1
|
Chen JC, Kaczmarczyk L, de-Faria FM, Szczot M, Jackson WS, Larsson M. Genetic targeting of myelinated primary afferent neurons using a new Nefh CreERT2 knock-in mouse. Sci Rep 2025; 15:10890. [PMID: 40158013 PMCID: PMC11954935 DOI: 10.1038/s41598-025-95874-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Primary afferent neurons that convey somatosensory modalities comprise two large, heterogeneous populations: small-diameter neurons that give rise to slowly conducting unmyelinated axonal C fibers and medium-to-large diameter neurons with fast myelinated A fibers. Despite these two major groupings, tools to differentiate between unmyelinated and myelinated primary afferent fibers by genetic targeting have not been available; in particular, whereas numerous mouse driver lines exist to target different C fiber populations, genetic tools that target myelinated primary afferent populations are scarce. Here we describe a knock-in mouse line expressing tamoxifen-dependent CreERT2 under control of the Nefh gene, which encodes neurofilament heavy chain (NFH or NF200), a protein that is highly enriched in myelinated fibers. This mouse enables highly selective and efficient recombination of Cre-dependent reporters for functional and anatomical interrogation of myelinated fibers while excluding unmyelinated C fibers. In combination with other recombinase-expressing mouse lines, this genetic tool will be valuable for intersectional targeting of subpopulations of myelinated primary afferent fibers.
Collapse
Affiliation(s)
- John Cy Chen
- Division of Cell- and Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, 581 85, Linköping, Sweden
| | - Lech Kaczmarczyk
- Division of Cell- and Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, 581 85, Linköping, Sweden
| | - Felipe Meira de-Faria
- Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience, Linköping University, 581 85, Linköping, Sweden
| | - Marcin Szczot
- Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience, Linköping University, 581 85, Linköping, Sweden
| | - Walker S Jackson
- Division of Cell- and Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, 581 85, Linköping, Sweden
| | - Max Larsson
- Division of Cell- and Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, 581 85, Linköping, Sweden.
| |
Collapse
|
2
|
Shukla S, Comerci CJ, Süel GM, Jahed Z. Bioelectronic tools for understanding the universal language of electrical signaling across species and kingdoms. Biosens Bioelectron 2025; 267:116843. [PMID: 39426280 DOI: 10.1016/j.bios.2024.116843] [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: 07/24/2023] [Revised: 09/10/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
Modern bioelectronic tools are rapidly advancing to detect electric potentials within networks of electrogenic cells, such as cardiomyocytes, neurons, and pancreatic beta cells. However, it is becoming evident that electrical signaling is not limited to the animal kingdom but may be a universal form of cell-cell communication. In this review, we discuss the existing evidence of, and tools used to collect, subcellular, single-cell and network-level electrical signals across kingdoms, including bacteria, plants, fungi, and even viruses. We discuss how cellular networks employ altered electrical "circuitry" and intercellular mechanisms across kingdoms, and we assess the functionality and scalability of cutting-edge nanobioelectronics to collect electrical signatures regardless of cell size, shape, or function. Researchers today aim to design micro- and nano-topographic structures which harness mechanosensitive membrane and cytoskeletal pathways that enable tight electrical coupling to subcellular compartments within high-throughput recording systems. Finally, we identify gaps in current knowledge of inter-species and inter-kingdom electrical signaling and propose critical milestones needed to create a central theory of electrical signaling across kingdoms. Our discussion demonstrates the need for high resolution, high throughput tools which can probe multiple, diverse cell types at once in their native or experimentally-modeled environments. These advancements will not only reveal the underlying biophysical laws governing the universal language of electrical communication, but can enable bidirectional electrical communication and manipulation of biological systems.
Collapse
Affiliation(s)
- Shivani Shukla
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, United States; Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, United States
| | - Colin J Comerci
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, United States
| | - Gürol M Süel
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, United States
| | - Zeinab Jahed
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, United States; Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, United States.
| |
Collapse
|
3
|
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
|
4
|
Kim J, Park J, Choe G, Jeong SI, Kim HS, Lee JY. A Gelatin/Alginate Double Network Hydrogel Nerve Guidance Conduit Fabricated by a Chemical-Free Gamma Radiation for Peripheral Nerve Regeneration. Adv Healthc Mater 2024; 13:e2400142. [PMID: 38566357 DOI: 10.1002/adhm.202400142] [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: 03/21/2024] [Indexed: 04/04/2024]
Abstract
Nerve guidance conduits (NGCs) are widely developed using various materials for the functional repair of injured or diseased peripheral nerves. Especially, hydrogels are considered highly suitable for the fabrication of NGCs due to their beneficial tissue-mimicking characteristics (e.g., high water content, softness, and porosity). However, the practical applications of hydrogel-based NGCs are hindered due to their poor mechanical properties and complicated fabrication processes. To bridge this gap, a novel double-network (DN) hydrogel using alginate and gelatin by a two-step crosslinking process involving chemical-free gamma irradiation and ionic crosslinking, is developed. DN hydrogels (1% alginate and 15% gelatin), crosslinked with 30 kGy gamma irradiation and barium ions, exhibit substantially improved mechanical properties, including tensile strength, elastic modulus, and fracture stain, compared to single network (SN) gelatin hydrogels. Additionally, the DN hydrogel NGC exhibits excellent kink resistance, mechanical stability to successive compression, suture retention, and enzymatic degradability. In vivo studies with a sciatic defect rat model indicate substantially improved nerve function recovery with the DN hydrogel NGC compared to SN gelatin and commercial silicone NGCs, as confirm footprint analysis, electromyography, and muscle weight measurement. Histological examination reveals that, in the DN NGC group, the expression of Schwann cell and neuronal markers, myelin sheath, and exon diameter are superior to the other controls. Furthermore, the DN NGC group demonstrates increased muscle fiber formation and reduced fibrotic scarring. These findings suggest that the mechanically robust, degradable, and biocompatible DN hydrogel NGC can serve as a novel platform for peripheral nerve regeneration and other biomedical applications, such as implantable tissue constructs.
Collapse
Affiliation(s)
- Junghyun Kim
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Junggeon Park
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Goeun Choe
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Sung-In Jeong
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, 56212, Republic of Korea
| | - Hyung-Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Jae Young Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| |
Collapse
|
5
|
Villalón Landeros E, Kho SC, Church TR, Brennan A, Türker F, Delannoy M, Caterina MJ, Margolis SS. The nociceptive activity of peripheral sensory neurons is modulated by the neuronal membrane proteasome. Cell Rep 2024; 43:114058. [PMID: 38614084 PMCID: PMC11157458 DOI: 10.1016/j.celrep.2024.114058] [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: 01/25/2023] [Revised: 03/05/2024] [Accepted: 03/20/2024] [Indexed: 04/15/2024] Open
Abstract
Proteasomes are critical for peripheral nervous system (PNS) function. Here, we investigate mammalian PNS proteasomes and reveal the presence of the neuronal membrane proteasome (NMP). We show that specific inhibition of the NMP on distal nerve fibers innervating the mouse hind paw leads to reduction in mechanical and pain sensitivity. Through investigating PNS NMPs, we demonstrate their presence on the somata and proximal and distal axons of a subset of dorsal root ganglion (DRG) neurons. Single-cell RNA sequencing experiments reveal that the NMP-expressing DRGs are primarily MrgprA3+ and Cysltr2+. NMP inhibition in DRG cultures leads to cell-autonomous and non-cell-autonomous changes in Ca2+ signaling induced by KCl depolarization, αβ-meATP, or the pruritogen histamine. Taken together, these data support a model whereby NMPs are expressed on a subset of somatosensory DRGs to modulate signaling between neurons of distinct sensory modalities and indicate the NMP as a potential target for controlling pain.
Collapse
Affiliation(s)
- Eric Villalón Landeros
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Samuel C Kho
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Taylor R Church
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Anna Brennan
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Fulya Türker
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael Delannoy
- Microscopy Facility, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael J Caterina
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurosurgery and Neurosurgery Pain Research Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Seth S Margolis
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
6
|
Merkulyeva N, Mikhalkin A. Transient expression of heavy-chain neurofilaments in the perigeniculate nucleus of cats. Brain Struct Funct 2024; 229:489-495. [PMID: 38265459 DOI: 10.1007/s00429-023-02752-6] [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: 09/17/2023] [Accepted: 12/12/2023] [Indexed: 01/25/2024]
Abstract
The perigeniculate nucleus (PGN) is a visual part of the thalamic reticular nucleus modulating the information transfer between the lateral geniculate nucleus and the visual cortex. This study focused on the postnatal development of the PGN in cats, using the SMI-32 antibody, which recognizes non-phosphorylated heavy-chain neurofilaments responsible for neuronal structural maturation and is also used as a marker for motion processing, or Y, stream. We questioned whether transient neuronal populations exist in the PGN and can they possibly be related to the Y processing stream. We uncovered a transient, robust SMI-32 staining in the PGN of kittens aged 0-34 days with the significant decline in the cellular density of labeled cells in older animals. According to the double-labeling, in all examined age groups, perigeniculate SMI-32-immunopositive cells are part of the main parvalbumin-positive population. The maximal cellular density of the double-stained cells appeared in animals aged 10-28 days. We also revealed that the most significant growth of perigeniculate cells's soma occurred at three postnatal weeks. The possible link of our data to the development of the Y visual processing stream and to the heterogeneity of the perigeniculate neuronal population is also discussed.
Collapse
Affiliation(s)
- Natalia Merkulyeva
- Pavlov Institute of Physiology RAS, Makarov Nab., 6, Saint-Petersburg, Russia, 199034.
| | - Aleksandr Mikhalkin
- Pavlov Institute of Physiology RAS, Makarov Nab., 6, Saint-Petersburg, Russia, 199034
| |
Collapse
|
7
|
Hovhannisyan AH, Lindquist KA, Belugin S, Mecklenburg J, Ibrahim T, Tram M, Corey TM, Salmon AB, Perez D, Ruparel S, Akopian AN. Sensory innervation of masseter, temporal and lateral pterygoid muscles in common marmosets. Sci Rep 2023; 13:23062. [PMID: 38155190 PMCID: PMC10754842 DOI: 10.1038/s41598-023-49882-9] [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: 04/26/2023] [Accepted: 12/13/2023] [Indexed: 12/30/2023] Open
Abstract
Myogenous temporomandibular disorders is associated with an increased responsiveness of nerves innervating the masseter (MM), temporal (TM), and lateral pterygoid muscles (LPM). This study aimed to examine sensory nerve types innervating MM, TM and LPM of adult non-human primate-common marmosets. Sensory nerves were localized in specific regions of these muscles. Pgp9.5, marker for all nerves, and NFH, a marker for A-fibers, showed that masticatory muscles were primarily innervated with A-fibers. The proportion of C- to A-fibers was highest in LPM, and lowest in MM. All C-fibers (pgp9.5+/NFH-) observed in masticatory muscles were peptidergic (CGRP+) and lacked mrgprD and CHRNA3, a silent nociceptive marker. TrpV1 was register in 17% of LPM nerves. All fibers in masticatory muscles were labeled with GFAP+, a myelin sheath marker. There were substantially more peptidergic A-fibers (CGRP+/NFH+) in TM and LPM compared to MM. MM, TM and LPM NFH+ fibers contained different percentages of trkC+ and parvalbumin+, but not trkB+ fibers. Tyrosine hydroxylase antibodies, which did not label TG, highlighted sympathetic fibers around blood vessels of the masticatory muscles. Overall, masticatory muscle types of marmosets have similarities and differences in innervation patterns.
Collapse
Affiliation(s)
- Anahit H Hovhannisyan
- Departments of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Karen A Lindquist
- Integrated Biomedical Sciences (IBMS) Program, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Sergei Belugin
- Departments of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Jennifer Mecklenburg
- Departments of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Tarek Ibrahim
- Departments of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Meilinn Tram
- Departments of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
- Integrated Biomedical Sciences (IBMS) Program, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Tatiana M Corey
- Departments of Laboratory Animal Resources, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Adam B Salmon
- Departments of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Sam and Ann Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, Geriatric Research Education and Clinical Center San Antonio, San Antonio, TX, 78229, USA
| | - Daniel Perez
- Oral and Maxillofacial Surgery, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Shivani Ruparel
- Departments of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
- Integrated Biomedical Sciences (IBMS) Program, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Departments of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Armen N Akopian
- Departments of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
- Integrated Biomedical Sciences (IBMS) Program, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
- Departments of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
8
|
Alex Thomas M, Cui X, Artinian LR, Cao Q, Jing J, Silva FC, Wang S, Zigman JM, Sun Y, Shi H, Xue B. Crosstalk between Gut Sensory Ghrelin Signaling and Adipose Tissue Sympathetic Outflow Regulates Metabolic Homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.25.568689. [PMID: 38076894 PMCID: PMC10705268 DOI: 10.1101/2023.11.25.568689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
The stomach-derived orexigenic hormone ghrelin is a key regulator of energy homeostasis and metabolism in humans. The ghrelin receptor, growth hormone secretagogue receptor 1a (GHSR), is widely expressed in the brain and gastrointestinal vagal sensory neurons, and neuronal GHSR knockout results in a profoundly beneficial metabolic profile and protects against diet-induced obesity (DIO) and insulin resistance. Here we show that in addition to the well characterized vagal GHSR, GHSR is robustly expressed in gastrointestinal sensory neurons emanating from spinal dorsal root ganglia. Remarkably, sensory neuron GHSR deletion attenuates DIO through increased energy expenditure and sympathetic outflow to adipose tissue independent of food intake. In addition, neuronal viral tract tracing reveals prominent crosstalk between gut non-vagal sensory afferents and adipose sympathetic outflow. Hence, these findings demonstrate a novel gut sensory ghrelin signaling pathway critical for maintaining energy homeostasis.
