1016/j.semcdb.2023.07.009] [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: 11/21/2022] [Revised: 06/07/2023] [Accepted: 07/20/2023] [Indexed: 08/11/2023]
Abstract
The communication between the gut and brain is crucial for regulating various essential physiological functions, such as energy balance, fluid homeostasis, immune response, and emotion. The vagal sensory pathway plays an indispensable role in connecting the gut to the brain. Recently, our knowledge of the vagal gut-brain axis has significantly advanced through molecular genetic studies, revealing a diverse range of vagal sensory cell types with distinct peripheral innervations, response profiles, and physiological functions. Here, we review the current understanding of how vagal sensory neurons contribute to gut-brain communication. First, we highlight recent transcriptomic and genetic approaches that have characterized different vagal sensory cell types. Then, we focus on discussing how different subtypes encode numerous gut-derived signals and how their activities are translated into physiological and behavioral regulations. The emerging insights into the diverse cell types and functional properties of vagal sensory neurons have paved the way for exciting future directions, which may provide valuable insights into potential therapeutic targets for disorders involving gut-brain communication.
Collapse
Affiliation(s)
- Yiyun Cao
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Rui Li
- Chinese Institute for Brain Research, Beijing 102206, China; State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
| | - Ling Bai
- Chinese Institute for Brain Research, Beijing 102206, China.
| |
Collapse
|
42
|
Kim JI, Imaizumi K, Thete MV, Hudacova Z, Jurjuţ O, Amin ND, Scherrer G, Paşca SP. Human assembloid model of the ascending neural sensory pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584539. [PMID: 38559133 PMCID: PMC10979925 DOI: 10.1101/2024.03.11.584539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The ascending somatosensory pathways convey crucial information about pain, touch, itch, and body part movement from peripheral organs to the central nervous system. Despite a significant need for effective therapeutics modulating pain and other somatosensory modalities, clinical translation remains challenging, which is likely related to species-specific features and the lack of in vitro models to directly probe and manipulate this polysynaptic pathway. Here, we established human ascending somatosensory assembloids (hASA)- a four-part assembloid completely generated from human pluripotent stem cells that integrates somatosensory, spinal, diencephalic, and cortical organoids to model the human ascending spinothalamic pathway. Transcriptomic profiling confirmed the presence of key cell types in this circuit. Rabies tracing and calcium imaging showed that sensory neurons connected with dorsal spinal cord projection neurons, which ascending axons further connected to thalamic neurons. Following noxious chemical stimulation, single neuron calcium imaging of intact hASA demonstrated coordinated response, while four-part concomitant extracellular recordings and calcium imaging revealed synchronized activity across the assembloid. Loss of the sodium channel SCN9A, which causes pain insensitivity in humans, disrupted synchrony across the four-part hASA. Taken together, these experiments demonstrate the ability to functionally assemble the essential components of the human sensory pathway. These findings could both accelerate our understanding of human sensory circuits and facilitate therapeutic development.
Collapse
|
43
|
Gu X, Huang C, Wang S, Deng J, Guo S, Sulitan A, Gu W, Lu Q, Yuan S, Yin X. Transcriptomic Analysis of the Rat Dorsal Root Ganglion After Fracture. Mol Neurobiol 2024; 61:1467-1478. [PMID: 37725213 DOI: 10.1007/s12035-023-03637-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 09/04/2023] [Indexed: 09/21/2023]
Abstract
In fractures, pain signals are transmitted from the dorsal root ganglion (DRG) to the brain, and the DRG generates efferent signals to the injured bone to participate in the injury response. However, little is known about how this process occurs. We analyzed DRG transcriptome at 3, 7, 14, and 28 days after fracture. We identified the key pathways through KEGG and GO enrichment analysis. We then used IPA analysis to obtain upstream regulators and disease pathways. Finally, we compared the sequencing results with those of nerve injury to identify the unique transcriptome changes in DRG after fracture. We found that the first 14 days after fracture were the main repair response period, the 3rd day was the peak of repair activity, the 14th day was dominated by the stimulus response, and on the 28th day, the repair response had reached a plateau. ECM-receptor interaction, protein digestion and absorption, and the PI3K-Akt signaling pathway were most significantly enriched, which may be involved in repair regeneration, injury response, and pain transmission. Compared with the nerve injury model, DRG after fracture produced specific alterations related to bone repair, and the bone density function was the most widely activated bone-related function. Our results obtained some important genes and pathways in DRG after fracture, and we also summarized the main features of transcriptome function at each time point through functional annotation clustering of GO pathway, which gave us a deeper understanding of the role played by DRG in fracture.