Collapse
Affiliation(s)
- M. Alex Thomas
- Department of Biology, Georgia State University, Atlanta, GA
| | - Xin Cui
- Department of Biology, Georgia State University, Atlanta, GA
| | | | - Qiang Cao
- Department of Biology, Georgia State University, Atlanta, GA
| | - Jia Jing
- Department of Biology, Georgia State University, Atlanta, GA
| | - Felipe C. Silva
- Department of Biology, Georgia State University, Atlanta, GA
| | - Shirong Wang
- Department of Biology, Georgia State University, Atlanta, GA
| | - Jeffrey M. Zigman
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Yuxiang Sun
- Department of Nutrition, Texas A & M University, College Station, TX
| | - Hang Shi
- Department of Biology, Georgia State University, Atlanta, GA
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, GA
| |
Collapse
|
9
|
Hovhannisyan AH, Lindquist K, Belugin S, Mecklenburg J, Ibrahim T, Tram M, Corey T, Salmon A, Ruparel S, Ruparel S, Akopian A. Sensory innervation of masseter, temporal and lateral pterygoid muscles in common marmosets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.10.528062. [PMID: 36798270 PMCID: PMC9934658 DOI: 10.1101/2023.02.10.528062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Myogenous temporomandibular disorders (TMDM) is associated with an increased responsiveness of nerves innervating the masseter (MM), temporal (TM), medial pterygoid (MPM) and lateral pterygoid muscles (LPM). This study aimed to examine sensory nerve types innervating MM, TM and LPM of adult non-human primate - common marmosets. Sensory nerves are localized in specific regions of these muscles. Pgp9.5, marker for all nerves, and NFH, a marker for A-fibers, showed that masticatory muscles were predominantly innervated with A-fibers. The proportion of C- to A-fibers was highest in LPM, and minimal (6-8%) in MM. All C-fibers (pgp9.5+/NFH-) observed in masticatory muscles were peptidergic (CGRP+) and lacked mrgprD, trpV1 and CHRNA3, a silent nociceptive marker. All fibers in masticatory muscles were labeled with GFAP+, a myelin sheath marker. There were substantially more peptidergic A-fibers (CGRP+/NFH+) in TM and LPM compared to MM. Almost all A-fibers in MM expressed trkC, with some of them having trkB and parvalbumin. In contrast, a lesser number of TM and LPM nerves expressed trkC, and lacked trkB. Tyrosine hydroxylase antibodies, which did not label TG, highlighted sympathetic fibers around blood vessels of the masticatory muscles. Overall, masticatory muscle types of marmosets have distinct and different innervation patterns.
Collapse
|
10
|
Fukazawa A, Hori A, Hotta N, Katanosaka K, Estrada JA, Ishizawa R, Kim HK, Iwamoto GA, Smith SA, Vongpatanasin W, Mizuno M. Antagonism of TRPV4 channels partially reduces mechanotransduction in rat skeletal muscle afferents. J Physiol 2023; 601:1407-1424. [PMID: 36869605 PMCID: PMC10106437 DOI: 10.1113/jp284026] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/02/2023] [Indexed: 03/05/2023] Open
Abstract
Mechanical distortion of working skeletal muscle induces sympathoexcitation via thin fibre afferents, a reflex response known as the skeletal muscle mechanoreflex. However, to date, the receptor ion channels responsible for mechanotransduction in skeletal muscle remain largely undetermined. Transient receptor potential vanilloid 4 (TRPV4) is known to sense mechanical stimuli such as shear stress or osmotic pressure in various organs. It is hypothesized that TRPV4 in thin-fibre primary afferents innervating skeletal muscle is involved in mechanotransduction. Fluorescence immunostaining revealed that 20.1 ± 10.1% of TRPV4 positive neurons were small dorsal root ganglion (DRG) neurons that were DiI-labelled, and among them 9.5 ± 6.1% of TRPV4 co-localized with the C-fibre marker peripherin. In vitro whole-cell patch clamp recordings from cultured rat DRG neurons demonstrated that mechanically activated current amplitude was significantly attenuated after the application of the TRPV4 antagonist HC067047 compared to control (P = 0.004). Such reductions were also observed in single-fibre recordings from a muscle-nerve ex vivo preparation where HC067047 significantly decreased afferent discharge to mechanical stimulation (P = 0.007). Likewise, in an in vivo decerebrate rat preparation, the renal sympathetic nerve activity (RSNA) and mean arterial pressure (MAP) responses to passive stretch of hindlimb muscle were significantly reduced by intra-arterial injection of HC067047 (ΔRSNA: P = 0.019, ΔMAP: P = 0.002). The findings suggest that TRPV4 plays an important role in mechanotransduction contributing to the cardiovascular responses evoked by the skeletal muscle mechanoreflex during exercise. KEY POINTS: Although a mechanical stimulus to skeletal muscle reflexively activates the sympathetic nervous system, the receptors responsible for mechanotransduction in skeletal muscle thin fibre afferents have not been fully identified. Evidence suggests that TRPV4 is a mechanosensitive channel that plays an important role in mechanotransduction within various organs. Immunocytochemical staining demonstrates that TRPV4 is expressed in group IV skeletal muscle afferents. In addition, we show that the TRPV4 antagonist HC067047 decreases the responsiveness of thin fibre afferents to mechanical stimulation at the muscle tissue level as well as at the level of dorsal root ganglion neurons. Moreover, we demonstrate that intra-arterial HC067047 injection attenuates the sympathetic and pressor responses to passive muscle stretch in decerebrate rats. These data suggest that antagonism of TRPV4 attenuates mechanotransduction in skeletal muscle afferents. The present study demonstrates a probable physiological role for TRPV4 in the regulation of mechanical sensation in somatosensory thin fibre muscle afferents.
Collapse
Affiliation(s)
- Ayumi Fukazawa
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Amane Hori
- Graduate School of Life and Health Sciences, Chubu University, Kasugai 487-850, Japan
- Japan Society for the Promotion of Science, Tokyo 102-8472, Japan
| | - Norio Hotta
- Graduate School of Life and Health Sciences, Chubu University, Kasugai 487-850, Japan
- College of Life and Health Sciences, Chubu University, Kasugai 487-850, Japan
| | - Kimiaki Katanosaka
- Graduate School of Life and Health Sciences, Chubu University, Kasugai 487-850, Japan
- College of Life and Health Sciences, Chubu University, Kasugai 487-850, Japan
| | - Juan A. Estrada
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rie Ishizawa
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Han-Kyul Kim
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Gary A. Iwamoto
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Scott A. Smith
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wanpen Vongpatanasin
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Masaki Mizuno
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
11
|
Lopez JA, Yamamoto A, Vecchi JT, Hagen J, Lee K, Sonka M, Hansen MR, Lee A. Caldendrin represses neurite regeneration and growth in dorsal root ganglion neurons. Sci Rep 2023; 13:2608. [PMID: 36788334 PMCID: PMC9929226 DOI: 10.1038/s41598-023-29622-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Caldendrin is a Ca2+ binding protein that interacts with multiple effectors, such as the Cav1 L-type Ca2+ channel, which play a prominent role in regulating the outgrowth of dendrites and axons (i.e., neurites) during development and in response to injury. Here, we investigated the role of caldendrin in Cav1-dependent pathways that impinge upon neurite growth in dorsal root ganglion neurons (DRGNs). By immunofluorescence, caldendrin was localized in medium- and large- diameter DRGNs. Compared to DRGNs cultured from WT mice, DRGNs of caldendrin knockout (KO) mice exhibited enhanced neurite regeneration and outgrowth. Strong depolarization, which normally represses neurite growth through activation of Cav1 channels, had no effect on neurite growth in DRGN cultures from female caldendrin KO mice. Remarkably, DRGNs from caldendrin KO males were no different from those of WT males in terms of depolarization-dependent neurite growth repression. We conclude that caldendrin opposes neurite regeneration and growth, and this involves coupling of Cav1 channels to growth-inhibitory pathways in DRGNs of females but not males.
Collapse
Affiliation(s)
- Josue A Lopez
- Department of Neuroscience, University of Texas-Austin, 100 E. 24th St., Austin, TX, 78712, USA
| | - Annamarie Yamamoto
- Department of Neuroscience, University of Texas-Austin, 100 E. 24th St., Austin, TX, 78712, USA
| | - Joseph T Vecchi
- Department of Molecular Physiology and Biophysics and Otolaryngology Head-Neck Surgery, Iowa Neuroscience Institute, Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Jussara Hagen
- Department of Molecular Physiology and Biophysics and Otolaryngology Head-Neck Surgery, Iowa Neuroscience Institute, Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Kyungmoo Lee
- Electrical and Computer Engineering, Iowa Institute for Biomedical Imaging, University of Iowa, 51 Newton Rd. Iowa City, Iowa, 52242, USA
| | - Milan Sonka
- Electrical and Computer Engineering, Iowa Institute for Biomedical Imaging, University of Iowa, 51 Newton Rd. Iowa City, Iowa, 52242, USA
| | - Marlan R Hansen
- Department of Molecular Physiology and Biophysics and Otolaryngology Head-Neck Surgery, Iowa Neuroscience Institute, Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Amy Lee
- Department of Neuroscience, University of Texas-Austin, 100 E. 24th St., Austin, TX, 78712, USA.
| |
Collapse
|
12
|
Xi H, Wang C, Li Q, Ye Q, Zhu Y, Mao Y. S-Propargyl-Cysteine Ameliorates Peripheral Nerve Injury through Microvascular Reconstruction. Antioxidants (Basel) 2023; 12:antiox12020294. [PMID: 36829853 PMCID: PMC9952745 DOI: 10.3390/antiox12020294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Microvascular reconstruction is essential for peripheral nerve repair. S-Propargyl-cysteine (SPRC), the endogenous hydrogen sulfide (H2S) donor, has been reported to promote angiogenesis. The aim of this study is to utilize the pro-angiogenic ability of SPRC to support peripheral nerve repair and to explore the potential mechanisms. The effects and mechanisms of SPRC on angiogenesis and peripheral nerve repair were examined under hypoxic condition by establishing a sciatic nerve crushed injury model in mice and rats, and a hypoxia model in human umbilical vascular endothelial cells (HUVECs) in vitro. We found that SPRC accelerated the function recovery of the injured sciatic nerve and alleviated atrophy of the gastrocnemius muscle in mice. It facilitated the viability of Schwann cells (SCs), the outgrowth and myelination of regenerated axons, and angiogenesis in rats. It enhanced the viability, proliferation, adhesion, migration, and tube formation of HUVECs under hypoxic condition. SPRC activated sirtuin1 (SIRT1) expression by promoting the production of endogenous H2S, and SIRT1 negatively regulated Notch signaling in endothelial cells (ECs), thereby promoting angiogenesis. Collectively, our study has provided important evidence that SPRC has an effective role in peripheral nerve repair through microvascular reconstruction, which could be a potentially effective medical therapy for peripheral nerve injury.
Collapse
Affiliation(s)
- Haiyan Xi
- Shanghai Key Laboratory of Bioactive Small Molecules, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Chenye Wang
- Shanghai Key Laboratory of Bioactive Small Molecules, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Qixiu Li
- Shanghai Key Laboratory of Bioactive Small Molecules, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Qing Ye
- Shanghai Key Laboratory of Bioactive Small Molecules, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yizhun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules, School of Pharmacy, Fudan University, Shanghai 201203, China
- School of Pharmacy, Macau University of Science and Technology, Macau, China
- Correspondence: (Y.Z.); (Y.M.)
| | - Yicheng Mao
- Shanghai Key Laboratory of Bioactive Small Molecules, School of Pharmacy, Fudan University, Shanghai 201203, China
- Correspondence: (Y.Z.); (Y.M.)
| |
Collapse
|
13
|
Uhelski ML, Johns ME, Horrmann A, Mohamed S, Sohail A, Khasabova IA, Simone DA, Banik RK. Adverse effects of methylene blue in peripheral neurons: An in vitro electrophysiology and cell culture study. Mol Pain 2022; 18:17448069221142523. [PMID: 36408567 PMCID: PMC9730009 DOI: 10.1177/17448069221142523] [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] [Indexed: 11/22/2022] Open
Abstract
Methylene blue (MB) is an effective treatment for methemoglobinemia, ifosfamide-induced encephalopathy, cyanide poisoning, and refractory vasoplegia. However, clinical case reports and preclinical studies indicate potentially neurotoxic activity of MB at certain concentrations. The exact mechanisms of MB neurotoxicity are not known, and while the effects of MB on neuronal tissue from different brain regions and myenteric ganglia have been examined, its effects on primary afferent neurons from dorsal root ganglia (DRG) have not been studied. Mouse DRG were exposed to MB (0.3-10 μM) in vitro to assess neurite outgrowth. Increasing concentrations of MB (0.3-10 μM) were associated with neurotoxicity as shown by a substantial loss of cells with neurite formation, particularly at 10 μM. In parallel experiments, cultured rat DRG neurons were treated with MB (100 μM) to examine how MB affects electrical membrane properties of small-diameter sensory neurons. MB decreased peak inward and outward current densities, decreased action potential amplitude, overshoot, afterhyperpolarization, increased action potential rise time, and decreased action potential firing in response to current stimulation. MB induced dose-dependent toxicity in peripheral neurons, in vitro. These findings are consistent with studies in brain and myenteric ganglion neurons showing increased neuronal loss and altered membrane electrical properties after MB application. Further research is needed to parse out the toxicity profile for MB to minimize damage to neuronal structures and reduce side effects in clinical settings.
Collapse
Affiliation(s)
- Megan L Uhelski
- Department of Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Malcolm E Johns
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Alec Horrmann
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Sadiq Mohamed
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Ayesha Sohail
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Iryna A Khasabova
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Donald A Simone
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Ratan K Banik
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA,Ratan K Banik, Department of Anesthesiology, University of Minnesota, B515 Mayo Memorial Building, 420 Delaware Street S.E., MMC 294, Minneapolis, MN 55455, USA.
| |
Collapse
|
14
|
Sex Differences in Neuropathy: The Paradigmatic Case of MetFormin. Int J Mol Sci 2022; 23:ijms232314503. [PMID: 36498830 PMCID: PMC9738696 DOI: 10.3390/ijms232314503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
As a widely prescribed anti-diabetic drug, metformin has been receiving novel attention for its analgesic potential. In the study of the complex etiology of neuropathic pain (NeP), male and female individuals exhibit quite different responses characterized by higher pain sensitivity and greater NeP incidence in women. This "gender gap" in our knowledge of sex differences in pain processing strongly limits the sex-oriented treatment of patients suffering from NeP. Besides, the current investigation of the analgesic potential of metformin has not addressed the "gender gap" problem. Hence, this study focuses on metformin and sex-dependent analgesia in a murine model of NeP induced by chronic constriction injury of the sciatic nerve. We investigated sexual dimorphism in signaling pathways involved by 7 days of metformin administration, such as changes in AMP-activated protein kinase and the positive regulation of autophagy machinery, discovering that metformin affected in a sexually dimorphic manner the immunological and inflammatory response to nerve lesion. These effects were complemented by morphological and adaptive changes occurring after peripheral nerve injury. Altogether these data can contribute to explaining a number of potential mechanisms responsible for the complete recovery from NeP found in male mice, as opposed to the failure of long-lasting recovery in female animals.
Collapse
|
15
|
Eller OC, Stair RN, Neal C, Rowe PS, Nelson-Brantley J, Young EE, Baumbauer KM. Comprehensive phenotyping of cutaneous afferents reveals early-onset alterations in nociceptor response properties, release of CGRP, and hindpaw edema following spinal cord injury. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2022; 12:100097. [PMID: 35756343 PMCID: PMC9218836 DOI: 10.1016/j.ynpai.2022.100097] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 11/30/2022]
Abstract
Spinal cord injury (SCI) is a complex syndrome that has profound effects on patient well-being, including the development of medically-resistant chronic pain. The mechanisms underlying SCI pain have been the subject of thorough investigation but remain poorly understood. While the majority of the research has focused on changes occurring within and surrounding the site of injury in the spinal cord, there is now a consensus that alterations within the peripheral nervous system, namely sensitization of nociceptors, contribute to the development and maintenance of chronic SCI pain. Using an ex vivo skin/nerve/DRG/spinal cord preparation to characterize afferent response properties following SCI, we found that SCI increased mechanical and thermal responding, as well as the incidence of spontaneous activity (SA) and afterdischarge (AD), in below-level C-fiber nociceptors 24 hr following injury relative to naïve controls. Interestingly, the distribution of nociceptors that exhibit SA and AD are not identical, and the development of SA was observed more frequently in nociceptors with low heat thresholds, while AD was found more frequently in nociceptors with high heat thresholds. We also found that SCI resulted in hindpaw edema and elevated cutaneous calcitonin gene-related peptide (CGRP) concentration that were not observed in naïve mice. These results suggest that SCI causes a rapidly developing nociceptor sensitization and peripheral inflammation that may contribute to the early emergence and persistence of chronic SCI pain.
Collapse
Affiliation(s)
- Olivia C. Eller
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Rena N. Stair
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Christopher Neal
- Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, Kansas City, KS, United States
| | - Peter S.N. Rowe
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, United States
- The Kidney Institute & Division of Nephrology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Jennifer Nelson-Brantley
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Erin E. Young
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, KS, United States
- Center for Advancement in Managing Pain, School of Nursing, University of Connecticut, Storrs, CT, United States
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, United States
- Department of Neuroscience, UConn Health, Farmington, CT, United States
| | - Kyle M. Baumbauer
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, KS, United States
- Center for Advancement in Managing Pain, School of Nursing, University of Connecticut, Storrs, CT, United States
- Department of Neuroscience, UConn Health, Farmington, CT, United States
| |
Collapse
|
16
|
Hu L, Jiang GY, Wang YP, Hu ZB, Zhou BY, Zhang L, Song NN, Huang Y, Chai GD, Chen JY, Lang B, Xu L, Liu JL, Li Y, Wang QX, Ding YQ. The role of PTEN in primary sensory neurons in processing itch and thermal information in mice. Cell Rep 2022; 39:110724. [PMID: 35443189 DOI: 10.1016/j.celrep.2022.110724] [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: 08/26/2021] [Revised: 03/03/2022] [Accepted: 03/30/2022] [Indexed: 12/01/2022] Open
Abstract
PTEN is known as a tumor suppressor and plays essential roles in brain development. Here, we report that PTEN in primary sensory neurons is involved in processing itch and thermal information in adult mice. Deletion of PTEN in the dorsal root ganglia (DRG) is achieved in adult Drg11-CreER: PTENflox/flox (PTEN CKO) mice with oral administration of tamoxifen, and CKO mice develop pathological itch and elevated itch responses on exposure to various pruritogens. PTEN deletion leads to ectopic expression of TRPV1 and MrgprA3 in IB4+ non-peptidergic DRG neurons, and the TRPV1 is responsive to capsaicin. Importantly, the elevated itch responses are no longer present in Drg11-CreER: PTENflox/flox: TRPV1flox/flox (PTEN: TRPV1 dCKO) mice. In addition, thermal stimulation is enhanced in PTEN CKO mice but blunted in dCKO mice. PTEN-involved regulation of itch-related gene expression in DRG neurons provides insights for understanding molecular mechanism of itch and thermal sensation at the spinal level.
Collapse
Affiliation(s)
- Ling Hu
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China; Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Guan-Yu Jiang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Ying-Ping Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Zhi-Bin Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Bing-Yao Zhou
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Lei Zhang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Ning-Ning Song
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Ying Huang
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China
| | - Guo-Dong Chai
- Department of Anesthesiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Jia-Yin Chen
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China; Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Bing Lang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Lin Xu
- Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Jun-Ling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Yong Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Qing-Xiu Wang
- Department of Anesthesiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Yu-Qiang Ding
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China; Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
17
|
Hori A, Hotta N, Fukazawa A, Estrada JA, Katanosaka K, Mizumura K, Sato J, Ishizawa R, Kim HK, Iwamoto GA, Vongpatanasin W, Mitchell JH, Smith SA, Mizuno M. Insulin potentiates the response to capsaicin in dorsal root ganglion neurons in vitro and muscle afferents ex vivo in normal healthy rodents. J Physiol 2022; 600:531-545. [PMID: 34967443 PMCID: PMC8810710 DOI: 10.1113/jp282740] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 12/23/2021] [Indexed: 02/03/2023] Open
Abstract
Systemic insulin administration evokes sympathoexcitatory actions, but the mechanisms underlying these observations are unknown. We reported that insulin sensitizes the response of thin-fibre primary afferents, as well as the dorsal root ganglion (DRG) that subserves them, to mechanical stimuli. However, little is known about the effects of insulin on primary neuronal responses to chemical stimuli. TRPV1, whose agonist is capsaicin (CAP), is widely expressed on chemically sensitive metaboreceptors and/or nociceptors. The aim of this investigation was to determine the effects of insulin on CAP-activated currents in small DRG neurons and CAP-induced action potentials in thin-fibre muscle afferents of normal healthy rodents. Additionally, we investigated whether insulin potentiates sympathetic nerve activity (SNA) responses to CAP. In whole-cell patch-clamp recordings from cultured mice DRG neurons in vitro, the fold change in CAP-activated current from pre- to post-application of insulin (n = 13) was significantly (P < 0.05) higher than with a vehicle control (n = 14). Similar results were observed in single-fibre recording experiments ex vivo as insulin potentiated CAP-induced action potentials compared to vehicle controls (n = 9 per group, P < 0.05). Furthermore, insulin receptor blockade with GSK1838705 significantly suppressed the insulin-induced augmentation in CAP-activated currents (n = 13) as well as the response magnitude of CAP-induced action potentials (n = 9). Likewise, the renal SNA response to CAP after intramuscular injection of insulin (n = 8) was significantly (P < 0.05) greater compared to vehicle (n = 9). The findings suggest that insulin potentiates TRPV1 responsiveness to CAP at the DRG and muscle tissue levels, possibly contributing to the augmentation in sympathoexcitation during activities such as physical exercise. KEY POINTS: Evidence suggests insulin centrally activates the sympathetic nervous system, and a chemical stimulus to tissues activates the sympathetic nervous system via thin fibre muscle afferents. Insulin is reported to modulate putative chemical-sensitive channels in the dorsal root ganglion neurons of these afferents. In the present study, it is demonstrated that insulin potentiates the responsiveness of thin fibre afferents to capsaicin at muscle tissue levels as well as at the level of dorsal root ganglion neurons. In addition, it is demonstrated that insulin augments the sympathetic nerve activity response to capsaicin in vivo. These data suggest that sympathoexcitation is peripherally mediated via insulin-induced chemical sensitization. The present study proposes a possible physiological role of insulin in the regulation of chemical sensitivity in somatosensory thin fibre muscle afferents.
Collapse
Affiliation(s)
- Amane Hori
- Graduate School of Life and Health Sciences, Chubu University, Kasugai 487-850, Japan;,Japan Society for the Promotion of Science, Tokyo 102-8472, Japan
| | - Norio Hotta
- Graduate School of Life and Health Sciences, Chubu University, Kasugai 487-850, Japan;,College of Life and Health Sciences, Chubu University, Kasugai 487-850, Japan
| | - Ayumi Fukazawa
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Juan A. Estrada
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kimiaki Katanosaka
- Graduate School of Life and Health Sciences, Chubu University, Kasugai 487-850, Japan;,College of Life and Health Sciences, Chubu University, Kasugai 487-850, Japan
| | - Kazue Mizumura
- Department of Physiology, Nihon University School of Dentistry, Tokyo 101-8310, Japan
| | - Jun Sato
- Graduate School of Life and Health Sciences, Chubu University, Kasugai 487-850, Japan;,College of Life and Health Sciences, Chubu University, Kasugai 487-850, Japan
| | - Rie Ishizawa
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Han-Kyul Kim
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Gary A. Iwamoto
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wanpen Vongpatanasin
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jere H. Mitchell
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Scott A. Smith
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Masaki Mizuno
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
18
|
Nieder C, Rosene DL, Mortazavi F, Oblak AL, Ketten DR. Morphology and unbiased stereology of the lateral superior olive in the short‐beaked common dolphin,
Delphinus delphis
(Cetacea, Delphinidae). J Morphol 2022; 283:446-461. [DOI: 10.1002/jmor.21453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/10/2022] [Accepted: 01/16/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Carolin Nieder
- Institute of Marine Science, University of Auckland, Leigh Marine Laboratory, 160 Goat Island Road, Leigh New Zealand
| | - Douglas L. Rosene
- Department of Anatomy and Neurobiology Boston University School of Medicine 72 East, Concord St (L 1004), Boston Massachusetts
| | - Farzad Mortazavi
- Department of Anatomy and Neurobiology Boston University School of Medicine 72 East, Concord St (L 1004), Boston Massachusetts
| | - Adrian L. Oblak
- Indiana University School of Medicine, Stark Neurosciences Research Institute, Department of Radiology & Imaging Sciences, 320 W. 15th Street Indianapolis IN
| | - Darlene R. Ketten
- Woods Hole Oceanographic Institution, Biology Department, Marine Research Facility, MS #50 Woods Hole MA USA
| |
Collapse
|
19
|
Abstract
When animals walk overground, mechanical stimuli activate various receptors located in muscles, joints, and skin. Afferents from these mechanoreceptors project to neuronal networks controlling locomotion in the spinal cord and brain. The dynamic interactions between the control systems at different levels of the neuraxis ensure that locomotion adjusts to its environment and meets task demands. In this article, we describe and discuss the essential contribution of somatosensory feedback to locomotion. We start with a discussion of how biomechanical properties of the body affect somatosensory feedback. We follow with the different types of mechanoreceptors and somatosensory afferents and their activity during locomotion. We then describe central projections to locomotor networks and the modulation of somatosensory feedback during locomotion and its mechanisms. We then discuss experimental approaches and animal models used to investigate the control of locomotion by somatosensory feedback before providing an overview of the different functional roles of somatosensory feedback for locomotion. Lastly, we briefly describe the role of somatosensory feedback in the recovery of locomotion after neurological injury. We highlight the fact that somatosensory feedback is an essential component of a highly integrated system for locomotor control. © 2021 American Physiological Society. Compr Physiol 11:1-71, 2021.
Collapse
Affiliation(s)
- Alain Frigon
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Quebec, Canada
| | - Turgay Akay
- Department of Medical Neuroscience, Atlantic Mobility Action Project, Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Boris I Prilutsky
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
20
|
Kudo M, Wupuer S, Kubota S, Seki K. Distribution of Large and Small Dorsal Root Ganglion Neurons in Common Marmosets. Front Syst Neurosci 2021; 15:801492. [PMID: 34924967 PMCID: PMC8674680 DOI: 10.3389/fnsys.2021.801492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/12/2021] [Indexed: 11/13/2022] Open
Abstract
The aim of this study was to elucidate the size and distribution of dorsal root ganglion (DRG) neurons in non-human primates and to compare them with those of rodent DRG neurons. By measuring the size of NeuN-, NF200-, and peripherin-positive DRG neurons in the lumbar spinal cord of rats and marmosets, we found that the cell size distribution pattern was comparable in both species, although DRG neurons in marmosets were larger than those of rodents. This is the first demonstration that DRG neurons in marmosets have a bimodal size distribution, which has been well established in rodents and humans.