Collapse
Affiliation(s)
- Xinyi Gu
- Department of Orthopedics and Traumatology, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Key Laboratory of Trauma and Neural Regeneration (Peking University), Beijing, 100000, China
| | - Chen Huang
- Department of Orthopedics and Traumatology, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Key Laboratory of Trauma and Neural Regeneration (Peking University), Beijing, 100000, China
| | - Shen Wang
- Department of Orthopedics and Traumatology, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Key Laboratory of Trauma and Neural Regeneration (Peking University), Beijing, 100000, China
| | - Jin Deng
- Department of Orthopedics and Traumatology, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Key Laboratory of Trauma and Neural Regeneration (Peking University), Beijing, 100000, China
| | - Shuhang Guo
- Department of Orthopedics and Traumatology, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Key Laboratory of Trauma and Neural Regeneration (Peking University), Beijing, 100000, China
| | - Aihaiti Sulitan
- School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing, 210023, China
| | - Wanjun Gu
- School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing, 210023, China
- Collaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese Medicine, Nanjing, 210023, China
| | - Qingguo Lu
- Trauma Center, Pizhou People's Hospital, Xuzhou, Jiangsu Province, 221300, China
| | - Shaoxun Yuan
- School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing, 210023, China.
| | - Xiaofeng Yin
- Department of Orthopedics and Traumatology, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration (Peking University), Beijing, 100000, China.
| |
Collapse
|
44
|
Guo Y, Xiang P, Sun X, Liu W, Zhou J, Yin B, Hou L, Qiang B, Li H, Shu P, Peng X. Docking protein 6 (DOK6) selectively docks the neurotrophic signaling transduction to restrain peripheral neuropathy. Signal Transduct Target Ther 2024; 9:32. [PMID: 38351062 PMCID: PMC10864363 DOI: 10.1038/s41392-024-01742-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 12/12/2023] [Accepted: 01/09/2024] [Indexed: 02/16/2024] Open
Abstract
The appropriate and specific response of nerve cells to various external cues is essential for the establishment and maintenance of neural circuits, and this process requires the proper recruitment of adaptor molecules to selectively activate downstream pathways. Here, we identified that DOK6, a member of the Dok (downstream of tyrosine kinases) family, is required for the maintenance of peripheral axons, and that loss of Dok6 can cause typical peripheral neuropathy symptoms in mice, manifested as impaired sensory, abnormal posture, paw deformities, blocked nerve conduction, and dysmyelination. Furthermore, Dok6 is highly expressed in peripheral neurons but not in Schwann cells, and genetic deletion of Dok6 in peripheral neurons led to typical peripheral myelin outfolding, axon destruction, and hindered retrograde axonal transport. Specifically, DOK6 acts as an adaptor protein for selectivity-mediated neurotrophic signal transduction and retrograde transport for TrkC and Ret but not for TrkA and TrkB. DOK6 interacts with certain proteins in the trafficking machinery and controls their phosphorylation, including MAP1B, Tau and Dynein for axonal transport, and specifically activates the downstream ERK1/2 kinase pathway to maintain axonal survival and homeostasis. This finding provides new clues to potential insights into the pathogenesis and treatment of hereditary peripheral neuropathies and other degenerative diseases.
Collapse
Affiliation(s)
- Yan Guo
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Pan Xiang
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Xiaojiao Sun
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Wei Liu
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Jiafeng Zhou
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Bin Yin
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, 100005, China
| | - Lin Hou
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, 100005, China
| | - Boqin Qiang
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, 100005, China
| | - Huiliang Li
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London, WC1E 6BT, UK
| | - Pengcheng Shu
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, 100005, China.