Collapse
Affiliation(s)
- Moeko Kudo
- Department of Neurophysiology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Sidikejiang Wupuer
- Department of Neurophysiology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Shinji Kubota
- Department of Neurophysiology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Kazuhiko Seki
- Department of Neurophysiology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| |
Collapse
|
21
|
Yang Z, Yang Y, Xu Y, Jiang W, Shao Y, Xing J, Chen Y, Han Y. Biomimetic nerve guidance conduit containing engineered exosomes of adipose-derived stem cells promotes peripheral nerve regeneration. Stem Cell Res Ther 2021; 12:442. [PMID: 34362437 PMCID: PMC8343914 DOI: 10.1186/s13287-021-02528-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/18/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Efficient and stable delivery of neurotrophic factors (NTFs) is crucial to provide suitable microenvironment for peripheral nerve regeneration. Neurotrophin-3 (NT-3) is an important NTF during peripheral nerve regeneration which is scarce in the first few weeks of nerve defect. Exosomes are nanovesicles and have been served as promising candidate for biocarrier. In this work, NT-3 mRNA was encapsulated in adipose-derived stem cell (ADSC)-derived exosomes (ExoNT-3). These engineered exosomes were applied as NT-3 mRNA carrier and then were loaded in nerve guidance conduit (ExoNT-3-NGC) to bridge rat sciatic nerve defect. METHOD NT-3 mRNA was encapsulated in exosomes by forcedly expression of NT-3 mRNA in the donor ADSCs. ExoNT-3 were co-cultured with SCs in vitro; after 24 h of culture, the efficiency of NT-3 mRNA delivery was evaluated by qPCR, western blotting and ELISA. Then, ExoNT-3 were loaded in alginate hydrogel to construct the nerve guidance conduits (ExoNT-3-NGC). ExoNT-3-NGC were implanted in vivo to reconstruct 10 mm rat sciatic nerve defect. The expression of NT-3 was measured 2 weeks after the implantation operation. The sciatic nerve functional index (SFI) was examined at 2 and 8 weeks after the operation. Moreover, the therapeutic effect of ExoNT-3-NGC was also evaluated by morphology assay, immunofluorescence staining of regenerated nerves, function evaluation of gastrocnemius muscles after 8 weeks of implantation. RESULTS The engineered exosomes could deliver NT-3 mRNA to the recipient cells efficiently and translated into functional protein. The constructed NGC could realize stable release of exosomes at least for 2 weeks. After NGC implantation in vivo, ExoNT-3-NGC group significantly promote nerve regeneration and improve the function recovery of gastrocnemius muscles compared with control exosomes (Exoempty-NGC) group. CONCLUSION In this work, NGC was constructed to allow exosome-mediated NT-3 mRNA delivery. After ExoNT-3-NGC implantation in vivo, the level of NT-3 could restore which enhance the nerve regeneration. Our study provide a potential approach to improve nerve regeneration.
Collapse
Affiliation(s)
- Zheng Yang
- Department of Plastic Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.,Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yang Yang
- Xi'an Daxing Hospital, Xi'an, 710016, Shaanxi, China
| | - Yichi Xu
- Department of Plastic Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Weiqian Jiang
- Department of Plastic Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.,Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yan Shao
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jiahua Xing
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Youbai Chen
- Department of Plastic Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yan Han
- Department of Plastic Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
22
|
Liao C, Zhou H, Chen H, Yang X, Zhong W, Zhang W. Patterns of nerve fiber impairments and neuronal activation in male diabetic rats with and without mechanical allodynia: a comparative study. Can J Diabetes 2021; 46:157-164. [DOI: 10.1016/j.jcjd.2021.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 06/28/2021] [Accepted: 08/05/2021] [Indexed: 11/17/2022]
|
23
|
Jakob MO, Kofoed-Branzk M, Deshpande D, Murugan S, Klose CSN. An Integrated View on Neuronal Subsets in the Peripheral Nervous System and Their Role in Immunoregulation. Front Immunol 2021; 12:679055. [PMID: 34322118 PMCID: PMC8312561 DOI: 10.3389/fimmu.2021.679055] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/15/2021] [Indexed: 12/21/2022] Open
Abstract
The peripheral nervous system consists of sensory circuits that respond to external and internal stimuli and effector circuits that adapt physiologic functions to environmental challenges. Identifying neurotransmitters and neuropeptides and the corresponding receptors on immune cells implies an essential role for the nervous system in regulating immune reactions. Vice versa, neurons express functional cytokine receptors to respond to inflammatory signals directly. Recent advances in single-cell and single-nuclei sequencing have provided an unprecedented depth in neuronal analysis and allowed to refine the classification of distinct neuronal subsets of the peripheral nervous system. Delineating the sensory and immunoregulatory capacity of different neuronal subsets could inform a better understanding of the response happening in tissues that coordinate physiologic functions, tissue homeostasis and immunity. Here, we summarize current subsets of peripheral neurons and discuss neuronal regulation of immune responses, focusing on neuro-immune interactions in the gastrointestinal tract. The nervous system as a central coordinator of immune reactions and tissue homeostasis may predispose for novel promising therapeutic approaches for a large variety of diseases including but not limited to chronic inflammation.
Collapse
Affiliation(s)
- Manuel O Jakob
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Michael Kofoed-Branzk
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Divija Deshpande
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Shaira Murugan
- Department of BioMedical Research, Group of Visceral Surgery and Medicine, University of Bern, Bern, Switzerland
| | - Christoph S N Klose
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
24
|
Malavasi EL, Ghosh A, Booth DG, Zagnoni M, Sherman DL, Brophy PJ. Dynamic early clusters of nodal proteins contribute to node of Ranvier assembly during myelination of peripheral neurons. eLife 2021; 10:68089. [PMID: 34240706 PMCID: PMC8289411 DOI: 10.7554/elife.68089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/07/2021] [Indexed: 12/31/2022] Open
Abstract
Voltage-gated sodium channels cluster in macromolecular complexes at nodes of Ranvier to promote rapid nerve impulse conduction in vertebrate nerves. Node assembly in peripheral nerves is thought to be initiated at heminodes at the extremities of myelinating Schwann cells, and fusion of heminodes results in the establishment of nodes. Here we show that assembly of 'early clusters' of nodal proteins in the murine axonal membrane precedes heminode formation. The neurofascin (Nfasc) proteins are essential for node assembly, and the formation of early clusters also requires neuronal Nfasc. Early clusters are mobile and their proteins are dynamically recruited by lateral diffusion. They can undergo fusion not only with each other but also with heminodes, thus contributing to the development of nodes in peripheral axons. The formation of early clusters constitutes the earliest stage in peripheral node assembly and expands the repertoire of strategies that have evolved to establish these essential structures.
Collapse
Affiliation(s)
- Elise Lv Malavasi
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Aniket Ghosh
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Daniel G Booth
- Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Michele Zagnoni
- Centre for Microsystems & Photonics, Dept. Electronic and Electrical Engineering, University of Strathclyde, Strathclyde, United Kingdom
| | - Diane L Sherman
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter J Brophy
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
25
|
Li Q, Qin L, Li J. IL-6 signaling pathway contributes to exercise pressor reflex in rats with femoral artery occlusion in association with Kv4 activity in muscle afferent nerves. Physiol Rep 2021; 9:e14935. [PMID: 34231965 PMCID: PMC8261684 DOI: 10.14814/phy2.14935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 05/26/2021] [Indexed: 12/02/2022] Open
Abstract
Interleukin-6 (IL-6) via trans-signaling pathway plays a role in modifying muscle sensory nerve-exaggerated exercise pressor reflex in rats with ligated femoral arteries, but the underlying mechanisms are poorly understood. It is known that voltage-gated potassium channel subfamily member Kv4 channels contribute to the excitabilities of sensory neurons and neuronal signaling transduction. Thus, in this study, we determined that 1) IL-6 regulates the exaggerated exercise pressor reflex in rats with peripheral artery disease (PAD) induced by femoral artery ligation and 2) Kv4 channels in muscle dorsal root ganglion (DRG) neurons are engaged in the role played by IL-6 trans-signaling pathway. We found that the protein levels of IL-6 and its receptor IL-6R expression were increased in the DRGs of PAD rats with 3-day of femoral artery occlusion. Inhibition of muscle afferents' IL-6 trans-signaling pathway (gp130) by intra-arterial administration of SC144, a gp130 inhibitor, into the hindlimb muscles of PAD rats alleviated blood pressure response to static muscle contraction. On the other hand, we found that 3-day femoral occlusion decreased amplitude of Kv4 currents in rat muscle DRG neurons. The homo IL-6/IL-6Rα fusion protein (H. IL-6/6Rα), but not IL-6 alone significantly inhibited Kv4 currents in muscle DRG neurons; and the effect of H. IL-6/6Rα was largely reverted by SC144. In conclusion, our data suggest that via trans-signaling pathway upregulated IL-6 in muscle afferent nerves by ischemic hindlimb muscles inhibits the activity of Kv4 channels and thus likely leads to adjustments of the exercise pressor reflex in PAD.
Collapse
Affiliation(s)
- Qin Li
- Heart and Vascular InstituteThe Pennsylvania State University College of MedicineHersheyPAUSA
| | - Lu Qin
- Heart and Vascular InstituteThe Pennsylvania State University College of MedicineHersheyPAUSA
| | - Jianhua Li
- Heart and Vascular InstituteThe Pennsylvania State University College of MedicineHersheyPAUSA
| |
Collapse
|
26
|
Li Q, Qin L, Li J. Effects of bradykinin on voltage-gated K V 4 channels in muscle dorsal root ganglion neurons of rats with experimental peripheral artery disease. J Physiol 2021; 599:3567-3580. [PMID: 34036586 PMCID: PMC8284427 DOI: 10.1113/jp281704] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 05/19/2021] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS During exercise, bradykinin (BK), a muscle metabolite in ischaemic muscles, exaggerates autonomic responses to activation of muscle afferent nerves in peripheral artery disease (PAD). We examined whether BK inhibits activity of KV 4 channels in muscle afferent neurons of PAD rats induced by femoral artery occlusion. We demonstrated that: 1) femoral occlusion attenuates KV 4 currents in dorsal root ganglion (DRG) neurons innervating the hindlimb muscles and decreases the threshold of action potential firing; 2) BK has a greater inhibitory effect on KV 4 currents in muscle DRG neurons of PAD rats; and 3) expression of KV 4.3 is downregulated in DRGs of PAD rats and inhibition of KV 4.3 significantly decreases activity of KV 4 currents in muscle DRG neurons. Femoral artery occlusion-induced limb ischaemia and/or ischaemia-induced metabolites (i.e. BK) inhibit activity of KV 4 channels in muscle afferent neurons and this is likely involved in the exaggerated exercise pressor reflex in PAD. ABSTRACT Muscle afferent nerve-activated reflex sympathetic nervous and blood pressure responses are exaggerated during exercise in patients with peripheral artery diseases (PAD) and in PAD rats induced by femoral artery occlusion. However, the precise signalling pathways and molecular mediators responsible for these abnormal autonomic responses in PAD are poorly understood. A-type voltage-gated K+ (KV ) channels are quintessential regulators of cellular excitability in the various tissues. Among KV channels, KV 4 (i.e. KV 4.1 and KV 4.3) in primary sensory neurons mainly participate in physiological functions in regulation of mechanical and chemical sensation. However, little is known about the role of KV 4 in regulating neuronal activity in muscle afferent neurons of PAD. In addition, bradykinin (BK) is considered as a muscle metabolite contributing to the exaggerated exercise pressor reflex in PAD rats with femoral artery occlusion. Our data demonstrated that: 1) KV 4 currents are attenuated in dorsal root ganglion (DRG) neurons innervating the hindlimb muscles of PAD rats, along with a decreasing threshold of action potential firing; 2) KV 4 currents are inhibited by application of BK onto muscle DRG neurons of PAD rats to a greater degree; and 3) expression of KV 4.3 is downregulated in the DRGs of PAD rats and KV 4.3 channel is a major contributor to the activity of KV 4 currents in muscle DRG neurons. In conclusion, data suggest that femoral artery occlusion-induced limb ischaemia and/or ischaemia-induced metabolites (i.e. BK) inhibit the activity of KV 4 channels in muscle afferent neurons likely leading to the exaggerated exercise pressor reflex observed in PAD.
Collapse
Affiliation(s)
- Qin Li
- Heart and Vascular Institute, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Lu Qin
- Heart and Vascular Institute, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Jianhua Li
- Heart and Vascular Institute, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| |
Collapse
|
27
|
Steverink JG, Oostinga D, van Tol FR, van Rijen MHP, Mackaaij C, Verlinde-Schellekens SAMW, Oosterman BJ, Van Wijck AJM, Roeling TAP, Verlaan JJ. Sensory Innervation of Human Bone: An Immunohistochemical Study to Further Understand Bone Pain. THE JOURNAL OF PAIN 2021; 22:1385-1395. [PMID: 33964414 DOI: 10.1016/j.jpain.2021.04.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/14/2021] [Accepted: 04/23/2021] [Indexed: 11/30/2022]
Abstract
Skeletal diseases and their surgical treatment induce severe pain. The innervation density of bone potentially explains the severe pain reported. Animal studies concluded that sensory myelinated A∂-fibers and unmyelinated C-fibers are mainly responsible for conducting bone pain, and that the innervation density of these nerve fibers was highest in periosteum. However, literature regarding sensory innervation of human bone is scarce. This observational study aimed to quantify sensory nerve fiber density in periosteum, cortical bone, and bone marrow of axial and appendicular human bones using immunohistochemistry and confocal microscopy. Multivariate Poisson regression analysis demonstrated that the total number of sensory and sympathetic nerve fibers was highest in periosteum, followed by bone marrow, and cortical bone for all bones studied. Bone from thoracic vertebral bodies contained most sensory nerve fibers, followed by the upper extremity, lower extremity, and parietal neurocranium. The number of nerve fibers declined with age and did not differ between male and female specimens. Sensory nerve fibers were organized as a branched network throughout the periosteum. The current results provide an explanation for the severe pain accompanying skeletal disease, fracture, or surgery. Further, the results could provide more insight into mechanisms that generate and maintain skeletal pain and might aid in developing new treatment strategies. PERSPECTIVE: This article presents the innervation of human bone and assesses the effect of age, gender, bone compartment and type of bone on innervation density. The presented data provide an explanation for the severity of bone pain arising from skeletal diseases and their surgical treatment.
Collapse
Affiliation(s)
- Jasper G Steverink
- Department of Orthopedic Surgery, University Medical Center Utrecht, The Netherlands; SentryX B.V., Woudenbergseweg 41, Austerlitz, The Netherlands.
| | - Douwe Oostinga
- Department of Orthopedic Surgery, University Medical Center Utrecht, The Netherlands
| | - Floris R van Tol
- Department of Orthopedic Surgery, University Medical Center Utrecht, The Netherlands; SentryX B.V., Woudenbergseweg 41, Austerlitz, The Netherlands
| | - Mattie H P van Rijen
- Department of Orthopedic Surgery, University Medical Center Utrecht, The Netherlands
| | - Claire Mackaaij
- Department of Anatomy, University Medical Center Utrecht, The Netherlands
| | | | - Bas J Oosterman
- SentryX B.V., Woudenbergseweg 41, Austerlitz, The Netherlands
| | - Albert J M Van Wijck
- Department of Anesthesiology, University Medical Center Utrecht, The Netherlands
| | - Tom A P Roeling
- Department of Anatomy, University Medical Center Utrecht, The Netherlands
| | - Jorrit-Jan Verlaan
- Department of Orthopedic Surgery, University Medical Center Utrecht, The Netherlands; SentryX B.V., Woudenbergseweg 41, Austerlitz, The Netherlands
| |
Collapse
|
28
|
Malfait AM, Miller RE, Miller RJ. Basic Mechanisms of Pain in Osteoarthritis: Experimental Observations and New Perspectives. Rheum Dis Clin North Am 2021; 47:165-180. [PMID: 33781488 DOI: 10.1016/j.rdc.2020.12.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The specific changes in the peripheral neuronal pathways underlying joint pain in osteoarthritis are the focus of this review. The plasticity of the nociceptive system in osteoarthritis and how this involves changes in the structural, physiologic, and genetic properties of neurons in pain pathways are discussed. The role of the neurotrophin, nerve growth factor, in these pathogenic processes is discussed. Finally, how neuronal pathways are modified by interaction with the degenerating joint tissues they innervate and with the innate immune system is considered. These extensive cellular interactions provide a substrate for identification of targets for osteoarthritis pain.
Collapse
Affiliation(s)
- Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Room 714, 1735 W Harrison Street, Chicago, IL 60612, USA.
| | - Rachel E Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Room 714, 1735 W Harrison Street, Chicago, IL 60612, USA
| | - Richard J Miller
- Department of Pharmacology, Northwestern University, Searle Building Room 8-510, 320 E Superior Street, Chicago, IL 60611, USA
| |
Collapse
|
29
|
Green DP. The role of Mrgprs in pain. Neurosci Lett 2021; 744:135544. [PMID: 33421487 DOI: 10.1016/j.neulet.2020.135544] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/16/2020] [Accepted: 11/26/2020] [Indexed: 11/17/2022]
Abstract
Owing to their functional diversity, the Mas-related G-protein-coupled receptor (Mrgpr) family has a role in both itch and pain modulation. While primarily linked to pruritis, Mrgprs were originally characterized in small-diameter nociceptive neurons of dorsal root ganglia (DRG) and trigeminal ganglia. This review will focus on the role Mrgpr's have in pain physiology, discussing recent discoveries as well as how Mrgpr's may provide a new target for the treatment of pathological pain.
Collapse
Affiliation(s)
- Dustin P Green
- Department of Neuroscience, Cell Biology, & Anatomy, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
30
|
Shenton FC, Campbell T, Jones JFX, Pyner S. Distribution and morphology of sensory and autonomic fibres in the subendocardial plexus of the rat heart. J Anat 2021; 238:36-52. [PMID: 32783212 PMCID: PMC7754995 DOI: 10.1111/joa.13284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/26/2020] [Accepted: 07/02/2020] [Indexed: 01/11/2023] Open
Abstract
Cardiac reflexes originating from sensory receptors in the heart ensure blood supply to vital tissues and organs in the face of constantly changing demands. Atrial volume receptors are mechanically sensitive vagal afferents which relay to the medulla and hypothalamus, affecting vasopressin release and renal sympathetic activity. To date, two anatomically distinct sensory endings have been identified which may subserve cardiac mechanosensation: end-nets and flower-spray endings. To map the distribution of atrial receptors in the subendocardial space, we have double-labelled rat right atrial whole mounts for neurofilament heavy chain (NFH) and synaptic vesicle protein 2 (SV2) and generated high-resolution maps of the rat subendocardial neural plexus at the cavo-atrial region. In order to elucidate the nature of these fibres, double labelling with synaptophysin (SYN) and either NFH, calcitonin gene-related peptide (CGRP), choline acetyltransferase (ChAT) or tyrosine hydroxylase (TH) was performed. The findings show that subendocardial nerve nets are denser at the superior cavo-atrial junction than the mid-atrial region. Adluminal plexuses had the finest diameters and stained positively for synaptic vesicles (SV2 and SYN), CGRP and TH. These plexuses may represent sympathetic post-ganglionic fibres and/or sensory afferents. The latter are candidate substrates for type B volume receptors which are excited by stretch during atrial filling. Deeper nerve fibres appeared coarser and may be cholinergic (positive staining for ChAT). Flower-spray endings were never observed using immunohistochemistry but were delineated clearly with the intravital stain methylene blue. We suggest that differing nerve fibre structures form the basis by which atrial deformation and hence atrial filling is reflected to the brain.
Collapse
Affiliation(s)
| | - Thomas Campbell
- Discipline of AnatomySchool of MedicineUniversity College DublinDublin 4Ireland
| | - James F. X. Jones
- Discipline of AnatomySchool of MedicineUniversity College DublinDublin 4Ireland
| | - Susan Pyner
- Department of BiosciencesDurham UniversityDurhamUK
| |
Collapse
|
31
|
Kampanis V, Tolou-Dabbaghian B, Zhou L, Roth W, Puttagunta R. Cyclic Stretch of Either PNS or CNS Located Nerves Can Stimulate Neurite Outgrowth. Cells 2020; 10:cells10010032. [PMID: 33379276 PMCID: PMC7824691 DOI: 10.3390/cells10010032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/15/2022] Open
Abstract
The central nervous system (CNS) does not recover from traumatic axonal injury, but the peripheral nervous system (PNS) does. We hypothesize that this fundamental difference in regenerative capacity may be based upon the absence of stimulatory mechanical forces in the CNS due to the protective rigidity of the vertebral column and skull. We developed a bioreactor to apply low-strain cyclic axonal stretch to adult rat dorsal root ganglia (DRG) connected to either the peripheral or central nerves in an explant model for inducing axonal growth. In response, larger diameter DRG neurons, mechanoreceptors and proprioceptors showed enhanced neurite outgrowth as well as increased Activating Transcription Factor 3 (ATF3).
Collapse
Affiliation(s)
- Vasileios Kampanis
- Laboratory for Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, 69118 Heidelberg, Germany; (V.K.); (B.T.-D.)
| | - Bahardokht Tolou-Dabbaghian
- Laboratory for Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, 69118 Heidelberg, Germany; (V.K.); (B.T.-D.)
| | - Luming Zhou
- Laboratory of NeuroRegeneration and Repair, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany;
| | - Wolfgang Roth
- Laboratory for Experimental Neurorehabilitation, Heidelberg University Hospital, 69118 Heidelberg, Germany;
| | - Radhika Puttagunta
- Laboratory for Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, 69118 Heidelberg, Germany; (V.K.); (B.T.-D.)
- Correspondence:
| |
Collapse
|
32
|
An Index Combining Lost and Remaining Nerve Fibers Correlates with Pain Hypersensitivity in Mice. Cells 2020; 9:cells9112414. [PMID: 33158176 PMCID: PMC7694241 DOI: 10.3390/cells9112414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/01/2020] [Accepted: 11/02/2020] [Indexed: 11/17/2022] Open
Abstract
Multiple peripheral nerves are known to degenerate after nerve compression injury but the correlation between the extent of nerve alteration and pain severity remains unclear. Here, we used intravital two-photon fluorescence microscopy to longitudinally observe changes in cutaneous fibers in the hind paw of Nav1.8-Cre-tdTomato mice after chronic constriction injury (CCI). Results showed that the CCI led to variable loss of the skin nerve plexus and intraepidermal nerve fibers. The timing of Nav1.8 nerve fiber loss correlated with the development of mechanical hypersensitivity. We compared a scoring approach that assessed whole-paw nerve degeneration with an index that quantified changes in the nerve plexus and terminals in multiple small regions of interest (ROI) from intravital images of the third and fifth toe tips. We found that the number of surviving nerve fibers was not linearly correlated with mechanical hypersensitivity. On the contrary, at 14 days after CCI, the moderately injured mice showed greater mechanical hypersensitivity than the mildly or severely injured mice. This indicates that both surviving and injured nerves are required for evoked neuropathic pain. In addition, these two methods may have the estimative effect as diagnostic and prognostic biomarkers for the assessment of neuropathic pain.
Collapse
|
33
|
Beul SF, Hilgetag CC. Systematic modelling of the development of laminar projection origins in the cerebral cortex: Interactions of spatio-temporal patterns of neurogenesis and cellular heterogeneity. PLoS Comput Biol 2020; 16:e1007991. [PMID: 33048930 PMCID: PMC7553356 DOI: 10.1371/journal.pcbi.1007991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 05/27/2020] [Indexed: 11/18/2022] Open
Abstract
The architectonic type principle conceptualizes structural connections between brain areas in terms of the relative architectonic differentiation of connected areas. It has previously been shown that spatio-temporal interactions between the time and place of neurogenesis could underlie multiple features of empirical mammalian connectomes, such as projection existence and the distribution of projection strengths. However, so far no mechanistic explanation for the emergence of typically observed laminar patterns of projection origins and terminations has been tested. Here, we expand an in silico model of the developing cortical sheet to explore which factors could potentially constrain the development of laminar projection patterns. We show that manipulations which rely solely on spatio-temporal interactions, namely the relative density of laminar compartments, a delay in the neurogenesis of infragranular layers relative to layer 1, and a delay in the neurogenesis of supragranular layers relative to infragranular layers, do not result in the striking correlation between supragranular contribution to projections and the relative differentiation of areas that is typically observed in the mammalian cortex. In contrast, we find that if we introduce systematic variation in cell-intrinsic properties, coupling them with architectonic differentiation, the resulting laminar projection patterns closely mirror the empirically observed patterns. We also find that the spatio-temporal interactions posited to occur during neurogenesis are necessary for the formation of the characteristic laminar patterns. Hence, our results indicate that the specification of the laminar patterns of projection origins may result from systematic variation in a number of cell-intrinsic properties, superimposed on the previously identified spatio-temporal interactions which are sufficient for the emergence of the architectonic type principle on the level of inter-areal connectivity in silico.
Collapse
Affiliation(s)
- Sarah F Beul
- Institute of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claus C Hilgetag
- Institute of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Neural Systems Laboratory, Department of Health Sciences, Boston University, Boston, Massachusetts, United States of America
| |
Collapse
|
34
|
Changes in expression of Kv7.5 and Kv7.2 channels in dorsal root ganglion neurons in the streptozotocin rat model of painful diabetic neuropathy. Neurosci Lett 2020; 736:135277. [PMID: 32739272 DOI: 10.1016/j.neulet.2020.135277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 11/21/2022]
Abstract
Diabetic peripheral neuropathic pain (DPNP), the most debilitating complication of diabetes mellitus, is resistant to current therapy. The pathogenesis of DPNP is still elusive, but several mechanisms have been proposed including abnormal hyperexcitability of dorsal root ganglion (DRG) neurons. The underlying molecular mechanisms of such aberrant hyperexcitability are incompletely understood. Using the streptozotocin (STZ) rat model of DPNP, we have recently provided evidence implicating neuronal Kv7 channels that normally exert a powerful stabilizing influence on neuronal excitability, in the abnormal hyperexcitability of DRG neurons and in pain hypersensitivity associated with DPNP. In the present immunohistochemical study, we sought to determine whether Kv7.2 and/or Kv7.5 channel expression is altered in DRG neurons in STZ rats. We found 35 days post-STZ: (1) a significant decrease in Kv7.5-immunoreactivity in small (<30 μm) DRG neurons (both IB4 positive and IB4 negative) and medium-sized (30-40 μm) neurons, and (2) a significant increase in Kv7.2-immunoreactivity in small (<30 μm) neurons, and a non-significant increase in medium/large neurons. The decrease in Kv7.5 channel expression in small and medium-sized DRG neurons in STZ rats is likely to contribute to the mechanisms of hyperexcitability of these neurons and thereby to the resulting pain hypersensitivity associated with DPNP. The upregulation of Kv7.2 subunit in small DRG neurons may be an activity dependent compensatory mechanism to limit STZ-induced hyperexcitability of DRG neurons and the associated pain hypersensitivity. The findings support the notion that Kv7 channels may represent a novel target for DPNP treatment.