- Chinese Institute for Brain Research, Beijing, 102206, China.
| | - Xiaozhong Peng
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, 100005, China.
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
45
|
Tsimpos P, Desiderio S, Cabochette P, Poelvoorde P, Kricha S, Vanhamme L, Poulard C, Bellefroid EJ. Loss of G9a does not phenocopy the requirement for Prdm12 in the development of the nociceptive neuron lineage. Neural Dev 2024; 19:1. [PMID: 38167468 PMCID: PMC10759634 DOI: 10.1186/s13064-023-00179-7] [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: 09/25/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
Prdm12 is an epigenetic regulator expressed in developing and mature nociceptive neurons, playing a key role in their specification during neurogenesis and modulating pain sensation at adulthood. In vitro studies suggested that Prdm12 recruits the methyltransferase G9a through its zinc finger domains to regulate target gene expression, but how Prdm12 interacts with G9a and whether G9a plays a role in Prdm12's functional properties in sensory ganglia remain unknown. Here we report that Prdm12-G9a interaction is likely direct and that it involves the SET domain of G9a. We show that both proteins are largely co-expressed in dorsal root ganglia during early murine development, opening the possibility that G9a plays a role in DRG and may act as a mediator of Prdm12's function in the development of nociceptive sensory neurons. To test this hypothesis, we conditionally inactivated G9a in neural crest using a Wnt1-Cre transgenic mouse line. We found that the specific loss of G9a in the neural crest lineage does not lead to dorsal root ganglia hypoplasia due to the loss of somatic nociceptive neurons nor to the ectopic expression of the visceral determinant Phox2b as observed upon Prdm12 ablation. These findings suggest that Prdm12 function in the initiation of the nociceptive lineage does not critically involves its interaction with G9a.
Collapse
Affiliation(s)
- Panagiotis Tsimpos
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, B- 6041, Belgium
| | - Simon Desiderio
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, B- 6041, Belgium
| | - Pauline Cabochette
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, B- 6041, Belgium
| | - Philippe Poelvoorde
- Department of Molecular Biology, Institute of Biology and Molecular Medicine, IBMM, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Sadia Kricha
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, B- 6041, Belgium
| | - Luc Vanhamme
- Department of Molecular Biology, Institute of Biology and Molecular Medicine, IBMM, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Coralie Poulard
- Cancer Research Cancer of Lyon, Université de Lyon, Lyon, F-69000, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France
| | - Eric J Bellefroid
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, B- 6041, Belgium.
| |
Collapse
|
46
|
Wyart C, Carbo-Tano M. Design of mechanosensory feedback during undulatory locomotion to enhance speed and stability. Curr Opin Neurobiol 2023; 83:102777. [PMID: 37666012 DOI: 10.1016/j.conb.2023.102777] [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: 06/06/2023] [Revised: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 09/06/2023]
Abstract
Undulatory locomotion relies on the propagation of a wave of excitation in the spinal cord leading to consequential activation of segmental skeletal muscles along the body. Although this process relies on self-generated oscillations of motor circuits in the spinal cord, mechanosensory feedback is crucial to entrain the underlying oscillatory activity and thereby, to enhance movement power and speed. This effect is achieved through directional projections of mechanosensory neurons either sensing stretching or compression of the trunk along the rostrocaudal axis. Different mechanosensory feedback pathways act in concert to shorten and fasten the excitatory wave propagating along the body. While inhibitory mechanosensory cells feedback inhibition on excitatory premotor interneurons and motor neurons, excitatory mechanosensory cells feedforward excitation to premotor excitatory interneurons. Together, diverse mechanosensory cells coordinate the activity of skeletal muscles controlling the head and tail to optimize speed and stabilize balance during fast locomotion.