Collapse
|
35
|
Meerschaert KA, Adelman PC, Friedman RL, Albers KM, Koerber HR, Davis BM. Unique Molecular Characteristics of Visceral Afferents Arising from Different Levels of the Neuraxis: Location of Afferent Somata Predicts Function and Stimulus Detection Modalities. J Neurosci 2020; 40:7216-7228. [PMID: 32817244 PMCID: PMC7534907 DOI: 10.1523/jneurosci.1426-20.2020] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/30/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023] Open
Abstract
Viscera receive innervation from sensory ganglia located adjacent to multiple levels of the brainstem and spinal cord. Here we examined whether molecular profiling could be used to identify functional clusters of colon afferents from thoracolumbar (TL), lumbosacral (LS), and nodose ganglia (NG) in male and female mice. Profiling of TL and LS bladder afferents was also performed. Visceral afferents were back-labeled using retrograde tracers injected into proximal and distal regions of colon or bladder, followed by single-cell qRT-PCR and analysis via an automated hierarchical clustering method. Genes were chosen for assay (32 for bladder; 48 for colon) based on their established role in stimulus detection, regulation of sensitivity/function, or neuroimmune interaction. A total of 132 colon afferents (from NG, TL, and LS ganglia) and 128 bladder afferents (from TL and LS ganglia) were analyzed. Retrograde labeling from the colon showed that NG and TL afferents innervate proximal and distal regions of the colon, whereas 98% of LS afferents only project to distal regions. There were clusters of colon and bladder afferents, defined by mRNA profiling, that localized to either TL or LS ganglia. Mixed TL/LS clustering also was found. In addition, transcriptionally, NG colon afferents were almost completely segregated from colon TL and LS neurons. Furthermore, colon and bladder afferents expressed genes at similar levels, although different gene combinations defined the clusters. These results indicate that genes implicated in both homeostatic regulation and conscious sensations are found at all anatomic levels, suggesting that afferents from different portions of the neuraxis have overlapping functions.SIGNIFICANCE STATEMENT Visceral organs are innervated by sensory neurons whose cell bodies are located in multiple ganglia associated with the brainstem and spinal cord. For the colon, this overlapping innervation is proposed to facilitate visceral sensation and homeostasis, where sensation and pain are mediated by spinal afferents and fear and anxiety (the affective aspects of visceral pain) are the domain of nodose afferents. The transcriptomic analysis performed here reveals that genes implicated in both homeostatic regulation and pain are found in afferents across all ganglia types, suggesting that conscious sensation and homeostatic regulation are the result of convergence, and not segregation, of sensory input.
Collapse
Affiliation(s)
- Kimberly A Meerschaert
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | | | - Robert L Friedman
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Kathryn M Albers
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - H Richard Koerber
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Brian M Davis
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
36
|
Ciglieri E, Vacca M, Ferrini F, Atteya MA, Aimar P, Ficarra E, Di Cataldo S, Merighi A, Salio C. Cytoarchitectural analysis of the neuron-to-glia association in the dorsal root ganglia of normal and diabetic mice. J Anat 2020; 237:988-997. [PMID: 32579747 DOI: 10.1111/joa.13252] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 11/28/2022] Open
Abstract
Dorsal root ganglia (DRGs) host the somata of sensory neurons which convey information from the periphery to the central nervous system. These neurons have heterogeneous size and neurochemistry, and those of small-to-medium size, which play an important role in nociception, form two distinct subpopulations based on the presence (peptidergic) or absence (non-peptidergic) of transmitter neuropeptides. Few investigations have so far addressed the spatial relationship between neurochemically different subpopulations of DRG neurons and glia. We used a whole-mount mouse lumbar DRG preparation, confocal microscopy and computer-aided 3D analysis to unveil that IB4+ non-peptidergic neurons form small clusters of 4.7 ± 0.26 cells, differently from CGRP+ peptidergic neurons that are, for the most, isolated (1.89 ± 0.11 cells). Both subpopulations of neurons are ensheathed by a thin layer of satellite glial cells (SGCs) that can be observed after immunolabeling with the specific marker glutamine synthetase (GS). Notably, at the ultrastructural level we observed that this glial layer was discontinuous, as there were patches of direct contact between the membranes of two adjacent IB4+ neurons. To test whether this cytoarchitectonic organization was modified in the diabetic neuropathy, one of the most devastating sensory pathologies, mice were made diabetic by streptozotocin (STZ). In diabetic animals, cluster organization of the IB4+ non-peptidergic neurons was maintained, but the neuro-glial relationship was altered, as STZ treatment caused a statistically significant increase of GS staining around CGRP+ neurons but a reduction around IB4+ neurons. Ultrastructural analysis unveiled that SGC coverage was increased at the interface between IB4+ cluster-forming neurons in diabetic mice, with a 50% reduction in the points of direct contacts between cells. These observations demonstrate the existence of a structural plasticity of the DRG cytoarchitecture in response to STZ.
Collapse
Affiliation(s)
- Elisa Ciglieri
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy.,Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Maurizia Vacca
- Department of Control and Computer Engineering, Politecnico di Torino, Torino, Italy
| | - Francesco Ferrini
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy.,Department of Psychiatry & Neuroscience, Université Laval, Québec, QC, Canada
| | - Mona A Atteya
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Patrizia Aimar
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Elisa Ficarra
- Department of Control and Computer Engineering, Politecnico di Torino, Torino, Italy
| | - Santa Di Cataldo
- Department of Control and Computer Engineering, Politecnico di Torino, Torino, Italy
| | - Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy.,National Institute of Neuroscience, Grugliasco, Italy
| | - Chiara Salio
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| |
Collapse
|
37
|
Roh J, Hwang SM, Lee SH, Lee K, Kim YH, Park CK. Functional Expression of Piezo1 in Dorsal Root Ganglion (DRG) Neurons. Int J Mol Sci 2020; 21:ijms21113834. [PMID: 32481599 PMCID: PMC7313462 DOI: 10.3390/ijms21113834] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/25/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022] Open
Abstract
Piezo channels are mechanosensitive ion channels. Piezo1 is primarily expressed in nonsensory tissues, whereas Piezo2 is predominantly found in sensory tissues, including dorsal root ganglion (DRG) neurons. However, a recent study demonstrated the intracellular calcium response to Yoda1, a selective Piezo1 agonist, in trigeminal ganglion (TG) neurons. Herein, we investigate the expression of Piezo1 mRNA and protein in mouse and human DRG neurons and the activation of Piezo1 via calcium influx by Yoda1. Yoda1 induces inward currents mainly in small- (< 25 μm) and medium-sized (25-35 μm) mouse DRG neurons. The Yoda1-induced Ca2+ response is inhibited by cationic channel blocker, ruthenium red and cationic mechanosensitive channel blocker, GsMTx4. To confirm the specific inhibition of Piezo1, we performed an adeno-associated virus serotype 2/5 (AAV2/5)-mediated delivery of short hairpin RNA (shRNA) into mouse DRG neurons. AAV2/5 transfection downregulates piezo1 mRNA expression and reduces Ca2+ response by Yoda1. Piezo1 also shows physiological functions with transient receptor potential vanilloid 1 (TRPV1) in the same DRG neurons and is regulated by the activation of TRPV1 in mouse DRG sensory neurons. Overall, we found that Piezo1 has physiological functions in DRG neurons and that TRPV1 activation inhibits an inward current induced by Yoda1.
Collapse
Affiliation(s)
- Jueun Roh
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Korea; (J.R.); (S.-M.H.); (K.L.)
| | - Sung-Min Hwang
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Korea; (J.R.); (S.-M.H.); (K.L.)
| | - Sun-Ho Lee
- Department of Neurosurgery, Spine tumor center, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea;
| | - Kihwan Lee
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Korea; (J.R.); (S.-M.H.); (K.L.)
| | - Yong Ho Kim
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Korea; (J.R.); (S.-M.H.); (K.L.)
- Correspondence: (Y.H.K.); (C.-K.P.); Tel.: +82-32-899-6691 (Y.H.K.); +82-32-899-6692 (C.-K.P.)
| | - Chul-Kyu Park
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Korea; (J.R.); (S.-M.H.); (K.L.)
- Correspondence: (Y.H.K.); (C.-K.P.); Tel.: +82-32-899-6691 (Y.H.K.); +82-32-899-6692 (C.-K.P.)
| |
Collapse
|
38
|
Oostinga D, Steverink JG, van Wijck AJM, Verlaan JJ. An understanding of bone pain: A narrative review. Bone 2020; 134:115272. [PMID: 32062002 DOI: 10.1016/j.bone.2020.115272] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 12/20/2022]
Abstract
Skeletal pathologies are often accompanied by bone pain, which has negative effects on the quality of life and functional status of patients. Bone pain can be caused by a wide variety of injuries and diseases including (poorly healed) fractures, bone cancer, osteoarthritis and also iatrogenic by skeletal interventions. Orthopedic interventions are considered to be the most painful surgical procedures overall. Two major groups of medication currently used to attenuate bone pain are NSAIDs and opioids. However, these systemic drugs frequently introduce adverse events, emphasizing the need for alternative therapies that are directed at the pathophysiological mechanisms underlying bone pain. The periosteum, cortical bone and bone marrow are mainly innervated by sensory A-delta fibers and C-fibers. These fibers are mostly present in the periosteum rendering this structure most sensitive to nociceptive stimuli. A-delta fibers and C-fibers can be activated upon mechanical distortion, acidic environment and increased intramedullary pressure. After activation, these fibers can be sensitized by inflammatory mediators, phosphorylation of acid-sensing ion channels and cytokine receptors, or by upregulation of transcription factors. This can result in a change of pain perception such that normally non-noxious stimuli are now perceived as noxious. Pathological conditions in the bone can produce neurotrophic factors that bind to receptors on A-delta fibers and C-fibers. These fibers then start to sprout and increase the innervation density of the bone, making it more sensitive to nociceptive stimuli. In addition, repetitive painful stimuli cause neurochemical and electrophysiological alterations in afferent sensory neurons in the spinal cord, which leads to central sensitization, and can contribute to chronic bone pain. Understanding the pathophysiological mechanisms underlying bone pain in different skeletal injuries and diseases is important for the development of alternative, targeted pain treatments. These pain mechanism-based alternatives have the potential to improve the quality of life of patients suffering from bone pain without introducing undesirable systemic effects.
Collapse
Affiliation(s)
- Douwe Oostinga
- Department of Orthopedics, University Medical Centre Utrecht, Heidelberglaan 100, 3508 GA Utrecht, the Netherlands.
| | - Jasper G Steverink
- Department of Orthopedics, University Medical Centre Utrecht, Heidelberglaan 100, 3508 GA Utrecht, the Netherlands.
| | - Albert J M van Wijck
- Department of Anesthesiology, University Medical Centre Utrecht, Heidelberglaan 100, 3508 GA Utrecht, the Netherlands.
| | - Jorrit-Jan Verlaan
- Department of Orthopedics, University Medical Centre Utrecht, Heidelberglaan 100, 3508 GA Utrecht, the Netherlands.
| |
Collapse
|
39
|
Lázár BA, Jancsó G, Sántha P. Modulation of Sensory Nerve Function by Insulin: Possible Relevance to Pain, Inflammation and Axon Growth. Int J Mol Sci 2020; 21:E2507. [PMID: 32260335 PMCID: PMC7177741 DOI: 10.3390/ijms21072507] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 12/16/2022] Open
Abstract
Insulin, besides its pivotal role in energy metabolism, may also modulate neuronal processes through acting on insulin receptors (InsRs) expressed by neurons of both the central and the peripheral nervous system. Recently, the distribution and functional significance of InsRs localized on a subset of multifunctional primary sensory neurons (PSNs) have been revealed. Systematic investigations into the cellular electrophysiology, neurochemistry and morphological traits of InsR-expressing PSNs indicated complex functional interactions among specific ion channels, proteins and neuropeptides localized in these neurons. Quantitative immunohistochemical studies have revealed disparate localization of the InsRs in somatic and visceral PSNs with a dominance of InsR-positive neurons innervating visceral organs. These findings suggested that visceral spinal PSNs involved in nociceptive and inflammatory processes are more prone to the modulatory effects of insulin than somatic PSNs. Co-localization of the InsR and transient receptor potential vanilloid 1 (TRPV1) receptor with vasoactive neuropeptides calcitonin gene-related peptide and substance P bears of crucial importance in the pathogenesis of inflammatory pathologies affecting visceral organs, such as the pancreas and the urinary bladder. Recent studies have also revealed significant novel aspects of the neurotrophic propensities of insulin with respect to axonal growth, development and regeneration.
Collapse
Affiliation(s)
| | - Gábor Jancsó
- Department of Physiology, University of Szeged, H-6720 Szeged, Hungary; (G.J.); (P.S.)
| | - Péter Sántha
- Department of Physiology, University of Szeged, H-6720 Szeged, Hungary; (G.J.); (P.S.)
| |
Collapse
|
40
|
Barry DM, Liu XT, Liu B, Liu XY, Gao F, Zeng X, Liu J, Yang Q, Wilhelm S, Yin J, Tao A, Chen ZF. Exploration of sensory and spinal neurons expressing gastrin-releasing peptide in itch and pain related behaviors. Nat Commun 2020; 11:1397. [PMID: 32170060 PMCID: PMC7070094 DOI: 10.1038/s41467-020-15230-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 02/27/2020] [Indexed: 12/12/2022] Open
Abstract
Gastrin-releasing peptide (GRP) functions as a neurotransmitter for non-histaminergic itch, but its site of action (sensory neurons vs spinal cord) remains controversial. To determine the role of GRP in sensory neurons, we generated a floxed Grp mouse line. We found that conditional knockout of Grp in sensory neurons results in attenuated non-histaminergic itch, without impairing histamine-induced itch. Using a Grp-Cre knock-in mouse line, we show that the upper epidermis of the skin is exclusively innervated by GRP fibers, whose activation via optogeneics and chemogenetics in the skin evokes itch- but not pain-related scratching or wiping behaviors. In contrast, intersectional genetic ablation of spinal Grp neurons does not affect itch nor pain transmission, demonstrating that spinal Grp neurons are dispensable for itch transmission. These data indicate that GRP is a neuropeptide in sensory neurons for non-histaminergic itch, and GRP sensory neurons are dedicated to itch transmission.
Collapse
Affiliation(s)
- Devin M Barry
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xue-Ting Liu
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Center for Immunology, Inflammation, Immune-mediated disease, Guangzhou Medical University, 510260, Guangzhou, Guangdong, P.R. China
| | - Benlong Liu
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xian-Yu Liu
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Fang Gao
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xiansi Zeng
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- College of Life Sciences, Xinyang Normal University, 237 Nanhu Road, 464000, Xinyang, P. R. China
| | - Juan Liu
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Qianyi Yang
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Steven Wilhelm
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jun Yin
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ailin Tao
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Center for Immunology, Inflammation, Immune-mediated disease, Guangzhou Medical University, 510260, Guangzhou, Guangdong, P.R. China
| | - Zhou-Feng Chen
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
41
|
Morellini N, Phillips JK, Wall RV, Drummond PD. Expression of the noradrenaline transporter in the peripheral nervous system. J Chem Neuroanat 2019; 104:101742. [PMID: 31891756 DOI: 10.1016/j.jchemneu.2019.101742] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 12/13/2019] [Accepted: 12/27/2019] [Indexed: 11/22/2022]
Abstract
The noradrenaline transporter (NAT) transfers noradrenaline released into the synaptic cleft back into the presynaptic terminal, thus terminating neurotransmission. Although the distribution of NAT within the central nervous system has been well-characterized, less is known about its distribution elsewhere in the peripheral nervous system and in organs such as the skin. To address this in the present study, NAT expression was investigated using immunohistochemistry in the hind paw skin and more proximally in the sciatic nerve, dorsal root ganglia and spinal cord of five male Wistar rats. It was hypothesised that NAT would be expressed exclusively on nerve fibres labelled by dopamine beta hydroxylase (DβH), an enzyme involved in the conversion of dopamine to noradrenaline. NAT co-localised with DβH in neurons in the spinal cord, dorsal root ganglia and sciatic nerve. Unexpectedly, however, NAT-like immunoreactivity was not observed in DβH immuno-reactive fibres that innervated dermal blood vessels, suggesting that a mechanism other than presynaptic re-uptake of noradrenaline through NAT regulates transmission at neurovascular junctions in the skin. Furthermore, a novel association between NAT-like immunoreactivity and the myelin marker myelin basic protein (MBP) was identified in peripheral nerves. Specifically, NAT and MBP appeared to congregate around primary afferent nerve fibres labelled by neurofilament 200, a marker of neurons with medium- and large-diameter axons. NAT-like immunoreactivity was also detected in cultured Schwann cells immunohistochemically and at the mRNA level. Together, these findings imply a hitherto unrecognised role of Schwann cells in clearance of noradrenaline in the peripheral nervous system.
Collapse
Affiliation(s)
- Natalie Morellini
- College of Science, Health, Engineering and Education, Murdoch University, Perth, Western Australia, Australia, and School of Medicine, University of Notre Dame, Fremantle, Western Australia, Australia
| | | | - Roshana Vander Wall
- Department of Biomedical Science, Macquarie University, Sydney, NSW, Australia
| | - Peter D Drummond
- College of Science, Health, Engineering and Education, Murdoch University, Perth, Western Australia, Australia, and School of Medicine, University of Notre Dame, Fremantle, Western Australia, Australia.
| |
Collapse
|
42
|
Hou Y, Wang X, Zhang Z, Luo J, Cai Z, Wang Y, Li Y. Repairing Transected Peripheral Nerve Using a Biomimetic Nerve Guidance Conduit Containing Intraluminal Sponge Fillers. Adv Healthc Mater 2019; 8:e1900913. [PMID: 31583854 DOI: 10.1002/adhm.201900913] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/11/2019] [Indexed: 12/29/2022]
Abstract
Nerve guide conduits (NGCs) with geometric design have shown significant advantages in guidance of nerve reinnervation across the defect of injured peripheral nerves. It is realized that intraluminal fillers with distinctive structure can effectively provide an inner guidance for sprouting of axons and improve the permeability of NGC. In this work, a poly(lactic-co-glycolic acid) (PLGA) NGC is prepared containing intraluminal sponge fillers (labeled as ISF-NGC) and used for reconstruction of a rat sciatic nerve with a 10 mm gap. For comparison, the same procedure is applied to a single hollow PLGA NGC (labeled as H-NGC) and an autologous nerve. As evidenced by significantly improved nerve morphology and function, the ISF-NGC achieves a superior nerve repair effect over H-NGC, which is comparable to autologous nerve grafting. It is likely that the H-NGC only provides a protected tunnel for nerve fiber regrowth and axonal extension, while ISF-NGC offers an extracellular matrix-mimetic architecture as autograft to provide contact guidance for nerve reinnervation. This newly developed ISF-NGC is a promising candidate to aid nerve reinnervation across longer gaps commonly encountered in clinical cases.
Collapse
Affiliation(s)
- Yuanjing Hou
- State Key Laboratory of Advanced Technology for Materials Synthesis and ProcessingWuhan University of Technology Wuhan 430070 China
- Biomedical Materials and Engineering Research Center of Hubei ProvinceWuhan University of Technology Wuhan 430070 China
| | - Xinyu Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and ProcessingWuhan University of Technology Wuhan 430070 China
- Biomedical Materials and Engineering Research Center of Hubei ProvinceWuhan University of Technology Wuhan 430070 China
| | - Zongrui Zhang
- College of Biochemical EngineeringAnhui Polytechnic University Wuhu 241000 China
| | - Jing Luo
- State Key Laboratory of Advanced Technology for Materials Synthesis and ProcessingWuhan University of Technology Wuhan 430070 China
- Biomedical Materials and Engineering Research Center of Hubei ProvinceWuhan University of Technology Wuhan 430070 China
| | - Zhengwei Cai
- State Key Laboratory of Advanced Technology for Materials Synthesis and ProcessingWuhan University of Technology Wuhan 430070 China
- Biomedical Materials and Engineering Research Center of Hubei ProvinceWuhan University of Technology Wuhan 430070 China
| | - Yiyu Wang
- School of Life Science TechnologyHubei Engineering University Xiaogan 432000 China
| | - Yi Li
- Institute of Textiles and ClothingThe Hong Kong Polytechnic University Hung Hom Kowloon Hong Kong 999077 China
| |
Collapse
|
43
|
Lawson SN, Fang X, Djouhri L. Nociceptor subtypes and their incidence in rat lumbar dorsal root ganglia (DRGs): focussing on C-polymodal nociceptors, Aβ-nociceptors, moderate pressure receptors and their receptive field depths. CURRENT OPINION IN PHYSIOLOGY 2019; 11:125-146. [PMID: 31956744 PMCID: PMC6959836 DOI: 10.1016/j.cophys.2019.10.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
A recent study with Ca++-sensitive-dyes in neurons in whole DRGs (Table 5) found that much lower percentages of nociceptors were polymodal-nociceptors (PMNs) (Emery et al., 2016), than the 50-80% values in many electrophysiological fiber studies. This conflict highlighted the lack of knowledge about percentages of nociceptor-subtypes in the DRG. This was analysed from intracellularly-recorded neurons in rat lumbar DRGs stimulated from outside the skin. Polymodal nociceptors (PMNs) were 11% of all neurons and 19% of all nociceptors. Most PMNs had C-fibers (CPMNs). Percentages of C-nociceptors that were CPMNs varied with receptive field (RF) depths, whether superficial (∼80%), dermal (25%), deep (0%) or cutaneous (superficial + dermal) (40%). This explains CPMN percentages 40-90%, being highest, in electrophysiological studies using cutaneous nerves, and lowest in studies that also include deep RFs, including ours, and the recent Ca++-imaging studies in whole DRGs. Despite having been originally described in 1967 (Burgess and Perl), both Aβ-nociceptors and Aβ-moderate pressure receptors (MPRs) remain overlooked. Most A-fiber nociceptors in rodents have Aβ-fibers. Of rat lumbar Aβ-nociceptors with superficial RFs, 50% were MPRs with variable medium-low trkA-expression. Despite having conduction velocities at the two extremes for nociceptors, both CPMNs and MPRs have relatively low thresholds, superficial/epidermal RFs and low trkA-expression. For abbreviations used see Table 5.
Collapse
Affiliation(s)
- Sally N Lawson
- The Physiology Department, University of Bristol, Bristol BS8 1TD, UK
| | - Xin Fang
- Qihan BioTech Co. Ltd, Hangzhou, China
| | - Laiche Djouhri
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
44
|
Hotta N, Katanosaka K, Mizumura K, Iwamoto GA, Ishizawa R, Kim HK, Vongpatanasin W, Mitchell JH, Smith SA, Mizuno M. Insulin potentiates the response to mechanical stimuli in small dorsal root ganglion neurons and thin fibre muscle afferents in vitro. J Physiol 2019; 597:5049-5062. [PMID: 31468522 DOI: 10.1113/jp278527] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 08/27/2019] [Indexed: 01/23/2023] Open
Abstract
KEY POINTS Insulin is known to activate the sympathetic nervous system centrally. A mechanical stimulus to tissues activates the sympathetic nervous system via thin fibre afferents. Evidence suggests that insulin modulates putative mechanosensitive channels in the dorsal root ganglion neurons of these afferents. In the present study, we report the novel finding that insulin augments the mechanical responsiveness of thin fibre afferents not only at dorsal root ganglion, but also at muscle tissue levels. Our data suggest that sympathoexcitation is mediated via the insulin-induced mechanical sensitization peripherally. The present study proposes a novel physiological role of insulin in the regulation of mechanical sensitivity in somatosensory thin fibre afferents. ABSTRACT Insulin activates the sympathetic nervous system, although the mechanism underlying insulin-induced sympathoexcitation remains to be determined. A mechanical stimulus to tissues such as skin and/or skeletal muscle, no matter whether the stimulation is noxious or not, activates the sympathetic nervous system via thin fibre afferents. Evidence suggests that insulin modulates putative mechanosensitive channels in the dorsal root ganglion (DRG) neurons of these afferents. Accordingly, we investigated whether insulin augments whole-cell current responses to mechanical stimuli in small DRG neurons of normal healthy mice. We performed whole-cell patch clamp recordings using cultured DRG neurons and observed mechanically-activated (MA) currents induced by mechanical stimuli applied to the cell surface. Local application of vehicle solution did not change MA currents or mechanical threshold in cultured DRG neurons. Insulin (500 mU mL-1 ) significantly augmented the amplitude of MA currents (P < 0.05) and decreased the mechanical threshold (P < 0.05). Importantly, pretreatment with the insulin receptor antagonist, GSK1838705, significantly suppressed the insulin-induced potentiation of the mechanical response. We further examined the impact of insulin on thin fibre muscle afferent activity in response to mechanical stimuli in normal healthy rats in vitro. Using a muscle-nerve preparation, we recorded single group IV fibre activity to a ramp-shaped mechanical stimulation. Insulin significantly decreased mechanical threshold (P < 0.05), although it did not significantly increase the response magnitude to the mechanical stimulus. In conclusion, these data suggest that insulin augments the mechanical responsiveness of small DRG neurons and potentially sensitizes group IV afferents to mechanical stimuli at the muscle tissue level, possibly contributing to insulin-induced sympathoexcitation.