Collapse
Affiliation(s)
- Claire Wyart
- Sorbonne Université, INSERM U1127, UMR CNRS 7225, Institut du Cerveau (ICM), 47 bld de l'hôpital, Paris 75013, France.
| | - Martin Carbo-Tano
- Sorbonne Université, INSERM U1127, UMR CNRS 7225, Institut du Cerveau (ICM), 47 bld de l'hôpital, Paris 75013, France. https://twitter.com/martincarbotano
| |
Collapse
|
47
|
Dey S, Barkai O, Gokhman I, Suissa S, Haffner-Krausz R, Wigoda N, Feldmesser E, Ben-Dor S, Kovalenko A, Binshtok A, Yaron A. Kinesin family member 2A gates nociception. Cell Rep 2023; 42:113257. [PMID: 37851573 DOI: 10.1016/j.celrep.2023.113257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 08/23/2023] [Accepted: 09/27/2023] [Indexed: 10/20/2023] Open
Abstract
Nociceptive axons undergo remodeling as they innervate their targets during development and in response to environmental insults and pathological conditions. How is nociceptive morphogenesis regulated? Here, we show that the microtubule destabilizer kinesin family member 2A (Kif2a) is a key regulator of nociceptive terminal structures and pain sensitivity. Ablation of Kif2a in sensory neurons causes hyperinnervation and hypersensitivity to noxious stimuli in young adult mice, whereas touch sensitivity and proprioception remain unaffected. Computational modeling predicts that structural remodeling is sufficient to explain the phenotypes. Furthermore, Kif2a deficiency triggers a transcriptional response comprising sustained upregulation of injury-related genes and homeostatic downregulation of highly specific channels and receptors at the late stage. The latter effect can be predicted to relieve the hyperexcitability of nociceptive neurons, despite persisting morphological aberrations, and indeed correlates with the resolution of pain hypersensitivity. Overall, we reveal a critical control node defining nociceptive terminal structure, which is regulating nociception.
Collapse
Affiliation(s)
- Swagata Dey
- Department of Biomolecular Sciences and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Omer Barkai
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah School of Medicine, Jerusalem 91120, Israel; The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel; F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Irena Gokhman
- Department of Biomolecular Sciences and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Sapir Suissa
- Department of Biomolecular Sciences and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Rebecca Haffner-Krausz
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Noa Wigoda
- Bioinformatics Unit, Life Science Core Facilities, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ester Feldmesser
- Bioinformatics Unit, Life Science Core Facilities, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Shifra Ben-Dor
- Bioinformatics Unit, Life Science Core Facilities, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Andrew Kovalenko
- Department of Biomolecular Sciences and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alexander Binshtok
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah School of Medicine, Jerusalem 91120, Israel; The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Avraham Yaron
- Department of Biomolecular Sciences and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
48
|
Koutsioumpa C, Santiago C, Jacobs K, Lehnert BP, Barrera V, Hutchinson JN, Schmelyun D, Lehoczky JA, Paul DL, Ginty DD. Skin-type-dependent development of murine mechanosensory neurons. Dev Cell 2023; 58:2032-2047.e6. [PMID: 37607547 PMCID: PMC10615785 DOI: 10.1016/j.devcel.2023.07.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/26/2023] [Accepted: 07/27/2023] [Indexed: 08/24/2023]
Abstract
Mechanosensory neurons innervating the skin underlie our sense of touch. Fast-conducting, rapidly adapting mechanoreceptors innervating glabrous (non-hairy) skin form Meissner corpuscles, while in hairy skin, they associate with hair follicles, forming longitudinal lanceolate endings. How mechanoreceptors develop axonal endings appropriate for their skin targets is unknown. We report that mechanoreceptor morphologies across different skin regions are indistinguishable during early development but diverge post-natally, in parallel with skin maturation. Neurons terminating along the glabrous and hairy skin border exhibit hybrid morphologies, forming both Meissner corpuscles and lanceolate endings. Additionally, molecular profiles of neonatal glabrous and hairy skin-innervating neurons largely overlap. In mouse mutants with ectopic glabrous skin, mechanosensory neurons form end-organs appropriate for the altered skin type. Finally, BMP5 and BMP7 are enriched in glabrous skin, and signaling through type I bone morphogenetic protein (BMP) receptors in neurons is critical for Meissner corpuscle morphology. Thus, mechanoreceptor morphogenesis is flexibly instructed by target tissues.
Collapse
Affiliation(s)
- Charalampia Koutsioumpa
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Celine Santiago
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Kiani Jacobs
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Brendan P Lehnert
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Victor Barrera
- Bioinformatics Core, Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - John N Hutchinson
- Bioinformatics Core, Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Dhane Schmelyun
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Jessica A Lehoczky
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - David L Paul
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
49
|
Santiago C, Sharma N, Africawala N, Siegrist J, Handler A, Tasnim A, Anjum R, Turecek J, Lehnert BP, Renauld S, Nolan-Tamariz M, Iskols M, Magee AR, Paradis S, Ginty DD. Activity-dependent development of the body's touch receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.23.559109. [PMID: 37790437 PMCID: PMC10542488 DOI: 10.1101/2023.09.23.559109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
We report a role for activity in the development of the primary sensory neurons that detect touch. Genetic deletion of Piezo2, the principal mechanosensitive ion channel in somatosensory neurons, caused profound changes in the formation of mechanosensory end organ structures and altered somatosensory neuron central targeting. Single cell RNA sequencing of Piezo2 conditional mutants revealed changes in gene expression in the sensory neurons activated by light mechanical forces, whereas other neuronal classes were less affected. To further test the role of activity in mechanosensory end organ development, we genetically deleted the voltage-gated sodium channel Nav1.6 (Scn8a) in somatosensory neurons throughout development and found that Scn8a mutants also have disrupted somatosensory neuron morphologies and altered electrophysiological responses to mechanical stimuli. Together, these findings indicate that mechanically evoked neuronal activity acts early in life to shape the maturation of the mechanosensory end organs that underlie our sense of gentle touch.
Collapse
Affiliation(s)
- Celine Santiago
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Nikhil Sharma
- Department of Molecular Pharmacology and Therapeutics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Nusrat Africawala
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Julianna Siegrist
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Annie Handler
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Aniqa Tasnim
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Rabia Anjum
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, MA, 02453, USA
| | - Josef Turecek
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Brendan P. Lehnert
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Sophia Renauld
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Michael Nolan-Tamariz
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Michael Iskols
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Alexandra R. Magee
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Suzanne Paradis
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, MA, 02453, USA
| | - David D. Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
- Lead Contact
| |
Collapse
|
50
|
Hong JY, Kim H, Yeo C, Lee J, Jeon WJ, Lee YJ, Ha IH. Epidural Injection of Harpagoside for the Recovery of Rats with Lumbar Spinal Stenosis. Cells 2023; 12:2281. [PMID: 37759506 PMCID: PMC10526993 DOI: 10.3390/cells12182281] [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: 08/08/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Epidural administration is the leading therapeutic option for the management of pain associated with lumbar spinal stenosis (LSS), which is characterized by compression of the nerve root due to narrowing of the spinal canal. Corticosteroids are effective in alleviating LSS-related pain but can lead to complications with long-term use. Recent studies have focused on identifying promising medications administered epidurally to affected spinal regions. In this study, we aimed to investigate the effectiveness of harpagoside (HAS) as an epidural medication in rats with LSS. HAS at various concentrations was effective for neuroprotection against ferrous sulfate damage and consequent promotion of axonal outgrowth in primary spinal cord neurons. When two concentrations of HAS (100 and 200 μg/kg) were administered to the rat LSS model via the epidural space once a day for 4 weeks, the inflammatory responses around the silicone block used for LSS were substantially reduced. Consistently, pain-related factors were significantly suppressed by the epidural administration of HAS. The motor functions of rats with LSS significantly improved. These findings suggest that targeted delivery of HAS directly to the affected area via epidural injection holds promise as a potential treatment option for the recovery of patients with LSS.
Collapse
Affiliation(s)
| | | | | | | | | | | | - In-Hyuk Ha
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul 135-896, Republic of Korea; (J.Y.H.); (H.K.); (C.Y.); (J.L.); (W.-J.J.); (Y.J.L.)
| |
Collapse
|