Collapse
Affiliation(s)
- Norio Hotta
- College of Life and Health Sciences, Chubu University, Kasugai, Japan.,Departments of Health Care Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Kazue Mizumura
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Gary A Iwamoto
- Departments of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rie Ishizawa
- Departments of Health Care Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Han-Kyul Kim
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wanpen Vongpatanasin
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jere H Mitchell
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Scott A Smith
- Departments of Health Care Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Masaki Mizuno
- Departments of Health Care Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
45
|
Increased Expression of Fibronectin Leucine-Rich Transmembrane Protein 3 in the Dorsal Root Ganglion Induces Neuropathic Pain in Rats. J Neurosci 2019; 39:7615-7627. [PMID: 31346030 DOI: 10.1523/jneurosci.0295-19.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/17/2019] [Accepted: 06/06/2019] [Indexed: 01/19/2023] Open
Abstract
Neuropathic pain is a chronic condition that occurs frequently after nerve injury and induces hypersensitivity or allodynia characterized by aberrant neuronal excitability in the spinal cord dorsal horn. Fibronectin leucine-rich transmembrane protein 3 (FLRT3) is a modulator of neurite outgrowth, axon pathfinding, and cell adhesion, which is upregulated in the dorsal horn following peripheral nerve injury. However, the function of FLRT3 in adults remains unknown. Therefore, we aimed to investigate the involvement of spinal FLRT3 in neuropathic pain using rodent models. In the dorsal horns of male rats, FLRT3 protein levels increased at day 4 after peripheral nerve injury. In the DRG, FLRT3 was expressed in activating transcription factor 3-positive, injured sensory neurons. Peripheral nerve injury stimulated Flrt3 transcription in the DRG but not in the spinal cord. Intrathecal administration of FLRT3 protein to naive rats induced mechanical allodynia and GluN2B phosphorylation in the spinal cord. DRG-specific FLRT3 overexpression using adeno-associated virus also produced mechanical allodynia. Conversely, a function-blocking FLRT3 antibody attenuated mechanical allodynia after partial sciatic nerve ligation. Therefore, FLRT3 derived from injured DRG neurons increases dorsal horn excitability and induces mechanical allodynia.SIGNIFICANCE STATEMENT Neuropathic pain occurs frequently after nerve injury and is associated with abnormal neuronal excitability in the spinal cord. Fibronectin leucine-rich transmembrane protein 3 (FLRT3) regulates neurite outgrowth and cell adhesion. Here, nerve injury increased FLRT3 protein levels in the spinal cord dorsal root, despite the fact that Flrt3 transcripts were only induced in the DRG. FLRT3 protein injection into the rat spinal cord induced mechanical hypersensitivity, as did virus-mediated FLRT3 overexpression in DRG. Conversely, FLRT3 inhibition with antibodies attenuated mechanically induced pain after nerve damage. These findings suggest that FLRT3 is produced by injured DRG neurons and increases neuronal excitability in the dorsal horn, leading to pain sensitization. Neuropathic pain induction is a novel function of FLRT3.
Collapse
|
46
|
Wang J, La JH, Hamill OP. PIEZO1 Is Selectively Expressed in Small Diameter Mouse DRG Neurons Distinct From Neurons Strongly Expressing TRPV1. Front Mol Neurosci 2019; 12:178. [PMID: 31379500 PMCID: PMC6659173 DOI: 10.3389/fnmol.2019.00178] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 07/04/2019] [Indexed: 11/13/2022] Open
Abstract
Using a high resolution in situ hybridization technique we have measured PIEZO1, PIEZO2, and TRPV1 transcripts in mouse dorsal root ganglion (DRG) neurons. Consistent with previous studies, PIEZO2 transcripts were highly expressed in DRG neurons of all sizes, including most notably the largest diameter neurons implicated in mediating touch and proprioception. In contrast, PIEZO1 transcripts were selectively expressed in smaller DRG neurons, which are implicated in mediating nociception. Moreover, the small neurons expressing PIEZO1 were mostly distinct from those neurons that strongly expressed TRPV1, one of the channels implicated in heat-nociception. Interestingly, while PIEZO1- and TRPV1- expressing neurons form essentially non-overlapping populations, PIEZO2 showed co-expression in both populations. Using an in vivo functional test for the selective expression, we found that Yoda1, a PIEZO1-specific agonist, induced a mechanical hyperalgesia that displayed a significantly prolonged time course compared with that induced by capsaicin, a TRPV1-specific agonist. Taken together, our results indicate that PIEZO1 should be considered a potential candidate in forming the long sought channel mediating mechano-nociception.
Collapse
Affiliation(s)
- Jigong Wang
- Department of Neuroscience, Cell Biology and Anatomy, The University of Texas Medical Branch, Galveston, TX, United States
| | - Jun-Ho La
- Department of Neuroscience, Cell Biology and Anatomy, The University of Texas Medical Branch, Galveston, TX, United States
| | - Owen P Hamill
- Department of Neuroscience, Cell Biology and Anatomy, The University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
47
|
Israel MR, Tanaka BS, Castro J, Thongyoo P, Robinson SD, Zhao P, Deuis JR, Craik DJ, Durek T, Brierley SM, Waxman SG, Dib-Hajj SD, Vetter I. Na V 1.6 regulates excitability of mechanosensitive sensory neurons. J Physiol 2019; 597:3751-3768. [PMID: 31087362 DOI: 10.1113/jp278148] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 05/08/2019] [Indexed: 12/20/2022] Open
Abstract
KEY POINTS Voltage-gated sodium channels are critical for peripheral sensory neuron transduction and have been implicated in a number of painful and painless disorders. The β-scorpion toxin, Cn2, is selective for NaV 1.6 in dorsal root ganglion neurons. NaV 1.6 plays an essential role in peripheral sensory neurons, specifically at the distal terminals of mechanosensing fibres innervating the skin and colon. NaV 1.6 activation also leads to enhanced response to mechanical stimulus in vivo. This works highlights the use of toxins in elucidating pain pathways moreover the importance of non-peripherally restricted NaV isoforms in pain generation. ABSTRACT Peripheral sensory neurons express multiple voltage-gated sodium channels (NaV ) critical for the initiation and propagation of action potentials and transmission of sensory input. Three pore-forming sodium channel isoforms are primarily expressed in the peripheral nervous system (PNS): NaV 1.7, NaV 1.8 and NaV 1.9. These sodium channels have been implicated in painful and painless channelopathies and there has been intense interest in them as potential therapeutic targets in human pain. Emerging evidence suggests NaV 1.6 channels are an important isoform in pain sensing. This study aimed to assess, using pharmacological approaches, the function of NaV 1.6 channels in peripheral sensory neurons. The potent and NaV 1.6 selective β-scorpion toxin Cn2 was used to assess the effect of NaV 1.6 channel activation in the PNS. The multidisciplinary approach included Ca2+ imaging, whole-cell patch-clamp recordings, skin-nerve and gut-nerve preparations and in vivo behavioural assessment of pain. Cn2 facilitates NaV 1.6 early channel opening, and increased persistent and resurgent currents in large-diameter dorsal root ganglion (DRG) neurons. This promotes enhanced excitatory drive and tonic action potential firing in these neurons. In addition, NaV 1.6 channel activation in the skin and gut leads to increased response to mechanical stimuli. Finally, intra-plantar injection of Cn2 causes mechanical but not thermal allodynia. This study confirms selectivity of Cn2 on NaV 1.6 channels in sensory neurons. Activation of NaV 1.6 channels, in terminals of the skin and viscera, leads to profound changes in neuronal responses to mechanical stimuli. In conclusion, sensory neurons expressing NaV 1.6 are important for the transduction of mechanical information in sensory afferents innervating the skin and viscera.
Collapse
Affiliation(s)
- Mathilde R Israel
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Brian S Tanaka
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT, 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Joel Castro
- Visceral Pain Research Group, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, 5042, Australia.,Hopwood Centre for Neurobiology and Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, 5000, Australia
| | - Panumart Thongyoo
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Samuel D Robinson
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Peng Zhao
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT, 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Jennifer R Deuis
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - David J Craik
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Thomas Durek
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, 5042, Australia.,Hopwood Centre for Neurobiology and Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, 5000, Australia
| | - Stephen G Waxman
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT, 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Sulayman D Dib-Hajj
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT, 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Irina Vetter
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia.,School of Pharmacy, Pharmacy Australia Centre of Excellence, 20 Cornwall St, Woolloongabba, Queensland, 4102, Australia
| |
Collapse
|
48
|
Restović I, Bočina I, Vukojević K, Kero D, Filipović N, Raonić J, Vučinić J, Vukmirović F, Vučković L, Saraga-Babić M. Time course and expression pattern of the neuronal markers in the developing human spinal cord. Int J Dev Neurosci 2019; 74:1-10. [PMID: 30753937 DOI: 10.1016/j.ijdevneu.2019.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/16/2019] [Accepted: 02/01/2019] [Indexed: 02/06/2023] Open
Abstract
The aim of this study was to examine the spatio-temporal appearance of different neuronal cell subtypes by analyzing expression patterns of several neuronal markers (calretinin, neurofilament 200 (NF200), vanilloid receptor 1(VR1) and calcitonin gene-related peptide (CGRP)) of the embryonic human spinal cord (SC). Developing human SCs from 11 human conceptuses beetwen 5-10 developmental weeks (DW) were examined by light and electron microscopy and immunofluorescence. Light and electron microscopy revealed different embryonic stages of recognizable structure of the SC. NF200, CGRP and VR1 positive cells were observed in SCs during 5th-6th DW. NF200 was predominantly expressed in the ventral part, indicating presence of motoneurons. As development advanced, NF200 was mainly expressed in the marginal zone. Expression of CGRP was intense during all of the investigated periods, predominantly during the 5th-6th DW pointing to neural sensory differentiation, as opposed to the last DW when reduced expression of CGRP in the marginal layer indicated the terminations of the sensory afferents. Expression of VR1 was highest in the intermediate zone, at the beginning and at the end of the investigated periods, pointing to VR1 spatial pattern in the visceral afferents in the grey matter, while the first signs of calretinin were found in the 9th-10th DW ventrally. Delineating the relationships between factors involved in processes of neuronal differentiation as well as spatial and temporal arrangement of SC interrelated neurons can provide a useful information about normal SC development as well as the insight in possible causes of anomalies and disorders during embryonic life.
Collapse
Affiliation(s)
- Ivana Restović
- Faculty of Humanities and Social Sciences, University of Split, Poljička cesta 35, 21 000 Split, Croatia.
| | - Ivana Bočina
- Faculty of Science, University of Split, Ruđera Boškovića 33, 21 000, Split, Croatia.
| | - Katarina Vukojević
- School of Medicine, University of Split, Department of Anatomy, Histology and Embryology, Laboratory for Early Human Development, Šoltanska 2, 21000, Split, Croatia.
| | - Darko Kero
- School of Medicine, University of Split, Department of Anatomy, Histology and Embryology, Laboratory for Early Human Development, Šoltanska 2, 21000, Split, Croatia.
| | - Natalija Filipović
- School of Medicine, University of Split, Department of Anatomy, Histology and Embryology, Laboratory for Early Human Development, Šoltanska 2, 21000, Split, Croatia; School of Medicine, University of Split, Department of Anatomy, Histology and Embryology, Laboratory Neurocardiology, Šoltanska 2, 21000, Split, Croatia.
| | - Janja Raonić
- Department of Histology and Embryology, Faculty of Medicine, University of Montenegro, Podgorica, Montenegro.
| | - Jelena Vučinić
- Department of Histology and Embryology, Faculty of Medicine, University of Montenegro, Podgorica, Montenegro.
| | - Filip Vukmirović
- Department of Pathology, Faculty of Medicine, University of Montenegro, Podgorica, Montenegro.
| | - Ljiljana Vučković
- Department of Histology and Embryology, Faculty of Medicine, University of Montenegro, Podgorica, Montenegro.
| | - Mirna Saraga-Babić
- School of Medicine, University of Split, Department of Anatomy, Histology and Embryology, Laboratory for Early Human Development, Šoltanska 2, 21000, Split, Croatia.
| |
Collapse
|
49
|
Paik SK, Yoo HI, Choi SK, Bae JY, Park SK, Bae YC. Distribution of excitatory and inhibitory axon terminals on the rat hypoglossal motoneurons. Brain Struct Funct 2019; 224:1767-1779. [PMID: 31006070 DOI: 10.1007/s00429-019-01874-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 04/11/2019] [Indexed: 12/19/2022]
Abstract
Detailed information about the excitatory and inhibitory synapses on the hypoglossal motoneurons may help understand the neural mechanism for control of the hypoglossal motoneuron excitability and hence the precise and coordinated movements of the tongue during chewing, swallowing and licking. For this, we investigated the distribution of GABA-, glycine (Gly)- and glutamate (Glut)-immunopositive (+) axon terminals on the genioglossal (GG) motoneurons by retrograde tracing, electron microscopic immunohistochemistry, and quantitative analysis. Small GG motoneurons (< 400 μm2 in cross-sectional area) had fewer primary dendrites, significantly higher nuclear/cytoplasmic ratio, and smaller membrane area covered by synaptic boutons than large GG motoneurons (> 400 μm2). The fraction of inhibitory boutons (GABA + only, Gly + only, and mixed GABA +/Gly + boutons) of all boutons was significantly higher for small GG motoneurons than for large ones, whereas the fraction of Glut + boutons was significantly higher for large GG motoneurons than for small ones. Almost all boutons (> 95%) on both small and large GG motoneurons were GABA + , Gly + or Glut + . The frequency of mixed GABA +/Gly + boutons was the highest among inhibitory boutons types for both small and large GG motoneurons. These findings may elucidate the anatomical substrate for precise regulation of the motoneuron firing required for the fine movements of the tongue, and also suggest that the excitability of small and large GG motoneurons may be regulated differently.
Collapse
Affiliation(s)
- Sang Kyoo Paik
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, 188-1, 2-Ga, Samdeok-Dong, Jung-Gu, Daegu, 700-412, South Korea
| | - Hong Il Yoo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, 77 Gyeryong-ro 771 beon-gil, Jung-Gu, Daejeon, 34824, South Korea
| | - Seung Ki Choi
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, 188-1, 2-Ga, Samdeok-Dong, Jung-Gu, Daegu, 700-412, South Korea
| | - Jin Young Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, 188-1, 2-Ga, Samdeok-Dong, Jung-Gu, Daegu, 700-412, South Korea
| | - Sook Kyung Park
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, 188-1, 2-Ga, Samdeok-Dong, Jung-Gu, Daegu, 700-412, South Korea
| | - Yong Chul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, 188-1, 2-Ga, Samdeok-Dong, Jung-Gu, Daegu, 700-412, South Korea.
| |
Collapse
|
50
|
Cortical Gradients and Laminar Projections in Mammals. Trends Neurosci 2018; 41:775-788. [DOI: 10.1016/j.tins.2018.06.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 06/10/2018] [Accepted: 06/11/2018] [Indexed: 12/30/2022]
|